1451
|
|
1452
|
Caturano A, Pafundi PC, Sasso FC, Dendramis G, Brugada P, Russo V. Brugada syndrome and COVID-19 vaccines. Europace 2021; 23:1871-1872. [PMID: 34383900 PMCID: PMC8385984 DOI: 10.1093/europace/euab211] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/16/2021] [Indexed: 01/25/2023] Open
Affiliation(s)
- Alfredo Caturano
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Piazza Luigi Miraglia 2, IT-80138 Naples, Italy
| | - Pia Clara Pafundi
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Piazza Luigi Miraglia 2, IT-80138 Naples, Italy
| | - Ferdinando Carlo Sasso
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Piazza Luigi Miraglia 2, IT-80138 Naples, Italy
| | - Gregory Dendramis
- Department of Cardiology, ARNAS Ospedale Civico e Benfratelli, Palermo, Italy
- Department of Cardiology, Heart Rhythm Managment Centre, UZ Brussel-VUB, Brussels, Belgium
| | - Pedro Brugada
- Department of Cardiology, Heart Rhythm Managment Centre, UZ Brussel-VUB, Brussels, Belgium
| | - Vincenzo Russo
- Division of Cardiology, Department of Medical Translational Sciences, Monaldi Hospital, University of Campania “Luigi Vanvitelli” Naples, Italy
| |
Collapse
|
1453
|
Hebbani AV, Pulakuntla S, Pannuru P, Aramgam S, Badri KR, Reddy VD. COVID-19: comprehensive review on mutations and current vaccines. Arch Microbiol 2021; 204:8. [PMID: 34873656 PMCID: PMC8647783 DOI: 10.1007/s00203-021-02606-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 11/09/2021] [Accepted: 11/22/2021] [Indexed: 12/15/2022]
Abstract
Viral outbreaks had been a threat for the human race for a long time. Several epidemics and pandemics have been reported in the past with serious consequences on human health and subsequent social and economic aspects. According to WHO, viral infections continue to be a major health concern globally. Novel coronavirus, SARS-CoV-2 (Severe acute respiratory syndrome coronavirus-2) causes the most recent infectious pandemic disease, COVID-19 (Coronavirus disease-19). As of now, there were 249 million infections of COVID-19 worldwide with a high mortality of more than 5 million deaths reported; and the number of new additional cases is drastically increasing. Development of therapies to treat the infected cases and prophylactic agents including vaccines that are effective towards different variants are crucial to curtail the COVID-19 pandemic. Owing to the fact that there is a high mortality and morbidity rate along with the risk of virus causing further epidemic outbursts, development of additional effective therapeutic and preventive strategies are highly warranted. Prevention, early detection and treatment will reduce the spread of COVID-19 pandemic. The present review highlights the novel mutations and therapeutic updates associated with coronaviruses along with the clinical manifestations-diagnosis, clinical management and, prophylactic and therapeutic strategies of COVID-19 infection.
Collapse
Affiliation(s)
| | - Swetha Pulakuntla
- Department of Biochemistry, REVA University, Bengaluru, 560064, India
| | - Padmavathi Pannuru
- DR Biosciences, Research and Development Institute, Bettahalasur, Bengaluru, 562157, India
| | - Sreelatha Aramgam
- Department of Biochemistry, REVA University, Bengaluru, 560064, India
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Kameswara Rao Badri
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, GA, 30310, USA.
- Clinical Analytical Chemistry Laboratory, Clinical Research Center, Morehouse School of Medicine, Atlanta, GA, 30310, USA.
| | | |
Collapse
|
1454
|
Weaver KN, Zhang X, Dai X, Watkins R, Adler J, Dubinsky MC, Kastl A, Bousvaros A, Strople JA, Cross RK, Higgins PDR, Ungaro RC, Bewtra M, Bellaguarda E, Farraye FA, Boccieri ME, Firestine A, Kappelman MD, Long MD. Impact of SARS-CoV-2 Vaccination on Inflammatory Bowel Disease Activity and Development of Vaccine-Related Adverse Events: Results From PREVENT-COVID. Inflamm Bowel Dis 2021; 28:1497-1505. [PMID: 34871388 PMCID: PMC8822409 DOI: 10.1093/ibd/izab302] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 vaccination is recommended for all individuals with inflammatory bowel disease (IBD), including those on immunosuppressive therapies; however, little is known about vaccine safety and efficacy in these patients or the impact of vaccination on IBD disease course. METHODS We evaluated coronavirus disease 2019 (COVID-19) vaccine-related adverse events (AEs) and the effect of vaccination on IBD disease course among participants in the PREVENT-COVID (Partnership to Report Effectiveness of Vaccination in populations Excluded from iNitial Trials of COVID) study, a prospective, observational cohort study. Localized and systemic reactions were assessed via questionnaire. Disease flare was defined by worsening IBD symptoms and change in IBD medications. Outcomes were stratified by vaccine type and IBD medication classes. RESULTS A total of 3316 individuals with IBD received at least 1 COVID-19 vaccine. Injection site tenderness (68%) and fatigue (46% dose 1, 68% dose 2) were the most commonly reported localized and systemic AEs after vaccination. Severe localized and systemic vaccine-related AEs were rare. The mRNA-1273 vaccine was associated with significantly greater severe AEs at dose 2 (localized 4% vs 2%, systemic 15% vs 10%; P < .001 for both). Prior COVID-19 infection, female sex, and vaccine type were associated with severe systemic reactions to dose 1, while age <50 years, female sex, vaccine type, and antitumor necrosis factor and vedolizumab use were associated with severe systemic reactions to dose 2. Overall rates (2%) of IBD flare were low following vaccination. CONCLUSIONS Our findings provide reassurance that the severe acute respiratory syndrome coronavirus 2 vaccine is safe and well tolerated among individuals with IBD, which may help to combat vaccine hesitancy and increase vaccine confidence.
Collapse
Affiliation(s)
- Kimberly N Weaver
- Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA,Address correspondence to: Kimberly N. Weaver, MD, Corresponding address: University of North Carolina at Chapel Hill, 130 Mason Farm Road, Campus Box #7080, Chapel Hill, NC 27599, USA ()
| | - Xian Zhang
- Division of Pediatric Gastroenterology, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Xiangfeng Dai
- Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Runa Watkins
- Division of Pediatric Gastroenterology and Nutrition, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jeremy Adler
- Susan B. Meister Child Health Evaluation and Research Center and Division of Pediatric Gastroenterology, University of Michigan, Ann Arbor, MI, USA
| | - Marla C Dubinsky
- Susan and Leonard Feinstein IBD Center, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Arthur Kastl
- Division of Gastroenterology, Children’s Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Athos Bousvaros
- Department of Pediatrics, Boston Children’s Hospital, Boston, MA, USA
| | - Jennifer A Strople
- Department of Pediatrics, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, USA
| | - Raymond K Cross
- Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Peter D R Higgins
- Division of Gastroenterology & Hepatology, University of Michigan, Ann Arbor, MI, USA
| | - Ryan C Ungaro
- Susan and Leonard Feinstein IBD Center, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Meenakshi Bewtra
- Division of Gastroenterology, Perelman School of Medicine, University of Pennsylvania, Philadelphia PA, USA,Department of Biostatistics and Epidemiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia PA, USA
| | - Emanuelle Bellaguarda
- Division of Gastroenterology and Hepatology, Northwestern University, Chicago, IL, USAand
| | - Francis A Farraye
- Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL, USA
| | - Margie E Boccieri
- Division of Pediatric Gastroenterology, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ann Firestine
- Division of Pediatric Gastroenterology, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Michael D Kappelman
- Division of Pediatric Gastroenterology, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA,Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Millie D Long
- Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA,Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
1455
|
Lu X, Hosono Y, Nagae M, Ishizuka S, Ishikawa E, Motooka D, Ozaki Y, Sax N, Maeda Y, Kato Y, Morita T, Shinnakasu R, Inoue T, Onodera T, Matsumura T, Shinkai M, Sato T, Nakamura S, Mori S, Kanda T, Nakayama EE, Shioda T, Kurosaki T, Takeda K, Kumanogoh A, Arase H, Nakagami H, Yamashita K, Takahashi Y, Yamasaki S. Identification of conserved SARS-CoV-2 spike epitopes that expand public cTfh clonotypes in mild COVID-19 patients. J Exp Med 2021; 218:212701. [PMID: 34647971 PMCID: PMC8641254 DOI: 10.1084/jem.20211327] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/21/2021] [Accepted: 09/28/2021] [Indexed: 11/04/2022] Open
Abstract
Adaptive immunity is a fundamental component in controlling COVID-19. In this process, follicular helper T (Tfh) cells are a subset of CD4+ T cells that mediate the production of protective antibodies; however, the SARS-CoV-2 epitopes activating Tfh cells are not well characterized. Here, we identified and crystallized TCRs of public circulating Tfh (cTfh) clonotypes that are expanded in patients who have recovered from mild symptoms. These public clonotypes recognized the SARS-CoV-2 spike (S) epitopes conserved across emerging variants. The epitope of the most prevalent cTfh clonotype, S864-882, was presented by multiple HLAs and activated T cells in most healthy donors, suggesting that this S region is a universal T cell epitope useful for booster antigen. SARS-CoV-2-specific public cTfh clonotypes also cross-reacted with specific commensal bacteria. In this study, we identified conserved SARS-CoV-2 S epitopes that activate public cTfh clonotypes associated with mild symptoms.
Collapse
Affiliation(s)
- Xiuyuan Lu
- Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Yuki Hosono
- Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, Japan.,Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan.,Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Masamichi Nagae
- Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, Japan.,Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Shigenari Ishizuka
- Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, Japan.,Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Eri Ishikawa
- Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, Japan.,Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Daisuke Motooka
- Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Yuki Ozaki
- Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | | | - Yuichi Maeda
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan.,Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yasuhiro Kato
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan.,Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Takayoshi Morita
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan.,Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Ryo Shinnakasu
- Laboratory of Lymphocyte Differentiation, Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Takeshi Inoue
- Laboratory of Lymphocyte Differentiation, Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Taishi Onodera
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan
| | - Takayuki Matsumura
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan
| | | | | | - Shota Nakamura
- Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Shunsuke Mori
- Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, Suita, Japan.,Laboratory of Immunochemistry, Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Teru Kanda
- Division of Microbiology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Emi E Nakayama
- Department of Viral Infections, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Tatsuo Shioda
- Department of Viral Infections, Research Institute for Microbial Diseases, Osaka University, Suita, Japan.,Center for Infectious Disease Education and Research, Osaka University, Suita, Japan
| | - Tomohiro Kurosaki
- Laboratory of Lymphocyte Differentiation, Immunology Frontier Research Center, Osaka University, Suita, Japan.,Center for Infectious Disease Education and Research, Osaka University, Suita, Japan.,Laboratory of Lymphocyte Differentiation, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Kiyoshi Takeda
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Suita, Japan.,Center for Infectious Disease Education and Research, Osaka University, Suita, Japan.,Department of Mucosal Immunology, Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Japan.,Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita, Japan.,Center for Infectious Disease Education and Research, Osaka University, Suita, Japan
| | - Hisashi Arase
- Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, Suita, Japan.,Laboratory of Immunochemistry, Immunology Frontier Research Center, Osaka University, Suita, Japan.,Center for Infectious Disease Education and Research, Osaka University, Suita, Japan
| | - Hironori Nakagami
- Department of Health Development and Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | | | - Yoshimasa Takahashi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan
| | - Sho Yamasaki
- Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, Japan.,Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan.,Center for Infectious Disease Education and Research, Osaka University, Suita, Japan.,Division of Molecular Immunology, Medical Mycology Research Center, Chiba University, Chiba, Japan.,Division of Molecular Design, Research Center for Systems Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| |
Collapse
|
1456
|
Reimann P, Ulmer H, Mutschlechner B, Benda M, Severgnini L, Volgger A, Lang T, Atzl M, Huynh M, Gasser K, Grabher C, Mink S, Fraunberger P, Petrausch U, Hartmann B, Winder T. Efficacy and safety of heterologous booster vaccination with Ad26.COV2.S after BNT162b2 mRNA COVID-19 vaccine in haemato-oncological patients with no antibody response. Br J Haematol 2021; 196:577-584. [PMID: 34872162 DOI: 10.1111/bjh.17982] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 11/18/2021] [Accepted: 11/19/2021] [Indexed: 01/11/2023]
Abstract
Patients with haemato-oncological malignancies are one of the high-risk groups for a severe course in case of COVID-19 infections. Furthermore, vaccination results in significantly lower response rates in haematological malignancies and lower antibody levels in patients with solid cancer. We investigated efficacy and safety of a heterologous booster vaccination with Ad26.COV2.S DNA vector vaccine in haemato-oncological patients without antibody response after double-dose BNT162b2 messenger (m-)RNA COVID-19 vaccine. A total of 32 haemato-oncological non-responders to double-dose BNT162b2 received a heterologous booster vaccination with Ad26.COV2.S. Blood samples were assessed directly before the vaccination (T0) and four weeks after (T1). Safety assessment was performed using a standardised questionnaire. The overall response rate was 31%, with a mean (SD) antibody titre of 693·79 (1 096·99) binding activity units (BAU)/ml. Patients with chronic lymphocytic leukaemia or lymphoma showed a significantly lower response rate (P = 0·048). Adverse events were reported in 29·6% of patients, of which 7·1% were graded as severe, including grade III and IV events following the Common Terminology Criteria of Adverse Events (CTCAE). The heterologous booster vaccination with Ad26.COV2.S led to a serological response in nine out of 29 patients without response after double-dose BNT162b2. Furthermore, the vaccination was safe in our cohort, leading to mainly mild local and systemic reactions. Overall, this vaccination regimen should be further evaluated to increase the response rate in the highly vulnerable population of haemato-oncological patients.
Collapse
Affiliation(s)
- Patrick Reimann
- Department of Internal Medicine II, Feldkirch Academic Teaching Hospital, Feldkirch, Austria.,Private University of the Principality of Liechtenstein, Triesen, Principality of Liechtenstein
| | - Hanno Ulmer
- Department of Medical Statistics, Informatics and Health Economics, Innsbruck Medical University, Innsbruck, Austria
| | - Beatrix Mutschlechner
- Private University of the Principality of Liechtenstein, Triesen, Principality of Liechtenstein.,Department of Internal Medicine I, Feldkirch Academic Teaching Hospital, Feldkirch, Austria
| | - Magdalena Benda
- Department of Internal Medicine II, Feldkirch Academic Teaching Hospital, Feldkirch, Austria.,Private University of the Principality of Liechtenstein, Triesen, Principality of Liechtenstein
| | - Luciano Severgnini
- Department of Internal Medicine II, Feldkirch Academic Teaching Hospital, Feldkirch, Austria
| | - Andreas Volgger
- Department of Internal Medicine II, Feldkirch Academic Teaching Hospital, Feldkirch, Austria
| | - Theresia Lang
- Department of Internal Medicine II, Feldkirch Academic Teaching Hospital, Feldkirch, Austria
| | - Michele Atzl
- Department of Internal Medicine II, Feldkirch Academic Teaching Hospital, Feldkirch, Austria
| | - Minh Huynh
- Department of Internal Medicine II, Feldkirch Academic Teaching Hospital, Feldkirch, Austria
| | - Klaus Gasser
- Department of Internal Medicine II, Feldkirch Academic Teaching Hospital, Feldkirch, Austria
| | | | - Sylvia Mink
- Private University of the Principality of Liechtenstein, Triesen, Principality of Liechtenstein.,Medical Central Laboratories, Feldkirch, Austria
| | | | - Ulf Petrausch
- University of Zurich, Zurich, Switzerland.,Onkozentrum Zürich, Swiss Tumour Immunology Institute, Zürich, Switzerland
| | - Bernd Hartmann
- Department of Internal Medicine II, Feldkirch Academic Teaching Hospital, Feldkirch, Austria
| | - Thomas Winder
- Department of Internal Medicine II, Feldkirch Academic Teaching Hospital, Feldkirch, Austria.,University of Zurich, Zurich, Switzerland
| |
Collapse
|
1457
|
Coburn SB, Humes E, Lang R, Stewart C, Hogan BC, Gebo KA, Napravnik S, Edwards JK, Browne LE, Park LS, Justice AC, Gordon K, Horberg MA, Certa JM, Watson E, Jefferson CR, Silverberg M, Skarbinski J, Leyden WA, Williams CF, Althoff KN. COVID-19 infections post-vaccination by HIV status in the United States. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.12.02.21267182. [PMID: 34909791 PMCID: PMC8669858 DOI: 10.1101/2021.12.02.21267182] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
IMPORTANCE Recommendations for additional doses of COVID vaccine are restricted to people with HIV who have advanced disease or unsuppressed HIV viral load. Understanding SARS-CoV-2 infection risk post-vaccination among PWH is essential for informing vaccination guidelines. OBJECTIVE Estimate the risk of breakthrough infections among fully vaccinated people with (PWH) and without (PWoH) HIV in the US. DESIGN SETTING AND PARTICIPANTS The Corona-Infectious-Virus Epidemiology Team (CIVET)-II cohort collaboration consists of 4 longitudinal cohorts from integrated health systems and academic health centers. Each cohort identified individuals ≥18 years old, in-care, and fully vaccinated for COVID-19 through 30 June 2021. PWH were matched to PWoH on date fully vaccinated, age group, race/ethnicity, and sex at birth. Incidence rates per 1,000 person-years and cumulative incidence of breakthrough infections with 95% confidence intervals ([,]) were estimated by HIV status. Cox proportional hazards models estimated adjusted hazard ratios (aHR) of breakthrough infections by HIV status adjusting for demographic factors, prior COVID-19 illness, vaccine type (BNT162b2, [Pfizer], mRNA-1273 [Moderna], Jansen Ad26.COV2.S [J&J]), calendar time, and cohort. Risk factors for breakthroughs among PWH, were also investigated. EXPOSURE HIV infection. OUTCOME COVID-19 breakthrough infections, defined as laboratory evidence of SARS-CoV-2 infection or COVID-19 diagnosis after an individual was fully vaccinated. RESULTS Among 109,599 individuals (31,840 PWH and 77,759 PWoH), the rate of breakthrough infections was higher in PWH versus PWoH: 44 [41, 48] vs. 31 [29, 33] per 1,000 person-years. Cumulative incidence at 210 days after date fully vaccinated was low, albeit higher in PWH versus PWoH overall (2.8% versus 2.1%, log-rank p<0.001, risk difference=0.7% [0.4%, 1.0%]) and within each vaccine type. Breakthrough infection risk was 41% higher in PWH versus PWoH (aHR=1.41 [1.28, 1.56]). Among PWH, younger age (18-24 versus 45-54), history of COVID-19 prior to fully vaccinated date, and J&J vaccination (versus Pfizer) were associated with increased risk of breakthroughs. There was no association of breakthrough with HIV viral load suppression or CD4 count among PWH. CONCLUSIONS AND RELEVANCE COVID-19 vaccination is effective against infection with SARS-CoV-2 strains circulating through 30 Sept 2021. PWH have an increased risk of breakthrough infections compared to PWoH. Recommendations for additional vaccine doses should be expanded to all PWH.
Collapse
Affiliation(s)
- Sally B Coburn
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Elizabeth Humes
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Raynell Lang
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Medicine, University of Calgary, Calgary, Canada
| | - Cameron Stewart
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Brenna C Hogan
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Kelly A Gebo
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Sonia Napravnik
- Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jessie K Edwards
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Lindsay E Browne
- Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Lesley S Park
- Stanford Center for Population Health Sciences, Palo Alto, CA, USA
| | - Amy C Justice
- Department of Health Policy and Management, Yale School of Public Health, New Haven, CT, USA
- Department of Medicine, Yale School of Medicine, New Haven, CT, USA
- VA Connecticut Healthcare System, West Haven, CT, USA
| | - Kirsha Gordon
- Department of Medicine, Yale School of Medicine, New Haven, CT, USA
- VA Connecticut Healthcare System, West Haven, CT, USA
| | - Michael A Horberg
- Kaiser Permanente Mid-Atlantic Permanente Research Institute, Rockville, MD, USA
| | - Julia M Certa
- Kaiser Permanente Mid-Atlantic Permanente Research Institute, Rockville, MD, USA
| | - Eric Watson
- Kaiser Permanente Mid-Atlantic Permanente Research Institute, Rockville, MD, USA
| | - Celeena R Jefferson
- Kaiser Permanente Mid-Atlantic Permanente Research Institute, Rockville, MD, USA
| | - Michael Silverberg
- Kaiser Permanente Northern California, Division of Research, Oakland CA, USA
| | - Jacek Skarbinski
- Kaiser Permanente Northern California, Division of Research, Oakland CA, USA
- Department of Infectious Diseases, Oakland Medical Center, Oakland CA, USA
| | - Wendy A Leyden
- Kaiser Permanente Northern California, Division of Research, Oakland CA, USA
| | - Carolyn F Williams
- Epidemiology Branch, Division of AIDS at National Institute of Allergy and Infectious Diseases (NIAID), National Institute of Health (NIH), Rockville, MD, USA
| | - Keri N Althoff
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
1458
|
Aksoyalp ZŞ, Nemutlu-Samur D. Sex-related susceptibility in coronavirus disease 2019 (COVID-19): Proposed mechanisms. Eur J Pharmacol 2021; 912:174548. [PMID: 34606834 PMCID: PMC8486578 DOI: 10.1016/j.ejphar.2021.174548] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/07/2021] [Accepted: 09/29/2021] [Indexed: 12/23/2022]
Abstract
The importance of sex differences is increasingly acknowledged in the incidence and treatment of disease. Accumulating clinical evidence demonstrates that sex differences are noticeable in COVID-19, and the prevalence, severity, and mortality rate of COVID-19 are higher among males than females. Sex-related genetic and hormonal factors and immunological responses may underlie the sex bias in COVID-19 patients. Angiotensin-converting enzyme 2 (ACE2) and transmembrane protease/serine subfamily member 2 (TMPRSS2) are essential proteins involved in the cell entry of SARS-CoV-2. Since ACE2 is encoded on the X-chromosome, a double copy of ACE2 in females may compensate for virus-mediated downregulation of ACE2, and thus ACE2-mediated cellular protection is greater in females. The X chromosome also contains the largest immune-related genes leading females to develop more robust immune responses than males. Toll-like receptor-7 (TLR-7), one of the key players in innate immunity, is linked to sex differences in autoimmunity and vaccine efficacy, and its expression is greater in females. Sex steroids also affect immune cell function. Estrogen contributes to higher CD4+ and CD8+ T cell activation levels, and females have more B cells than males. Sex differences not only affect the severity and progression of the disease, but also alter the efficacy of pharmacological treatment and adverse events related to the drugs/vaccines used against COVID-19. Administration of different drugs/vaccines in different doses or intervals may be useful to eliminate sex differences in efficacy and side/adverse effects. It should be noted that studies should include sex-specific analyses to develop further sex-specific treatments for COVID-19.
Collapse
Affiliation(s)
- Zinnet Şevval Aksoyalp
- Izmir Katip Celebi University, Faculty of Pharmacy, Department of Pharmacology, 35620, Izmir, Turkey.
| | - Dilara Nemutlu-Samur
- Alanya Alaaddin Keykubat University, Faculty of Medicine, Department of Pharmacology, 07450, Antalya, Turkey.
| |
Collapse
|
1459
|
Weinberger B. Vaccines and Vaccination against SARS-CoV-2: Considerations for the Older Population. Vaccines (Basel) 2021; 9:1435. [PMID: 34960181 PMCID: PMC8704374 DOI: 10.3390/vaccines9121435] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/23/2021] [Accepted: 12/02/2021] [Indexed: 12/23/2022] Open
Abstract
Age is among the most prominent risk factors for developing severe COVID-19 disease, and therefore older adults are a major target group for vaccination against SARS-CoV-2. This review focusses on age-associated aspects of COVID-19 vaccines and vaccination strategies, and summarizes data on immunogenicity, efficacy and effectiveness of the four COVID-19 vaccines, which are licensed in the US and/or Europe; namely, the two mRNA vaccines by BioNTech/Pfizer (BNT162b2) and Moderna (mRNA-1273), and the adenovector vaccines developed by AstraZeneca/University Oxford (ChAdOx1-nCoV-19, AZD1222) and Janssen/Johnson&Johnson (Ad26.COV2-S), respectively. After very high protection rates in the first months after vaccination even in the older population, effectiveness of the vaccines, particularly against asymptomatic infection and mild disease, declined at later time points and with the emergence of virus variants. Many high-income countries have recently started administration of additional doses to older adults and other high-risk groups, whereas other parts of the world are still struggling to acquire and distribute vaccines for primary vaccination. Other vaccines are available in other countries and clinical development for more vaccine candidates is ongoing, but a complete overview of COVID-19 vaccine development is beyond the scope of this article.
Collapse
Affiliation(s)
- Birgit Weinberger
- Institute for Biomedical Aging Research, Universität Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
1460
|
Singh A, Kumar S. Immune variants of SARS-CoV-2 could be a significant challenge for developing a pan genotype-specific vaccine. Hum Vaccin Immunother 2021; 17:5145-5147. [PMID: 34856855 DOI: 10.1080/21645515.2021.1997487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
The circulating variants of SARS-CoV-2 pose a threat to the public health response worldwide, especially in the case of developing countries, lacking vaccines and having compromised health-care facilities. This article highlights several recent studies conducted to determine the efficacy of COVID-19 vaccines against variants of concern. These studies comprise clinical trials and neutralization assay-based studies conducted on messenger RNA (mRNA), recombinant, viral vector-based, and inactivated vaccines.
Collapse
Affiliation(s)
- Abhay Singh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| | - Sachin Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| |
Collapse
|
1461
|
Pescarini JM, Teixeira CSS, Cruz EP, Ortelan N, Pinto PFPS, Ferreira AJF, Alves FJO, Pinto Junior EP, Falcão IR, Rocha ADS, Silva NBD, Ortiz RF, Saavedra RDC, Oliveira VDA, Ribeiro-Silva RDC, Ichihara MYT, Boaventura V, Barral Netto M, Kerr LRFS, Werneck GL, Barreto ML. Methods to evaluate COVID-19 vaccine effectiveness, with an emphasis on quasi-experimental approaches. CIENCIA & SAUDE COLETIVA 2021; 26:5599-5614. [PMID: 34852093 DOI: 10.1590/1413-812320212611.18622021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 09/29/2021] [Indexed: 11/22/2022] Open
Abstract
The evaluation of vaccine effectiveness is conducted with real-world data. They are essential to monitor the performance of vaccination programmes over time, and in the context of the emergence of new variants. Until now, the effectiveness of COVID-19 vaccines has been assessed based on classic methods, such as cohort and test-negative case-control studies, which may often not allow for adequate control of inherent biases in the assignment of vaccination campaigns. The aim of this review was to discuss the study designs available to evaluate vaccine effectiveness, highlighting quasi-experimental studies, which seek to mimic randomized trials, by introducing an exogenous component to allocate to treatment, in addition to the advantages, limitations, and applicability in the context of Brazilian data. The use of quasi-experimental approaches, such as interrupted time series, difference-in-differences, propensity scores, instrumental variables, and regression discontinuity design, are relevant due to the possibility of providing more accurate estimates of COVID-19 vaccine effectiveness. This is especially important in scenarios such as the Brazilian, which characterized by the use of various vaccines, with the respective numbers and intervals between doses, applied to different age groups, and introduced at different times during the pandemic.
Collapse
Affiliation(s)
- Julia Moreira Pescarini
- Centro de Integração de Dados e Conhecimentos para Saúde, Instituto Gonçalo Muniz, Fundação Oswaldo Cruz. Rua Waldemar Falcão 121, Candeal. 40296-710 Salvador BA Brasil.
| | - Camila Silveira Silva Teixeira
- Centro de Integração de Dados e Conhecimentos para Saúde, Instituto Gonçalo Muniz, Fundação Oswaldo Cruz. Rua Waldemar Falcão 121, Candeal. 40296-710 Salvador BA Brasil.
| | - Enny Paixão Cruz
- Centro de Integração de Dados e Conhecimentos para Saúde, Instituto Gonçalo Muniz, Fundação Oswaldo Cruz. Rua Waldemar Falcão 121, Candeal. 40296-710 Salvador BA Brasil.
| | - Naia Ortelan
- Centro de Integração de Dados e Conhecimentos para Saúde, Instituto Gonçalo Muniz, Fundação Oswaldo Cruz. Rua Waldemar Falcão 121, Candeal. 40296-710 Salvador BA Brasil.
| | - Priscila Fernanda Porto Scaff Pinto
- Centro de Integração de Dados e Conhecimentos para Saúde, Instituto Gonçalo Muniz, Fundação Oswaldo Cruz. Rua Waldemar Falcão 121, Candeal. 40296-710 Salvador BA Brasil.
| | - Andrêa Jacqueline Fortes Ferreira
- Centro de Integração de Dados e Conhecimentos para Saúde, Instituto Gonçalo Muniz, Fundação Oswaldo Cruz. Rua Waldemar Falcão 121, Candeal. 40296-710 Salvador BA Brasil.
| | - Flavia Jôse Oliveira Alves
- Centro de Integração de Dados e Conhecimentos para Saúde, Instituto Gonçalo Muniz, Fundação Oswaldo Cruz. Rua Waldemar Falcão 121, Candeal. 40296-710 Salvador BA Brasil.
| | - Elzo Pereira Pinto Junior
- Centro de Integração de Dados e Conhecimentos para Saúde, Instituto Gonçalo Muniz, Fundação Oswaldo Cruz. Rua Waldemar Falcão 121, Candeal. 40296-710 Salvador BA Brasil.
| | - Ila Rocha Falcão
- Centro de Integração de Dados e Conhecimentos para Saúde, Instituto Gonçalo Muniz, Fundação Oswaldo Cruz. Rua Waldemar Falcão 121, Candeal. 40296-710 Salvador BA Brasil.
| | - Aline Dos Santos Rocha
- Centro de Integração de Dados e Conhecimentos para Saúde, Instituto Gonçalo Muniz, Fundação Oswaldo Cruz. Rua Waldemar Falcão 121, Candeal. 40296-710 Salvador BA Brasil.
| | - Nivea Bispo da Silva
- Centro de Integração de Dados e Conhecimentos para Saúde, Instituto Gonçalo Muniz, Fundação Oswaldo Cruz. Rua Waldemar Falcão 121, Candeal. 40296-710 Salvador BA Brasil.
| | - Renzo Flores Ortiz
- Centro de Integração de Dados e Conhecimentos para Saúde, Instituto Gonçalo Muniz, Fundação Oswaldo Cruz. Rua Waldemar Falcão 121, Candeal. 40296-710 Salvador BA Brasil.
| | | | | | - Rita de Cássia Ribeiro-Silva
- Centro de Integração de Dados e Conhecimentos para Saúde, Instituto Gonçalo Muniz, Fundação Oswaldo Cruz. Rua Waldemar Falcão 121, Candeal. 40296-710 Salvador BA Brasil.
| | - Maria Yury Travassos Ichihara
- Centro de Integração de Dados e Conhecimentos para Saúde, Instituto Gonçalo Muniz, Fundação Oswaldo Cruz. Rua Waldemar Falcão 121, Candeal. 40296-710 Salvador BA Brasil.
| | - Viviane Boaventura
- Centro de Integração de Dados e Conhecimentos para Saúde, Instituto Gonçalo Muniz, Fundação Oswaldo Cruz. Rua Waldemar Falcão 121, Candeal. 40296-710 Salvador BA Brasil.
| | - Manoel Barral Netto
- Centro de Integração de Dados e Conhecimentos para Saúde, Instituto Gonçalo Muniz, Fundação Oswaldo Cruz. Rua Waldemar Falcão 121, Candeal. 40296-710 Salvador BA Brasil.
| | | | | | - Mauricio L Barreto
- Centro de Integração de Dados e Conhecimentos para Saúde, Instituto Gonçalo Muniz, Fundação Oswaldo Cruz. Rua Waldemar Falcão 121, Candeal. 40296-710 Salvador BA Brasil.
| |
Collapse
|
1462
|
Joshi G, Borah P, Thakur S, Sharma P, Mayank, Poduri R. Exploring the COVID-19 vaccine candidates against SARS-CoV-2 and its variants: where do we stand and where do we go? Hum Vaccin Immunother 2021; 17:4714-4740. [PMID: 34856868 PMCID: PMC8726002 DOI: 10.1080/21645515.2021.1995283] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/20/2021] [Accepted: 10/15/2021] [Indexed: 12/23/2022] Open
Abstract
As of September 2021, 117 COVID-19 vaccines are in clinical development, and 194 are in preclinical development as per the World Health Organization (WHO) published draft landscape. Among the 117 vaccines undergoing clinical trials, the major platforms include protein subunit; RNA; inactivated virus; viral vector, among others. So far, USFDA recognized to approve the Pfizer-BioNTech (Comirnaty) COVID-19 vaccine for its full use in individuals of 16 years of age and older. Though the approved vaccines are being manufactured at a tremendous pace, the wealthiest countries have about 28% of total vaccines despite possessing only 10.8% of the total world population, suggesting an inequity of vaccine distribution. The review comprehensively summarizes the history of vaccines, mainly focusing on vaccines for SARS-CoV-2. The review also connects relevant topics, including measurement of vaccines efficacy against SARS-CoV-2 and its variants, associated challenges, and limitations, as hurdles in global vaccination are also kept forth.
Collapse
Affiliation(s)
- Gaurav Joshi
- School of Pharmacy, Graphic Era Hill University, Dehradun, India
- Department of Pharmaceutical Sciences and Natural Products, School of Pharmaceutical Sciences, Central University of Punjab, Bathinda, India
| | - Pobitra Borah
- School of Pharmacy, Graphic Era Hill University, Dehradun, India
| | - Shweta Thakur
- Laboratory of Genomic Instability and Diseases, Department of Infectious Disease Biology, Institute of Life Sciences, Bhubaneshwar, India
| | - Praveen Sharma
- Department of Pharmaceutical Sciences and Natural Products, School of Pharmaceutical Sciences, Central University of Punjab, Bathinda, India
| | - Mayank
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Ramarao Poduri
- Department of Pharmaceutical Sciences and Natural Products, School of Pharmaceutical Sciences, Central University of Punjab, Bathinda, India
| |
Collapse
|
1463
|
Yu F, Lau LT, Fok M, Lau JYN, Zhang K. COVID-19 Delta variants-Current status and implications as of August 2021. PRECISION CLINICAL MEDICINE 2021; 4:287-292. [PMID: 35692860 PMCID: PMC8499995 DOI: 10.1093/pcmedi/pbab024] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/07/2021] [Accepted: 09/07/2021] [Indexed: 01/03/2023] Open
Abstract
The SARS-CoV-2 Delta variant has evolved as the dominant strain of the current pandemic. Studies have shown that this variant has increased infectivity/viral load, and reduced neutralization by the host antibodies from convalescent patients/vaccinees. Clinically, Delta variant infection has been observed/documented in convalescent patients/vaccinees, although with less incidence of severe diseases, but can serve as reservoir to spread the infection to the unvaccinated. The current understanding (as of 18 August 2021) on the virologic aspect (including the amino acid substitutions), clinical implications, and public health implications will be discussed in this mini review, and recommendations to health authorities will be provided.
Collapse
Affiliation(s)
- Flora Yu
- Department of Natural Sciences and Mathematics, University of Texas at Dallas, Richardson, Texas 75080-3021, USA
| | - Lok-Ting Lau
- Hong Kong Baptist University, Kowloon Tong, Hong Kong SAR 999077, China
| | - Manson Fok
- University Hospital and Faculty of Medicine, Macau University of Science and Technology, Macau SAR 999078, China
| | | | - Kang Zhang
- University Hospital and Faculty of Medicine, Macau University of Science and Technology, Macau SAR 999078, China
| |
Collapse
|
1464
|
Does infection with or vaccination against SARS-CoV-2 lead to lasting immunity? THE LANCET. RESPIRATORY MEDICINE 2021; 9:1450-1466. [PMID: 34688434 PMCID: PMC8530467 DOI: 10.1016/s2213-2600(21)00407-0] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/26/2021] [Accepted: 08/21/2021] [Indexed: 12/17/2022]
Abstract
Many nations are pursuing the rollout of SARS-CoV-2 vaccines as an exit strategy from unprecedented COVID-19-related restrictions. However, the success of this strategy relies critically on the duration of protective immunity resulting from both natural infection and vaccination. SARS-CoV-2 infection elicits an adaptive immune response against a large breadth of viral epitopes, although the duration of the response varies with age and disease severity. Current evidence from case studies and large observational studies suggests that, consistent with research on other common respiratory viruses, a protective immunological response lasts for approximately 5-12 months from primary infection, with reinfection being more likely given an insufficiently robust primary humoral response. Markers of humoral and cell-mediated immune memory can persist over many months, and might help to mitigate against severe disease upon reinfection. Emerging data, including evidence of breakthrough infections, suggest that vaccine effectiveness might be reduced significantly against emerging variants of concern, and hence secondary vaccines will need to be developed to maintain population-level protective immunity. Nonetheless, other interventions will also be required, with further outbreaks likely to occur due to antigenic drift, selective pressures for novel variants, and global population mobility.
Collapse
|
1465
|
Beatty AL, Peyser ND, Butcher XE, Cocohoba JM, Lin F, Olgin JE, Pletcher MJ, Marcus GM. Analysis of COVID-19 Vaccine Type and Adverse Effects Following Vaccination. JAMA Netw Open 2021; 4:e2140364. [PMID: 34935921 PMCID: PMC8696570 DOI: 10.1001/jamanetworkopen.2021.40364] [Citation(s) in RCA: 247] [Impact Index Per Article: 61.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 10/29/2021] [Indexed: 01/24/2023] Open
Abstract
Importance Little is known about the factors associated with COVID-19 vaccine adverse effects in a real-world population. Objective To evaluate factors potentially associated with participant-reported adverse effects after COVID-19 vaccination. Design, Setting, and Participants The COVID-19 Citizen Science Study, an online cohort study, includes adults aged 18 years and older with a smartphone or internet access. Participants complete daily, weekly, and monthly surveys on health and COVID-19-related events. This analysis includes participants who provided consent between March 26, 2020, and May 19, 2021, and received at least 1 COVID-19 vaccine dose. Exposures Participant-reported COVID-19 vaccination. Main Outcomes and Measures Participant-reported adverse effects and adverse effect severity. Candidate factors in multivariable logistic regression models included age, sex, race, ethnicity, subjective social status, prior COVID-19 infection, medical conditions, substance use, vaccine dose, and vaccine brand. Results The 19 586 participants had a median (IQR) age of 54 (38-66) years, and 13 420 (68.8%) were women. Allergic reaction or anaphylaxis was reported in 26 of 8680 participants (0.3%) after 1 dose of the BNT162b2 (Pfizer/BioNTech) or mRNA-1273 (Moderna) vaccine, 27 of 11 141 (0.2%) after 2 doses of the BNT162b2 or mRNA-1273 vaccine or 1 dose of the JNJ-78436735 (Johnson & Johnson) vaccine. The strongest factors associated with adverse effects were vaccine dose (2 doses of BNT162b2 or mRNA-1273 or 1 dose of JNJ-78436735 vs 1 dose of BNT162b2 or mRNA-1273; odds ratio [OR], 3.10; 95% CI, 2.89-3.34; P < .001), vaccine brand (mRNA-1273 vs BNT162b2, OR, 2.00; 95% CI, 1.86-2.15; P < .001; JNJ-78436735 vs BNT162b2: OR, 0.64; 95% CI, 0.52-0.79; P < .001), age (per 10 years: OR, 0.74; 95% CI, 0.72-0.76; P < .001), female sex (OR, 1.65; 95% CI, 1.53-1.78; P < .001), and having had COVID-19 before vaccination (OR, 2.17; 95% CI, 1.77-2.66; P < .001). Conclusions and Relevance In this real-world cohort, serious COVID-19 vaccine adverse effects were rare and comparisons across brands could be made, revealing that full vaccination dose, vaccine brand, younger age, female sex, and having had COVID-19 before vaccination were associated with greater odds of adverse effects. Large digital cohort studies may provide a mechanism for independent postmarket surveillance of drugs and devices.
Collapse
Affiliation(s)
- Alexis L. Beatty
- Department of Epidemiology and Biostatistics, University of California, San Francisco
- Division of Cardiology, Department of Medicine, University of California, San Francisco
| | - Noah D. Peyser
- Division of Cardiology, Department of Medicine, University of California, San Francisco
| | - Xochitl E. Butcher
- Division of Cardiology, Department of Medicine, University of California, San Francisco
| | - Jennifer M. Cocohoba
- Department of Clinical Pharmacy, University of California San Francisco School of Pharmacy
| | - Feng Lin
- Department of Epidemiology and Biostatistics, University of California, San Francisco
| | - Jeffrey E. Olgin
- Division of Cardiology, Department of Medicine, University of California, San Francisco
| | - Mark J. Pletcher
- Department of Epidemiology and Biostatistics, University of California, San Francisco
| | - Gregory M. Marcus
- Division of Cardiology, Department of Medicine, University of California, San Francisco
| |
Collapse
|
1466
|
Marfe G, Perna S, Shukla AK. Effectiveness of COVID-19 vaccines and their challenges (Review). Exp Ther Med 2021; 22:1407. [PMID: 34676000 PMCID: PMC8524740 DOI: 10.3892/etm.2021.10843] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 08/24/2021] [Indexed: 12/13/2022] Open
Abstract
At the end of 2019, a new disease recognized such as severe acute respiratory syndrome (SARS), was reported in Wuhan, China. This disease was caused by an unknown SARS coronavirus 2 (SARS-CoV-2); a virus is characterized by high infectivity among humans. In some cases, this disease can be asymptomatic, while in other cases can induce flu-like symptoms or acute respiratory distress syndrome, pneumonia and death. For this reason, the World Health Organization and Public Health Emergency of International Concern declared a pandemic status in January 2020. Currently, numerous countries have been involved in the development of effective vaccines to protect humans against SARS-CoV-2 infection. The present review will discuss the four vaccines, AZD1222 (AstraZeneca or Vaxzevria), Janssen (Ad26.COV2.S), Moderna/mRNA-1273 and BioNTech/Fosun/Pfizer BNT162b1, that are currently in use worldwide to understand their efficacy, but also evaluate the difficulties and challenges of vaccine development. Although several questions should be addressed regarding these vaccines, the current review will examine the viral elements used in the coronavirus-19 vaccine that can play a crucial role in inducing a strong immune response, as well as the different adverse effects that they can cause to individuals.
Collapse
Affiliation(s)
- Gabriella Marfe
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania ‘Luigi Vanvitelli’, 81100 Caserta, Italy
| | - Stefania Perna
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania ‘Luigi Vanvitelli’, 81100 Caserta, Italy
| | - Arvind Kumar Shukla
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan, Gyeongsangnam-do 50612, Republic of Korea
- Inventra Medclin Biomedical Healthcare and Research Center, Katemanivli, Kalyan, Thane, Maharashtra 421306, India
| |
Collapse
|
1467
|
Canas LS, Österdahl MF, Deng J, Hu C, Selvachandran S, Polidori L, May A, Molteni E, Murray B, Chen L, Kerfoot E, Klaser K, Antonelli M, Hammers A, Spector T, Ourselin S, Steves C, Sudre CH, Modat M, Duncan EL. Disentangling post-vaccination symptoms from early COVID-19. EClinicalMedicine 2021; 42:101212. [PMID: 34873584 PMCID: PMC8635464 DOI: 10.1016/j.eclinm.2021.101212] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/28/2021] [Accepted: 11/08/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Identifying and testing individuals likely to have SARS-CoV-2 is critical for infection control, including post-vaccination. Vaccination is a major public health strategy to reduce SARS-CoV-2 infection globally. Some individuals experience systemic symptoms post-vaccination, which overlap with COVID-19 symptoms. This study compared early post-vaccination symptoms in individuals who subsequently tested positive or negative for SARS-CoV-2, using data from the COVID Symptom Study (CSS) app. METHODS We conducted a prospective observational study in 1,072,313 UK CSS participants who were asymptomatic when vaccinated with Pfizer-BioNTech mRNA vaccine (BNT162b2) or Oxford-AstraZeneca adenovirus-vectored vaccine (ChAdOx1 nCoV-19) between 8 December 2020 and 17 May 2021, who subsequently reported symptoms within seven days (N=362,770) (other than local symptoms at injection site) and were tested for SARS-CoV-2 (N=14,842), aiming to differentiate vaccination side-effects per se from superimposed SARS-CoV-2 infection. The post-vaccination symptoms and SARS-CoV-2 test results were contemporaneously logged by participants. Demographic and clinical information (including comorbidities) were recorded. Symptom profiles in individuals testing positive were compared with a 1:1 matched population testing negative, including using machine learning and multiple models considering UK testing criteria. FINDINGS Differentiating post-vaccination side-effects alone from early COVID-19 was challenging, with a sensitivity in identification of individuals testing positive of 0.6 at best. Most of these individuals did not have fever, persistent cough, or anosmia/dysosmia, requisite symptoms for accessing UK testing; and many only had systemic symptoms commonly seen post-vaccination in individuals negative for SARS-CoV-2 (headache, myalgia, and fatigue). INTERPRETATION Post-vaccination symptoms per se cannot be differentiated from COVID-19 with clinical robustness, either using symptom profiles or machine-derived models. Individuals presenting with systemic symptoms post-vaccination should be tested for SARS-CoV-2 or quarantining, to prevent community spread. FUNDING UK Government Department of Health and Social Care, Wellcome Trust, UK Engineering and Physical Sciences Research Council, UK National Institute for Health Research, UK Medical Research Council and British Heart Foundation, Chronic Disease Research Foundation, Zoe Limited.
Collapse
Key Words
- AUC, Area under the curve
- BMI, Body mass index
- CI, Confidence interval
- COVID-19 detection
- COVID-19, Coronavirus disease 2019
- CSS, COVID Symptoms Study
- DI, Data invalid
- Early detection
- IQR, inter quartile range
- KCL, King's College London
- LFAT, Lateral flow antigen test
- LR, Logistic Regression
- Mobile technology
- NHS UK, National Health Service of the United Kingdom
- O-AZ, Oxford-AstraZeneca adenovirus-vectored vaccine
- PB, Pfizer-BoiNTech mRNA vaccine
- RF, Random forest
- ROC, Receiver operating curve
- SARS-CoV-2, Severe acute respiratory syndrome-related coronavirus-2
- Self-reported symptoms
- Side-effects
- UK, United Kingdom of Great Britain and Nothern Ireland
- Vaccination
- bMEM, Bayesian mixed-effect model
- rtPCR, Reverse transcription polymerase chain reaction
- severe acute respiratory syndrome‐related coronavirus 2 (SARS-CoV-2)
Collapse
Affiliation(s)
- Liane S. Canas
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Marc F. Österdahl
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, UK
| | - Jie Deng
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | | | | | | | | | - Erika Molteni
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Benjamin Murray
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Liyuan Chen
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Eric Kerfoot
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Kerstin Klaser
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Michela Antonelli
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Alexander Hammers
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
- King's College London & Guy's and St Thomas’ PET Centre, London, UK
| | - Tim Spector
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, UK
| | - Sebastien Ourselin
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Claire Steves
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, UK
| | - Carole H. Sudre
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
- Medical Research Council Unit for Lifelong Health and Ageing, Department of Population Science and Experimental Medicine. UK
- Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
| | - Marc Modat
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Emma L. Duncan
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, UK
| |
Collapse
|
1468
|
Ludwig H, Sonneveld P, Facon T, San-Miguel J, Avet-Loiseau H, Mohty M, Mateos MV, Moreau P, Cavo M, Pawlyn C, Zweegman S, Engelhardt M, Driessen C, Cook G, Dimopoulos MA, Gay F, Einsele H, Delforge M, Caers J, Weisel K, Jackson G, Garderet L, van de Donk N, Leleu X, Goldschmidt H, Beksac M, Nijhof I, Schreder M, Abildgaard N, Hajek R, Zojer N, Kastritis E, Broijl A, Schjesvold F, Boccadoro M, Terpos E. COVID-19 vaccination in patients with multiple myeloma: a consensus of the European Myeloma Network. Lancet Haematol 2021; 8:e934-e946. [PMID: 34756169 PMCID: PMC8553271 DOI: 10.1016/s2352-3026(21)00278-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/19/2021] [Accepted: 08/26/2021] [Indexed: 12/14/2022]
Abstract
Patients with multiple myeloma frequently present with substantial immune impairment and an increased risk for infections and infection-related mortality. The risk for infection with SARS-CoV-2 virus and resulting mortality is also increased, emphasising the importance of protecting patients by vaccination. Available data in patients with multiple myeloma suggest a suboptimal anti-SARS-CoV-2 immune response, meaning a proportion of patients are unprotected. Factors associated with poor response are uncontrolled disease, immunosuppression, concomitant therapy, more lines of therapy, and CD38 antibody-directed and B-cell maturation antigen-directed therapy. These facts suggest that monitoring the immune response to vaccination in patients with multiple myeloma might provide guidance for clinical management, such as administration of additional doses of the same or another vaccine, or even temporary treatment discontinuation, if possible. In those who do not exhibit a good response, prophylactic treatment with neutralising monoclonal antibody cocktails might be considered. In patients deficient of a SARS-CoV-2 immune response, adherence to measures for infection risk reduction is particularly recommended. This consensus was generated by members of the European Multiple Myeloma Network and some external experts. The panel members convened in virtual meetings and conducted an extensive literature research and evaluated recently published data and work presented at meetings, as well as findings from their own studies. The outcome of the discussions on establishing consensus recommendations for COVID-19 vaccination in patients with multiple myeloma was condensed into this Review.
Collapse
Affiliation(s)
- Heinz Ludwig
- Wilhelminen Cancer Research Institute, First Department of Medicine, Center for Oncology, Hematology, and Palliative Care, Clinic Ottakring, Vienna, Austria.
| | - Pieter Sonneveld
- Erasmus Medical Center Cancer Institute-Erasmus University Rotterdam, Rotterdam, Netherlands
| | - Thierry Facon
- University of Lille, CHU Lille, Service des Maladies du Sang, Lille, France
| | - Jesus San-Miguel
- Clínica Universidad de Navarra, CIMA, CIBERONC, IDISNA, Pamplona, Spain
| | | | - Mohamad Mohty
- Service d'Hematologie Clinique et Therapie Cellulaire, Hopital Saint-Antoine, Assistance Publique-Hopitaux de Paris (AP-HP), Sorbonne University, INSERM Unite Mixte de Recherche (UMR) 938, Paris, France
| | - Maria-Victoria Mateos
- Hospital Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca, Instituto de Biología Molecular y Celular del Cáncer (Universidad de Salamanca-Consejo Superior de Investigaciones Científicas), CIBERONC, Salamanca, Spain
| | - Philippe Moreau
- Department of Hematology, University hospital Hotel-Dieu, Nantes, France
| | - Michele Cavo
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Bologna, Italy
| | - Charlotte Pawlyn
- Institute of Cancer Research, London, UK; Royal Marsden Hospital, London, UK
| | - Sonja Zweegman
- Department of Hematology, Amsterdam UMC, VU University, Amsterdam, Netherlands
| | - Monika Engelhardt
- Department of Medicine I and Department of Hematology, Oncology, and Stem-Cell Transplantation, Clinical Cancer Research Group, University Hospital of Freiburg, Freiburg, Germany
| | - Christoph Driessen
- Department of Oncology and Hematology, Kantonsspital St Gallen, St Gallen, Switzerland
| | - Gordon Cook
- Leeds Institute of Clinical Trial Research, University of Leeds, Leeds, UK
| | - Melitios A Dimopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Francesca Gay
- Myeloma Unit, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza, Torino, Italy
| | - Hermann Einsele
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | | | - Jo Caers
- Department of Hematology, CHU de Liège, Liège, Belgium
| | - Katja Weisel
- Universitätsklinikum Hamburg-Eppendorf II, Medizinische Klinik und Poliklinik, Hamburg, Germany
| | - Graham Jackson
- Northern Centre for Cancer Care, Freeman Hospital, Newcastle Upon Tyne Hospitals trust, Newcastle Upon Tyne, UK
| | - Laurent Garderet
- Sorbonne Université-INSERM, UMR-S 938, Centre de Recherche Saint-Antoine-Team Hematopoietic and leukemic development, Assistance Publique-Hôpitaux de Paris, Hôpital Pitié Salpetrière, Département d'Hématologie et de Thérapie Cellulaire, Paris, France
| | - Niels van de Donk
- Department of Hematology, Amsterdam UMC, VU University, Amsterdam, Netherlands
| | - Xavier Leleu
- CHU Poitiers, Poitiers, France; Inserm, Poitiers, France
| | - Hartmut Goldschmidt
- University Hospital Heidelberg, Internal Medicine V and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Meral Beksac
- Department of Hematology, Ankara University, Ankara, Turkey
| | - Inger Nijhof
- Department of Hematology, Amsterdam UMC, VU University, Amsterdam, Netherlands
| | - Martin Schreder
- First Department of Medicine, Center for Oncology, Hematology, and Palliative Care, Clinic Ottakring, Vienna, Austria
| | - Niels Abildgaard
- Hematology Research Unit, Department of Hematology, Odense University Hospital, and Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Roman Hajek
- Department of Hematooncology, University Hospital Ostrava and Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Niklas Zojer
- First Department of Medicine, Center for Oncology, Hematology, and Palliative Care, Clinic Ottakring, Vienna, Austria
| | - Efstathios Kastritis
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Annemiek Broijl
- Erasmus Medical Center Cancer Institute-Erasmus University Rotterdam, Rotterdam, Netherlands
| | | | - Mario Boccadoro
- KG Jebsen Center for B Cell Malignancies, University of Oslo, Oslo, Norway; European Myeloma Network (EMN) Italy, Torino, Italy
| | - Evangelos Terpos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
1469
|
Gresele P, Momi S, Marcucci R, Ramundo F, De Stefano V, Tripodi A. Interactions of adenoviruses with platelets and coagulation and the vaccine-induced immune thrombotic thrombocytopenia syndrome. Haematologica 2021; 106:3034-3045. [PMID: 34407607 PMCID: PMC8634187 DOI: 10.3324/haematol.2021.279289] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 08/04/2021] [Indexed: 11/10/2022] Open
Abstract
The COVID-19 pandemic has had a heavy impact on global health and economy and vaccination remains the primary way of controlling the infection. During the ongoing vaccination campaign some unexpected thrombotic events have emerged in subjects who had recently received the AstraZeneca (Vaxzevria) vaccine or the Johnson and Johnson (Janssen) vaccine, two adenovirus vector-based vaccines. Epidemiological studies confirm that the observed/expected ratio of these unusual thromboses is abnormally increased, especially in women in fertile age. The characteristics of this complication, with venous thromboses at unusual sites, most frequently in the cerebral vein sinuses but also in splanchnic vessels, often with multiple associated thromboses, thrombocytopenia, and sometimes disseminated intravascular coagulation, are unique and the time course and tumultuous evolution are suggestive of an acute immunological reaction. Indeed, plateletactivating anti-PF4 antibodies have been detected in a large proportion of the affected patients. Several data suggest that adenoviruses may interact with platelets, the endothelium and the blood coagulation system. Here we review interactions between adenoviral vectors and the hemostatic system that are of possible relevance in vaccine-associated thrombotic thrombocytopenia syndrome. We systematically analyze the clinical data on the reported thrombotic complications of adenovirus-based therapeutics and discuss all the current hypotheses on the mechanisms triggering this novel syndrome. Although, considering current evidence, the benefit of vaccination clearly outweighs the potential risks, it is of paramount importance to fully unravel the mechanisms leading to vaccineassociated thrombotic thrombocytopenia syndrome and to identify prognostic factors through further research.
Collapse
Affiliation(s)
- Paolo Gresele
- Department of Medicine and Surgery, Section of Internal and Cardiovascular Medicine, University of Perugia, Perugia.
| | - Stefania Momi
- Department of Medicine and Surgery, Section of Internal and Cardiovascular Medicine, University of Perugia, Perugia
| | - Rossella Marcucci
- Department of Experimental and Clinical Medicine, University of Florence; Atherothrombotic Center, AOU Careggi, Florence
| | - Francesco Ramundo
- Section of Hematology, Department of Radiological and Hematological Sciences, Catholic University, Fondazione Policlinico A. Gemelli - IRCCS - Rome
| | - Valerio De Stefano
- Section of Hematology, Department of Radiological and Hematological Sciences, Catholic University, Fondazione Policlinico A. Gemelli - IRCCS - Rome
| | - Armando Tripodi
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Angelo Bianchi Bonomi Hemophilia and Thromboses Center, Milan
| |
Collapse
|
1470
|
Tao K, Tzou PL, Nouhin J, Gupta RK, de Oliveira T, Kosakovsky Pond SL, Fera D, Shafer RW. The biological and clinical significance of emerging SARS-CoV-2 variants. Nat Rev Genet 2021; 22:757-773. [PMID: 34535792 PMCID: PMC8447121 DOI: 10.1038/s41576-021-00408-x] [Citation(s) in RCA: 700] [Impact Index Per Article: 175.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2021] [Indexed: 12/13/2022]
Abstract
The past several months have witnessed the emergence of SARS-CoV-2 variants with novel spike protein mutations that are influencing the epidemiological and clinical aspects of the COVID-19 pandemic. These variants can increase rates of virus transmission and/or increase the risk of reinfection and reduce the protection afforded by neutralizing monoclonal antibodies and vaccination. These variants can therefore enable SARS-CoV-2 to continue its spread in the face of rising population immunity while maintaining or increasing its replication fitness. The identification of four rapidly expanding virus lineages since December 2020, designated variants of concern, has ushered in a new stage of the pandemic. The four variants of concern, the Alpha variant (originally identified in the UK), the Beta variant (originally identified in South Africa), the Gamma variant (originally identified in Brazil) and the Delta variant (originally identified in India), share several mutations with one another as well as with an increasing number of other recently identified SARS-CoV-2 variants. Collectively, these SARS-CoV-2 variants complicate the COVID-19 research agenda and necessitate additional avenues of laboratory, epidemiological and clinical research.
Collapse
Affiliation(s)
- Kaiming Tao
- Division of Infectious Diseases, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Philip L Tzou
- Division of Infectious Diseases, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Janin Nouhin
- Division of Infectious Diseases, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Ravindra K Gupta
- Cambridge Institute for Therapeutic Immunology and Infectious Diseases, University of Cambridge, Cambridge, UK
| | - Tulio de Oliveira
- KwaZulu-Natal Research Innovation and Sequencing Platform (KRISP), University of KwaZulu-Natal, Durban, South Africa
| | | | - Daniela Fera
- Department of Chemistry and Biochemistry, Swarthmore College, Swarthmore, PA, USA
| | - Robert W Shafer
- Division of Infectious Diseases, Department of Medicine, Stanford University, Stanford, CA, USA.
- Department of Pathology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
1471
|
Madewell ZJ, Dean NE, Berlin JA, Coplan PM, Davis KJ, Struchiner CJ, Halloran ME. Challenges of evaluating and modelling vaccination in emerging infectious diseases. Epidemics 2021; 37:100506. [PMID: 34628108 PMCID: PMC8491997 DOI: 10.1016/j.epidem.2021.100506] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/25/2021] [Accepted: 10/04/2021] [Indexed: 12/17/2022] Open
Abstract
Outbreaks of emerging pathogens pose unique methodological and practical challenges for the design, implementation, and evaluation of vaccine efficacy trials. Lessons learned from COVID-19 highlight the need for innovative and flexible study design and application to quickly identify promising candidate vaccines. Trial design strategies should be tailored to the dynamics of the specific pathogen, location of the outbreak, and vaccine prototypes, within the regional socioeconomic constraints. Mathematical and statistical models can assist investigators in designing infectious disease clinical trials. We introduce key challenges for planning, evaluating, and modelling vaccine efficacy trials for emerging pathogens.
Collapse
Affiliation(s)
- Zachary J Madewell
- Department of Biostatistics, University of Florida, Gainesville, FL, USA.
| | - Natalie E Dean
- Department of Biostatistics, University of Florida, Gainesville, FL, USA
| | - Jesse A Berlin
- Global Epidemiology, Johnson & Johnson, Titusville, NJ, USA
| | - Paul M Coplan
- Medical Device Epidemiology and Real World Data Sciences, Johnson & Johnson, New Brunswick, NJ, USA; Department of Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, PA, USA
| | | | | | - M Elizabeth Halloran
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Department of Biostatistics, University of Washington, Seattle, WA, USA
| |
Collapse
|
1472
|
Shahgolzari M, Yavari A, Arjeini Y, Miri SM, Darabi A, Mozaffari Nejad AS, Keshavarz M. Immunopathology and Immunopathogenesis of COVID-19, what we know and what we should learn. GENE REPORTS 2021; 25:101417. [PMID: 34778602 PMCID: PMC8570409 DOI: 10.1016/j.genrep.2021.101417] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 10/28/2021] [Accepted: 11/01/2021] [Indexed: 02/08/2023]
Abstract
Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) directly interacts with host's epithelial and immune cells, leading to inflammatory response induction, which is considered the hallmark of infection. The host immune system is programmed to facilitate the clearance of viral infection by establishing a modulated response. However, SARS-CoV-2 takes the initiative and its various structural and non-structural proteins directly or indirectly stimulate the uncontrolled activation of injurious inflammatory pathways through interaction with innate immune system mediators. Upregulation of cell-signaling pathways such as mitogen-activate protein kinase (MAPK) in response to recognition of SARS-CoV-2 antigens by innate immune system receptors mediates unbridled production of proinflammatory cytokines and cells causing cytokine storm, tissue damage, increased pulmonary edema, acute respiratory distress syndrome (ARDS), and mortality. Moreover, this acute inflammatory state hinders the immunomodulatory effect of T helper cells and timely response of CD4+ and CD8+ T cells against infection. Furthermore, inflammation-induced overproduction of Th17 cells can downregulate the antiviral response of Th1 and Th2 cells. In fact, the improperly severe response of the innate immune system is the key to conversion from a non-severe to severe disease state and needs to be investigated more deeply. The virus can also modulate the protective immune responses by developing immune evasion mechanisms, and thereby provide a more stable niche. Overall, combination of detrimental immunostimulatory and immunomodulatory properties of both the SARS-CoV-2 and immune cells does complicate the immune interplay. Thorough understanding of immunopathogenic basis of immune responses against SARS-CoV-2 has led to developing several advanced vaccines and immune-based therapeutics and should be expanded more rapidly. In this review, we tried to delineate the immunopathogenesis of SARS-CoV-2 in humans and to provide insight into more effective therapeutic and prophylactic strategies.
Collapse
Affiliation(s)
- Mehdi Shahgolzari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Afagh Yavari
- Department of Biology, Payame Noor University, Tehran, Iran
| | - Yaser Arjeini
- Department of Research and Development, Production and Research Complex, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Mohammad Miri
- Freelance Researcher of Biomedical Sciences, No 32, Vaezi Street, Tehran, Iran
| | - Amirhossein Darabi
- The Persian Gulf Tropical Medicine Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Amir Sasan Mozaffari Nejad
- Department of Microbiology, Nutrition Health Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohsen Keshavarz
- The Persian Gulf Tropical Medicine Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| |
Collapse
|
1473
|
Nordström P, Ballin M, Nordström A. Association Between Risk of COVID-19 Infection in Nonimmune Individuals and COVID-19 Immunity in Their Family Members. JAMA Intern Med 2021; 181:1589-1595. [PMID: 34633407 PMCID: PMC8506298 DOI: 10.1001/jamainternmed.2021.5814] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
IMPORTANCE The association between COVID-19 immunity within families and the risk of infection in nonimmune family members is unknown. OBJECTIVE To investigate the association between risk of COVID-19 in nonimmune individuals and the number of their family members with known immunity acquired from a previous COVID-19 infection or full vaccination (2 vaccine doses). DESIGN, SETTING, AND PARTICIPANTS In this cohort study of data from nationwide registries in Sweden, all individuals who acquired immunity from either previous COVID-19 infection or full vaccination until May 26, 2021, were considered for inclusion. Each person with immunity was matched 1:1 to an individual without immunity from an identified cohort of individuals with families comprising 2 to 5 members. EXPOSURES Number of immune family members in each family on April 14, 2021 (index date), who acquired immunity from a previous COVID-19 infection or full vaccination (2 doses of the mRNA-1273, BNT162b2 mRNA, or ChAdOx1 nCoV-19 vaccine). MAIN OUTCOMES AND MEASURES Incident COVID-19 infection in nonimmune family members from April 15 to May 26, 2021. RESULTS A total of 1 789 728 individuals from 814 806 families were included in the analysis. Each family comprised 2 to 5 family members, with a mean (SD) age at baseline of 51.3 (19.5) years. During a mean (range) follow-up time of 26.3 (1-40) days, 88 797 of 1 549 989 (5.7%) nonimmune family members (mean [SD] age, 51.6 [17.7] years; 790 276 men [51.0%]) were diagnosed with COVID-19. There was an inverse dose-response association between the number of immune members in each family and the risk of incident COVID-19 infection in nonimmune family members. Nonimmune families with 1 immune family member had a 45% to 61% lower risk of contracting COVID-19 (hazard ratio [HR], 0.39-0.55; 95% CI, 0.37-0.61, P < .001). The risk reduction increased to 75% to 86% in families with 2 immune family members (HR, 0.14-0.25; 95% CI, 0.11-0.27; P < .001), 91% to 94% with 3 immune family members (HR, 0.06-0.09; 95% CI, 0.04-0.10; P < .001), and 97% with 4 immune family members (HR, 0.03; 95% CI, 0.02-0.05; P < .001). The results were similar for the outcome of COVID-19 infection that was severe enough to warrant a hospital stay. CONCLUSIONS AND RELEVANCE In this cohort study, family members without immunity had a 45% to 97% lower risk of contracting COVID-19 as the number of immune family members increased. Vaccination is a key strategy for decreasing the transmission of the virus within families.
Collapse
Affiliation(s)
- Peter Nordström
- Unit of Geriatric Medicine, Department of Community Medicine and Rehabilitation, Umeå University, Umeå, Sweden
| | - Marcel Ballin
- Unit of Geriatric Medicine, Department of Community Medicine and Rehabilitation, Umeå University, Umeå, Sweden.,Section of Sustainable Health, Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Anna Nordström
- Section of Sustainable Health, Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden.,School of Sport Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
1474
|
Aparicio B, Casares N, Egea J, Ruiz M, Llopiz D, Maestro S, Olagüe C, González-Aseguinolaza G, Smerdou C, López-Díaz de Cerio A, Inogés S, Prósper F, Yuste JR, Carmona-Torre F, Reina G, Lasarte JJ, Sarobe P. Preclinical evaluation of a synthetic peptide vaccine against SARS-CoV-2 inducing multiepitopic and cross-reactive humoral neutralizing and cellular CD4 and CD8 responses. Emerg Microbes Infect 2021; 10:1931-1946. [PMID: 34538222 PMCID: PMC8480813 DOI: 10.1080/22221751.2021.1978823] [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] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/24/2021] [Accepted: 09/06/2021] [Indexed: 11/01/2022]
Abstract
Identification of relevant epitopes is crucial for the development of subunit peptide vaccines inducing neutralizing and cellular immunity against SARS-CoV-2. Our aim was the characterization of epitopes in the receptor-binding domain (RBD) of SARS-CoV-2 spike (S) protein to generate a peptide vaccine. Epitope mapping using a panel of 10 amino acid overlapped 15-mer peptides covering region 401-515 from RBD did not identify linear epitopes when tested with sera from infected individuals or from RBD-immunized mice. However, immunization of mice with these 15-mer peptides identified four peptides located at region 446-480 that induced antibodies recognizing the peptides and RBD/S1 proteins. Immunization with peptide 446-480 from S protein formulated with Freund's adjuvant or with CpG oligodeoxinucleotide/Alum induced polyepitopic antibody responses in BALB/c and C56BL/6J mice, recognizing RBD (titres of 3 × 104-3 × 105, depending on the adjuvant) and displaying neutralizing capacity (80-95% inhibition capacity; p < 0.05) against SARS-CoV-2. Murine CD4 and CD8T-cell epitopes were identified in region 446-480 and vaccination experiments using HLA transgenic mice suggested the presence of multiple human T-cell epitopes. Antibodies induced by peptide 446-480 showed broad recognition of S proteins and S-derived peptides belonging to SARS-CoV-2 variants of concern. Importantly, vaccination with peptide 446-480 or with a cyclic version of peptide 446-488 containing a disulphide bridge between cysteines 480 and 488, protected humanized K18-hACE2 mice from a lethal dose of SARS-CoV-2 (62.5 and 75% of protection; p < 0.01 and p < 0.001, respectively). This region could be the basis for a peptide vaccine or other vaccine platforms against Covid-19.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Animals
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
- COVID-19/immunology
- COVID-19/prevention & control
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/standards
- Cross Reactions/immunology
- Epitope Mapping
- Epitopes, B-Lymphocyte
- Epitopes, T-Lymphocyte/immunology
- Humans
- Immunity, Cellular
- Immunity, Humoral
- Immunization
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Transgenic
- SARS-CoV-2/genetics
- SARS-CoV-2/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/immunology
- Vaccines, Subunit/immunology
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- Belén Aparicio
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Pamplona, Spain
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Noelia Casares
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Josune Egea
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Pamplona, Spain
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Marta Ruiz
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Pamplona, Spain
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Diana Llopiz
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Pamplona, Spain
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Sheila Maestro
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Cristina Olagüe
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Gloria González-Aseguinolaza
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Cristian Smerdou
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | | | | | - Felipe Prósper
- Clínica Universidad de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, Spain
| | | | | | | | - Juan J. Lasarte
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Pablo Sarobe
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Pamplona, Spain
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| |
Collapse
|
1475
|
Desai A, Desai P, Mehta J, Sachora W, Bharti N, Patel T, Sukhwani K, Jain A, Sorathiya D, Nanda V, Mehta P, Desai A. Measuring the impact of a single dose of ChAdOx1 nCoV-19 (recombinant) coronavirus vaccine on hospital stay, ICU requirement, and mortality outcome in a tertiary care centre. Int J Infect Dis 2021; 113:282-287. [PMID: 34688949 PMCID: PMC8529545 DOI: 10.1016/j.ijid.2021.10.032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/12/2021] [Accepted: 10/14/2021] [Indexed: 01/04/2023] Open
Abstract
OBJECTIVE To comparatively evaluate ICU requirement, length of stay, and mortality between single-dose vaccinated and non-vaccinated hospitalized COVID-19 patients. DESIGN A retrospective observational study was carried out in a tertiary care hospital in western Indian, from April 1 to June 30, 2021. RESULTS Of the 569 patients who fulfilled the eligibility criteria and were enrolled in the study, 137 (24.08%) patients had received a single dose of ChAdOx1 nCoV-19 vaccine, while 432 (75.92%) patients had not received any form of vaccination. The overall length of stay in hospital was similar for both groups; however, a significant difference was seen in length of stay in the ward and in the ICU. Vaccinated patients were admitted to the ward for 6.21 ± 3.204 days, while non-vaccinated patients were admitted for 5.56 ± 4.55 days (p < 0.001). The mean length of ICU stay for the 21 vaccinated patients requiring intensive care was 4.47 ± 2.3 days, while that for the 145 non-vaccinated patients was 6.29 ± 2.19 days (p < 0.001). Mortality was observed in four patients in the vaccinated group and in 95 patients in the non-vaccinated group. CONCLUSION A single dose of ChAdOx1 nCoV-19 vaccine was associated with a significantly lower severity of SARS-CoV-2 infection compared with no vaccination.
Collapse
Affiliation(s)
- Anuja Desai
- Department of Ophthalmology, Kusum Dhirajlal Hospital, Vaishnodevi Circle, SG Road, Ahmedabad, 382421, Gujarat, India.
| | - Parth Desai
- Kusum Dhirajlal Hospital, Vaishnodevi Circle, SG Road, Ahmedabad, 382421, Gujarat, India.
| | - Jigar Mehta
- Department of Critical Care Medicine, Kusum Dhirajlal Hospital, Vaishnodevi Circle, SG Road, Ahmedabad, 382421, Gujarat, India.
| | - Wasimahmed Sachora
- Department of General Medicine, Kusum Dhirajlal Hospital, Vaishnodevi Circle, SG Road, Ahmedabad, 382421, Gujarat, India.
| | - Neeraj Bharti
- Department of General Medicine, Kusum Dhirajlal Hospital, Vaishnodevi Circle, SG Road, Ahmedabad, 382421, Gujarat, India.
| | - Tushar Patel
- Department of Pulmonary Medicine, Kusum Dhirajlal Hospital, Vaishnodevi Circle, SG Road, Ahmedabad, 382421, Gujarat, India.
| | - Kalpesh Sukhwani
- Department of Infectious Disease, Kusum Dhirajlal Hospital, Vaishnodevi Circle, SG Road, Ahmedabad, 382421, Gujarat, India.
| | - Ankita Jain
- Department of Obstetrics and Gynecology, Kusum Dhirajlal Hospital, Vaishnodevi Circle, SG Road, Ahmedabad, 382421, Gujarat, India.
| | - Dipesh Sorathiya
- Department of In-vitro Fertilization, Kusum Dhirajlal Hospital, Vaishnodevi Circle, SG Road, Ahmedabad, 382421, Gujarat, India.
| | - Vivek Nanda
- Department of Emergency Medicine, Kusum Dhirajlal Hospital, Vaishnodevi Circle, SG Road, Ahmedabad,382421, Gujarat, India.
| | - Parin Mehta
- Department of Ophthalmology, Kusum Dhirajlal Hospital, Vaishnodevi Circle, SG Road, Ahmedabad, 382421, Gujarat, India.
| | - Adit Desai
- Kusum Dhirajlal Hospital, Vaishnodevi Circle, SG Road, Ahmedabad, 382421, Gujarat, India.
| |
Collapse
|
1476
|
Luxi N, Giovanazzi A, Capuano A, Crisafulli S, Cutroneo PM, Fantini MP, Ferrajolo C, Moretti U, Poluzzi E, Raschi E, Ravaldi C, Reno C, Tuccori M, Vannacci A, Zanoni G, Trifirò G. COVID-19 Vaccination in Pregnancy, Paediatrics, Immunocompromised Patients, and Persons with History of Allergy or Prior SARS-CoV-2 Infection: Overview of Current Recommendations and Pre- and Post-Marketing Evidence for Vaccine Efficacy and Safety. Drug Saf 2021; 44:1247-1269. [PMID: 34739716 PMCID: PMC8569292 DOI: 10.1007/s40264-021-01131-6] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2021] [Indexed: 12/24/2022]
Abstract
To date, four vaccines have been authorised for emergency use and under conditional approval by the European Medicines Agency to prevent COVID-19: Comirnaty, COVID-19 Vaccine Janssen, Spikevax (previously COVID-19 Vaccine Moderna) and Vaxzevria (previously COVID-19 Vaccine AstraZeneca). Although the benefit-risk profile of these vaccines was proven to be largely favourable in the general population, evidence in special cohorts initially excluded from the pivotal trials, such as pregnant and breastfeeding women, children/adolescents, immunocompromised people and persons with a history of allergy or previous SARS-CoV-2 infection, is still limited. In this narrative review, we critically overview pre- and post-marketing evidence on the potential benefits and risks of marketed COVID-19 vaccines in the above-mentioned special cohorts. In addition, we summarise the recommendations of the scientific societies and regulatory agencies about COVID-19 primary prevention in the same vaccinee categories.
Collapse
Affiliation(s)
- Nicoletta Luxi
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, Piazzale L.A. Scuro 10, 37134, Verona, Italy
| | - Alexia Giovanazzi
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, Piazzale L.A. Scuro 10, 37134, Verona, Italy
| | - Annalisa Capuano
- Section of Pharmacology "L. Donatelli", Department of Experimental Medicine, Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Salvatore Crisafulli
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Paola Maria Cutroneo
- Sicilian Regional Pharmacovigilance Centre, University Hospital of Messina, Messina, Italy
| | - Maria Pia Fantini
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Carmen Ferrajolo
- Section of Pharmacology "L. Donatelli", Department of Experimental Medicine, Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Ugo Moretti
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, Piazzale L.A. Scuro 10, 37134, Verona, Italy
| | - Elisabetta Poluzzi
- Department of Medical and Surgical Science, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Emanuel Raschi
- Department of Medical and Surgical Science, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Claudia Ravaldi
- PeaRL-Perinatal Research Laboratory, NEUROFARBA Department, University of Florence and CiaoLapo Foundation for Perinatal Health, Florence, Italy
| | - Chiara Reno
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Marco Tuccori
- Unit of Adverse Drug Reactions Monitoring, University Hospital of Pisa, Pisa, Italy
| | - Alfredo Vannacci
- PeaRL-Perinatal Research Laboratory, NEUROFARBA Department, University of Florence and CiaoLapo Foundation for Perinatal Health, Florence, Italy
| | | | - Gianluca Trifirò
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, Piazzale L.A. Scuro 10, 37134, Verona, Italy.
| |
Collapse
|
1477
|
An Overview of COVID-19 and Its Vaccines. BIOLOGY BULLETIN REVIEWS 2021. [PMCID: PMC8762443 DOI: 10.1134/s2079086421070069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Coronavirus pandemic, is a continuing catastrophe (COVID-19) triggered by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). The virus passes into the target cells by attaching itself to a receptor i.e., Human Angiotensin-converting enzyme 2 (hACE2). It consists of Spike structures created from glycoproteins that promote the virus entry into the target cells of host. The RBD of the S1 subunit on Spike proteins binds to the hACE2 receptor, which is mostly found in the lungs, particularly type-2 pneumocytes, causing human ACE2 receptors to be downregulated. Apart from nausea, vomiting, and chest tightness, which are unusual symptoms of COVID-19, the most common causes of death and severity are respiratory failure (69.5%), sepsis or multi-organ failure (28%), cardiac failure (14.6%), and renal failure (14.6%). Viral antigen-based or viral nucleic acid-based real-time RT-PCR is recommended for the diagnosis of COVID-19 suspects. Vaccination is essential for antiviral treatment. The study was conducted on viruses based on live-attenuated or non-activated viruses, recombinant viral vectors, DNA, VLPs and soluble proteins. Vaccine from Pfizer and BioNTech was the first that showed promising data on effectiveness. 90% efficacy of the vaccine was reported. BNT-162b2 (Pfizer, BioNTech) & mRNA-1273 (Moderna) are mRNA based; AZD-1222 Ad5‑CoV (AstraZeneca; Oxford University); Ad26.COV2.S (Johnson & Johnson) are viral vector based and other vaccines have been granted emergency use authorization by FDA. Since RNA viruses are able to mutate readily and quickly the mutation in the existing strain can be Variants of concern (VOCs) that might diminish vaccine effectiveness. The latest Delta variant (B.1.671.2) has rapidly spread in India is emerging in the United States in mid-2021. It turns out to be the chief which is 6–8 fold more resistant to neutralization by sera from COVID-19 convalescent and mRNA vaccinated individuals.
Collapse
|
1478
|
Toscano S, Chisari CG, Patti F. Multiple Sclerosis, COVID-19 and Vaccines: Making the Point. Neurol Ther 2021; 10:627-649. [PMID: 34625925 PMCID: PMC8500471 DOI: 10.1007/s40120-021-00288-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 09/22/2021] [Indexed: 12/15/2022] Open
Abstract
On 11 March 2020, the World Health Organization declared the coronavirus disease 19 (COVID-19) outbreak a pandemic. In this context, several studies and clinical trials have been conducted since then, and many are currently ongoing, leading to the development of several COVID-19 vaccines with different mechanisms of action. People affected by multiple sclerosis (MS) have been considered high-risk subjects in most countries and prioritized for COVID-19 vaccination. However, the management of MS during the COVID-19 pandemic has represented a new challenge for MS specialists, particularly because of the initial lack of guidelines and differing recommendations. Despite an initial hesitation in prescribing disease-modifying drugs (DMDs) in naïve and already treated patients with MS, most national neurology associations and organizations agree on not stopping treatment. However, care is needed especially for patients treated with immune-depleting drugs, which also require some attentions in programming vaccine administration. Many discoveries and new research results have accumulated in a short time on COVID-19, resulting in a need for summarizing the existing evidence on this topic. In this review, we describe the latest research results on the immunological aspects of SARS-CoV-2 infection speculating about their impact on COVID-19 vaccines' mechanisms of action and focused on the management of MS during the COVID pandemic according to the most recent guidelines and recommendations. Finally, the efficacy of COVID-19 and other well-known vaccines against infectious disease in patients with MS on DMDs is discussed.
Collapse
Affiliation(s)
- Simona Toscano
- Department G. F. Ingrassia, Section of Neurosciences, University of Catania, Via Santa Sofia 78, 95123, Catania, Italy
| | - Clara G Chisari
- Department G. F. Ingrassia, Section of Neurosciences, University of Catania, Via Santa Sofia 78, 95123, Catania, Italy
| | - Francesco Patti
- Department G. F. Ingrassia, Section of Neurosciences, University of Catania, Via Santa Sofia 78, 95123, Catania, Italy.
| |
Collapse
|
1479
|
Milota T, Strizova Z, Smetanova J, Sediva A. An immunologist's perspective on anti-COVID-19 vaccines. Curr Opin Allergy Clin Immunol 2021; 21:545-552. [PMID: 34545040 DOI: 10.1097/aci.0000000000000788] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
PURPOSE OF REVIEW Antisevere acute respiratory syndrome-corona virus 2 (SARS-CoV-2) vaccines may provide prompt, effective, and safe solution for the COVID-19 pandemic. Several vaccine candidates have been evaluated in randomized clinical trials (RCTs). Furthermore, data from observational studies mimicking real-life practice and studies on specific groups, such as pregnant women or immunocompromised patients who were excluded from RCTs, are currently available. The main aim of the review is to summarize and provide an immunologist's view on mechanism of action, efficacy and safety, and future challenges in vaccination against SARS-CoV-2. RECENT FINDINGS mRNA and recombinant viral vector-based vaccines have been approved for conditional use in Europe and the USA. They show robust humoral and cellular responses, high with efficacy in prevention of COVID-19 infection (66.9 95%) and favorable safety profile in RCTs. High efficacy of 80-92% was observed in real-life practice. A pilot study also confirmed good safety profile of the mRNA vaccines in pregnant women. Unlike in those with secondary immunodeficiencies where postvaccination responses did not occur, encouraging results were obtained in patients with inborn errors of immunity. SUMMARY Although both RCTs and observational studies suggest good efficacy and safety profiles of the vaccines, their long-term efficacy and safety are still being discussed. Despite the promising results, clinical evidence for specific groups such as children, pregnant and breastfeeding women, and immunocompromised patients, and for novel virus variants are lacking. VIDEO ABSTRACT http://links.lww.com/COAI/A21.
Collapse
Affiliation(s)
- Tomas Milota
- Department of Immunology, Second Faculty of Medicine, Charles University
- Department of Pediatric and Adult Rheumatology, Motol University Hospital, Prague, Czech Republic
| | - Zuzana Strizova
- Department of Immunology, Second Faculty of Medicine, Charles University
| | - Jitka Smetanova
- Department of Immunology, Second Faculty of Medicine, Charles University
| | - Anna Sediva
- Department of Immunology, Second Faculty of Medicine, Charles University
| |
Collapse
|
1480
|
Rodríguez C, Pérez-Nieva A, Máiz L, Meijón MDM, Llamas P, Monreal M, Bikdeli B, Jiménez D. Vaccine-induced immune thrombotic thrombocytopenia after the BNT162b2 mRNA Covid-19 vaccine: A case study. Thromb Res 2021; 208:1-3. [PMID: 34649161 PMCID: PMC8501513 DOI: 10.1016/j.thromres.2021.10.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/30/2021] [Accepted: 10/03/2021] [Indexed: 12/16/2022]
Affiliation(s)
- Carmen Rodríguez
- Respiratory Department, Ramón y Cajal Hospital, IRYCIS, Madrid, Spain
| | - Alberto Pérez-Nieva
- Department of Internal Medicine, Ramón y Cajal Hospital, IRYCIS, Madrid, Spain
| | - Luis Máiz
- Respiratory Department, Ramón y Cajal Hospital, IRYCIS, Madrid, Spain
| | | | - Pilar Llamas
- Hematology Department, Fundación Jiménez Díaz, Madrid, Spain
| | - Manuel Monreal
- Department of Internal Medicine, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain,CIBER de Enfermedades Respiratorias, CIBERES, Madrid, Spain
| | - Behnood Bikdeli
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA,Center for Outcomes Research and Evaluation (CORE), Yale School of Medicine, New Haven, CT, USA,Clinical Trials Center, Cardiovascular Research Foundation, New York, NY, USA
| | - David Jiménez
- Respiratory Department, Ramón y Cajal Hospital, IRYCIS, Madrid, Spain,CIBER de Enfermedades Respiratorias, CIBERES, Madrid, Spain,Corresponding author at: Respiratory Department, Ramón y Cajal Hospital, IRYCIS, Alcalá University, 28034 Madrid, Spain
| |
Collapse
|
1481
|
Nordström P, Ballin M, Nordström A. Effectiveness of heterologous ChAdOx1 nCoV-19 and mRNA prime-boost vaccination against symptomatic Covid-19 infection in Sweden: A nationwide cohort study. THE LANCET REGIONAL HEALTH. EUROPE 2021; 11:100249. [PMID: 34693387 PMCID: PMC8520818 DOI: 10.1016/j.lanepe.2021.100249] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND The effectiveness of heterologous prime-boost Coronavirus disease 2019 (Covid-19) vaccination is currently unknown. METHODS From individuals vaccinated with two doses against Covid-19 in Sweden until July 5, 2021 (N=3,445,061), we formed a study cohort including 94,569 individuals that had received heterologous ChAdOx1 nCoV-19 / BNT162b2 prime-boost vaccination, 16,402 individuals that received heterologous ChAdOx1 nCoV-19 / mRNA-1273 prime-boost vaccination, and 430,100 individuals that received homologous ChAdOx1 nCoV-19 / ChAdOx1 nCoV-19 prime-boost vaccination. In addition, 180,716 individuals were selected who were unvaccinated at the date of vaccination in the corresponding case. Unvaccinated individuals were censored at first dose of any vaccine. Baseline was the date of the second dose of any vaccine, with the same date in the corresponding unvaccinated individual. The outcome included incident symptomatic Covid-19 infection occurring >14 days after baseline. FINDINGS During a mean follow-up time of 76 (range 1-183) days, symptomatic Covid-19 infection was confirmed in 187 individuals with heterologous vaccine schedules (incidence rate: 2.0/100,000 person-days) and in 306 individuals from the unvaccinated control group (incidence rate: 7.1/100,000 person-days). The adjusted vaccine effectiveness was 67% (95% CI, 59-73, P<0.001) for heterologous ChAdOx1 nCoV-19 / BNT162b2 prime-boost vaccination, and 79% (95% CI, 62-88, P<0.001) for heterologous ChAdOx1 nCoV-19 / mRNA-1273 prime-boost vaccination. When combined and analysed together, the two heterologous vaccine schedules had an effectiveness of 68% (95% CI, 61-74, P<0.001) which was significantly greater (Pinteraction<0.001) than the 50% effectiveness for homologous ChAdOx1 nCoV-19 / ChAdOx1 nCoV-19 (95% CI, 41-58, P<0.001). INTERPRETATION The findings of this study suggest that the use of heterologous ChAdOx1 nCoV-19 and mRNA prime-boost vaccination is an effective alternative to increase population immunity against Covid-19, including against the Delta variant which dominated the confirmed cases during the study period. These findings could have important implications for vaccination strategies and logistics, and consequently in the battle against the Covid-19 pandemic.
Collapse
Affiliation(s)
- Peter Nordström
- Department of Community Medicine and Rehabilitation, Unit of Geriatric Medicine, Umeå University, Umeå, Sweden (Peter Nordström and Marcel Ballin)
| | - Marcel Ballin
- Department of Community Medicine and Rehabilitation, Unit of Geriatric Medicine, Umeå University, Umeå, Sweden (Peter Nordström and Marcel Ballin)
- Department of Public Health and Clinical Medicine, Section of Sustainable Health, Umeå University, Umeå, Sweden (Marcel Ballin and Anna Nordström)
| | - Anna Nordström
- Department of Public Health and Clinical Medicine, Section of Sustainable Health, Umeå University, Umeå, Sweden (Marcel Ballin and Anna Nordström)
- School of Sport Sciences, UiT the Arctic University of Norway, Tromsø, Norway (Anna Nordström)
| |
Collapse
|
1482
|
Chi G, Memar Montazerin S, Lee JJ, Kazmi SHA, Shojaei F, Fitzgerald C, Gibson CM. Effect of azithromycin and hydroxychloroquine in patients hospitalized with COVID-19: Network meta-analysis of randomized controlled trials. J Med Virol 2021; 93:6737-6749. [PMID: 34370328 PMCID: PMC8427058 DOI: 10.1002/jmv.27259] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 08/04/2021] [Indexed: 12/15/2022]
Abstract
Chloroquine or its derivative hydroxychloroquine (HCQ) combined with or without azithromycin (AZ) have been widely investigated in observational studies as a treatment option for coronavirus 2019 (COVID-19) infection. The network meta-analysis aims to summarize evidence from randomized controlled trials (RCTs) to determine if AZ or HCQ is associated with improved clinical outcomes. PubMed and Embase were searched from inception to March 7, 2021. We included published RCTs that investigated the efficacy of AZ, HCQ, or its combination among hospitalized patients with COVID-19 infection. The outcomes of interest were all-cause mortality and the use of mechanical ventilation. The pooled odds ratio was calculated using a random-effect model. A total of 10 RCTs were analyzed. Participant's mean age ranged from 40.4 to 66.5 years. There was no significant effect on mortality associated with AZ plus HCQ (odds ratio [OR] = 0.562 [95% confidence interval {CI}: 0.168-1.887]), AZ alone (OR = 0.965 [95% CI: 0.865-1.077]), or HCQ alone (OR = 1.122 [95% CI: 0.995-1.266]; p = 0.06). Similarly, based on pooled effect sizes derived from direct and indirect evidence, none of the treatments had a significant benefit in decreasing the use of mechanical ventilation. No heterogeneity was identified (Cochran's Q = 1.68; p = 0.95; τ2 = 0; I2 = 0% [95% CI: 0%-0%]). Evidence from RCTs suggests that AZ with or without HCQ was not associated with a significant effect on the mortality or mechanical ventilation rates in hospitalized patients with COVID-19. More research is needed to explore therapeutics agents that can effectively reduce the mortality or severity of COVID-19.
Collapse
Affiliation(s)
- Gerald Chi
- Division of Cardiovascular, Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Sahar Memar Montazerin
- Division of Cardiovascular, Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Jane J. Lee
- Department of Trial Design and DevelopmentBaim Institute for Clinical ResearchBostonMassachusettsUSA
| | - Syed Hassan A. Kazmi
- Division of Cardiovascular, Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Fahimehalsadat Shojaei
- Division of Cardiovascular, Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Clara Fitzgerald
- Division of Cardiovascular, Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - C. Michael Gibson
- Division of Cardiovascular, Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
1483
|
Sengsayadeth S, Broglie L, Savani BN. Effectiveness of COVID-19 vaccination in patients after allogeneic haematopoietic cell transplant: how much protection are we getting? Br J Haematol 2021; 196:809-811. [PMID: 34850381 DOI: 10.1111/bjh.17955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 10/28/2021] [Indexed: 11/27/2022]
Affiliation(s)
- Salyka Sengsayadeth
- Section of Hematology and Stem Cell Transplant, Division of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Larisa Broglie
- Department of Pediatrics, Division of Hematology/Oncology/Blood and Marrow Transplantation, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Bipin N Savani
- Section of Hematology and Stem Cell Transplant, Division of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
1484
|
Hassan W, Kazmi SK, Tahir MJ, Ullah I, Royan HA, Fahriani M, Nainu F, Rosa SGV. Global acceptance and hesitancy of COVID-19 vaccination: A narrative review. NARRA J 2021; 1:e57. [PMID: 38450215 PMCID: PMC10914054 DOI: 10.52225/narra.v1i3.57] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 11/29/2021] [Indexed: 02/05/2023]
Abstract
The coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a major global health threat to human civilization and has disrupted many aspects of the community around the globe. Vaccination is one of the prominent measures to control the COVID-19 pandemic. More than 120 vaccines have entered human clinical trials and at least 8 vaccines have been fully approved. However, the success of the COVID-19 vaccination programs depends on how the community accepts the vaccines. Despite COVID-19 vaccination having been initiated for a while now, more than 50% of the global population have not been vaccinated. In some low- and middle-income countries (LMICs), the vaccine coverage is less than 20%. Since the decision to accept the new vaccine is complex, understanding the factors underpinning vaccine acceptance is critical. This review aimed to summarize the COVID-19 vaccine acceptance rate around the globe as well as its associated determinants. Information from this study might be important to formulate effective strategies to increase the COVID-19 vaccine coverage, and to be able to achieve herd immunity.
Collapse
Affiliation(s)
- Wardah Hassan
- Dow University of Health Sciences, Karachi, Pakistan
| | - Syeda K Kazmi
- Dow University of Health Sciences, Karachi, Pakistan
| | - Muhammad J Tahir
- Lahore General Hospital, Lahore, Pakistan
- Ameer-ud-Din Medical College, Affiliated with University of Health and Sciences, Lahore, Pakistan
| | - Irfan Ullah
- Kabir Medical College, Gandhara University, Peshawar, Pakistan
- Naseer Teaching Hospital, Peshawar, Pakistan
| | - Hibban A Royan
- Center for Indonesian Medical Students' Activities (CIMSA), Banda Aceh, Indonesia
| | - Marhami Fahriani
- Medical Research Unit, School of Medicine, Universitas Syiah Kuala, Banda Aceh, 23111, Indonesia
| | - Firzan Nainu
- Faculty of Pharmacy, Hasanuddin University, Makassar, Sulawesi Selatan, Indonesia
| | - Sandro G V Rosa
- Diretoria de Patentes, Divisão de Farmácia - Instituto Nacional da Propriedade Industrial, Rio de Janeiro, Brasil
- Programa de Pós-Graduação em Ciências Aplicadas a Produtos para Saúde, Faculdade de Farmácia, Universidade Federal Fluminense, Brasil
| |
Collapse
|
1485
|
Chojecka D, Pytlos J, Zawadka M, Andruszkiewicz P, Szarpak Ł, Dzieciątkowski T, Jaguszewski MJ, Filipiak KJ, Gąsecka A. How to Maintain Safety and Maximize the Efficacy of Cardiopulmonary Resuscitation in COVID-19 Patients: Insights from the Recent Guidelines. J Clin Med 2021; 10:jcm10235667. [PMID: 34884368 PMCID: PMC8658351 DOI: 10.3390/jcm10235667] [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: 10/18/2021] [Revised: 11/22/2021] [Accepted: 11/30/2021] [Indexed: 11/16/2022] Open
Abstract
Since December 2019, the novel coronavirus disease 2019 (COVID-19) caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has remained a challenge for governments and healthcare systems all around the globe. SARS-CoV-2 infection is associated with increased rates of hospital admissions and significant mortality. The pandemic increased the rate of cardiac arrest and the need for cardiopulmonary resuscitation (CPR). COVID-19, with its pathophysiology and detrimental effects on healthcare, influenced the profile of patients suffering from cardiac arrest, as well as the conditions of performing CPR. To ensure both the safety of medical personnel and the CPR efficacy for patients, resuscitation societies have published modified guidelines addressing the specific reality of the COVID-19 pandemic. In this review, we briefly describe the transmission and pathophysiology of COVID-19, present the challenges of CPR in SARS-CoV-2-infected patients, summarize the current recommendations regarding the algorithms of basic life support (BLS), advanced life support (ALS) and pediatric life support, and discuss other aspects of CPR in COVID-19 patients, which potentially affect the risk-to-benefit ratio of medical procedures and therefore should be considered while formulating further recommendations.
Collapse
Affiliation(s)
- Dominika Chojecka
- 1st Chair and Department of Cardiology, Medical University of Warsaw, 02-091 Warsaw, Poland; (D.C.); (J.P.)
| | - Jakub Pytlos
- 1st Chair and Department of Cardiology, Medical University of Warsaw, 02-091 Warsaw, Poland; (D.C.); (J.P.)
| | - Mateusz Zawadka
- 2nd Department of Anesthesia and Intensive Care, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.Z.); (P.A.)
| | - Paweł Andruszkiewicz
- 2nd Department of Anesthesia and Intensive Care, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.Z.); (P.A.)
| | - Łukasz Szarpak
- Department of Clinical Sciences, Maria Sklodowska-Curie Bialystok Oncology Center, 15-027 Bialystok, Poland;
- Department of Clinical Sciences, Maria Sklodowska-Curie Medical Academy in Warsaw, 00-136 Warsaw, Poland;
| | - Tomasz Dzieciątkowski
- Chair and Department of Medical Microbiology, Medical University of Warsaw, 02-091 Warsaw, Poland;
| | | | - Krzysztof Jerzy Filipiak
- Department of Clinical Sciences, Maria Sklodowska-Curie Medical Academy in Warsaw, 00-136 Warsaw, Poland;
| | - Aleksandra Gąsecka
- 1st Chair and Department of Cardiology, Medical University of Warsaw, 02-091 Warsaw, Poland; (D.C.); (J.P.)
- Correspondence:
| |
Collapse
|
1486
|
Prates-Syed WA, Chaves LCS, Crema KP, Vuitika L, Lira A, Côrtes N, Kersten V, Guimarães FEG, Sadraeian M, Barroso da Silva FL, Cabral-Marques O, Barbuto JAM, Russo M, Câmara NOS, Cabral-Miranda G. VLP-Based COVID-19 Vaccines: An Adaptable Technology against the Threat of New Variants. Vaccines (Basel) 2021; 9:1409. [PMID: 34960155 PMCID: PMC8708688 DOI: 10.3390/vaccines9121409] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/18/2021] [Accepted: 11/20/2021] [Indexed: 12/23/2022] Open
Abstract
Virus-like particles (VLPs) are a versatile, safe, and highly immunogenic vaccine platform. Recently, there are developmental vaccines targeting SARS-CoV-2, the causative agent of COVID-19. The COVID-19 pandemic affected humanity worldwide, bringing out incomputable human and financial losses. The race for better, more efficacious vaccines is happening almost simultaneously as the virus increasingly produces variants of concern (VOCs). The VOCs Alpha, Beta, Gamma, and Delta share common mutations mainly in the spike receptor-binding domain (RBD), demonstrating convergent evolution, associated with increased transmissibility and immune evasion. Thus, the identification and understanding of these mutations is crucial for the production of new, optimized vaccines. The use of a very flexible vaccine platform in COVID-19 vaccine development is an important feature that cannot be ignored. Incorporating the spike protein and its variations into VLP vaccines is a desirable strategy as the morphology and size of VLPs allows for better presentation of several different antigens. Furthermore, VLPs elicit robust humoral and cellular immune responses, which are safe, and have been studied not only against SARS-CoV-2 but against other coronaviruses as well. Here, we describe the recent advances and improvements in vaccine development using VLP technology.
Collapse
Affiliation(s)
- Wasim A. Prates-Syed
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (ICB/USP), São Paulo 05508000, SP, Brazil; (W.A.P.-S.); (K.P.C.); (L.V.); (A.L.); (N.C.); (V.K.); (O.C.-M.); (J.A.M.B.); (M.R.); (N.O.S.C.)
- Institute of Research and Education in Child Health (PENSI), São Paulo 01228200, SP, Brazil
| | - Lorena C. S. Chaves
- Department of Microbiology and Immunology, School of Medicine, Emory University, Claudia Nance Rollins Building, Atlanta, GA 30329, USA;
| | - Karin P. Crema
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (ICB/USP), São Paulo 05508000, SP, Brazil; (W.A.P.-S.); (K.P.C.); (L.V.); (A.L.); (N.C.); (V.K.); (O.C.-M.); (J.A.M.B.); (M.R.); (N.O.S.C.)
- Institute of Research and Education in Child Health (PENSI), São Paulo 01228200, SP, Brazil
| | - Larissa Vuitika
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (ICB/USP), São Paulo 05508000, SP, Brazil; (W.A.P.-S.); (K.P.C.); (L.V.); (A.L.); (N.C.); (V.K.); (O.C.-M.); (J.A.M.B.); (M.R.); (N.O.S.C.)
| | - Aline Lira
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (ICB/USP), São Paulo 05508000, SP, Brazil; (W.A.P.-S.); (K.P.C.); (L.V.); (A.L.); (N.C.); (V.K.); (O.C.-M.); (J.A.M.B.); (M.R.); (N.O.S.C.)
- Institute of Research and Education in Child Health (PENSI), São Paulo 01228200, SP, Brazil
| | - Nelson Côrtes
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (ICB/USP), São Paulo 05508000, SP, Brazil; (W.A.P.-S.); (K.P.C.); (L.V.); (A.L.); (N.C.); (V.K.); (O.C.-M.); (J.A.M.B.); (M.R.); (N.O.S.C.)
- Institute of Research and Education in Child Health (PENSI), São Paulo 01228200, SP, Brazil
| | - Victor Kersten
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (ICB/USP), São Paulo 05508000, SP, Brazil; (W.A.P.-S.); (K.P.C.); (L.V.); (A.L.); (N.C.); (V.K.); (O.C.-M.); (J.A.M.B.); (M.R.); (N.O.S.C.)
| | | | - Mohammad Sadraeian
- São Carlos Institute of Physics, IFSC-USP, São Carlos 13566590, SP, Brazil; (F.E.G.G.); (M.S.)
- Institute for Biomedical Materials & Devices (IBMD), Faculty of Science, University of Technology, Sydney, NSW 2007, Australia
| | - Fernando L. Barroso da Silva
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040903, SP, Brazil;
- Department of Chemical and Biomolecular Engeneering, North Carolina State University, Raleigh, NC 27695, USA
| | - Otávio Cabral-Marques
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (ICB/USP), São Paulo 05508000, SP, Brazil; (W.A.P.-S.); (K.P.C.); (L.V.); (A.L.); (N.C.); (V.K.); (O.C.-M.); (J.A.M.B.); (M.R.); (N.O.S.C.)
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508000, SP, Brazil
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Children’s Medical Center, Tehran 1419733151, Iran
| | - José A. M. Barbuto
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (ICB/USP), São Paulo 05508000, SP, Brazil; (W.A.P.-S.); (K.P.C.); (L.V.); (A.L.); (N.C.); (V.K.); (O.C.-M.); (J.A.M.B.); (M.R.); (N.O.S.C.)
- Laboratory of Medical Investigation in Pathogenesis and Targeted Therapy in Onco-Immuno-Hematology (LIM-31), Department of Hematology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo 0124690, SP, Brazil
| | - Momtchilo Russo
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (ICB/USP), São Paulo 05508000, SP, Brazil; (W.A.P.-S.); (K.P.C.); (L.V.); (A.L.); (N.C.); (V.K.); (O.C.-M.); (J.A.M.B.); (M.R.); (N.O.S.C.)
| | - Niels O. S. Câmara
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (ICB/USP), São Paulo 05508000, SP, Brazil; (W.A.P.-S.); (K.P.C.); (L.V.); (A.L.); (N.C.); (V.K.); (O.C.-M.); (J.A.M.B.); (M.R.); (N.O.S.C.)
| | - Gustavo Cabral-Miranda
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (ICB/USP), São Paulo 05508000, SP, Brazil; (W.A.P.-S.); (K.P.C.); (L.V.); (A.L.); (N.C.); (V.K.); (O.C.-M.); (J.A.M.B.); (M.R.); (N.O.S.C.)
- Institute of Research and Education in Child Health (PENSI), São Paulo 01228200, SP, Brazil
| |
Collapse
|
1487
|
The Function of Immunoproteasomes-An Immunologists' Perspective. Cells 2021; 10:cells10123360. [PMID: 34943869 PMCID: PMC8699091 DOI: 10.3390/cells10123360] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/23/2021] [Accepted: 11/25/2021] [Indexed: 01/02/2023] Open
Abstract
Proteasomes are responsible for intracellular proteolysis and play an important role in cellular protein homeostasis. Cells of the immune system assemble a specialized form of proteasomes, known as immunoproteasomes, in which the constitutive catalytic sites are replaced for cytokine-inducible homologues. While immunoproteasomes may fulfill all standard proteasome’ functions, they seem specially adapted for a role in MHC class I antigen processing and CD8+ T-cell activation. In this way, they may contribute to CD8+ T-cell-mediated control of intracellular infections, but also to the immunopathogenesis of autoimmune diseases. Starting at the discovery of its catalytic subunits in the genome, here, we review the observations shaping our current understanding of immunoproteasome function, and the consequential novel opportunities for immune intervention.
Collapse
|
1488
|
Lin L, Liu Y, Tang X, He D. The Disease Severity and Clinical Outcomes of the SARS-CoV-2 Variants of Concern. Front Public Health 2021; 9:775224. [PMID: 34917580 PMCID: PMC8669511 DOI: 10.3389/fpubh.2021.775224] [Citation(s) in RCA: 156] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 11/08/2021] [Indexed: 01/06/2023] Open
Abstract
With the continuation of the pandemic, many severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants have appeared around the world. Owing to a possible risk of increasing the transmissibility of the virus, severity of the infected individuals, and the ability to escape the antibody produced by the vaccines, the four SARS-CoV-2 variants of Alpha (B.1.1.7), Beta (B.1.351), Gamma (P.1), and Delta (B.1.617.2) have attracted the most widespread attention. At present, there is a unified conclusion that these four variants have increased the transmissibility of SARS-CoV-2, but the severity of the disease caused by them has not yet been determined. Studies from June 1, 2020 to October 15, 2021 were considered, and a meta-analysis was carried out to process the data. Alpha, Beta, Gamma, and Delta variants are all more serious than the wild-type virus in terms of hospitalization, ICU admission, and mortality, and the Beta and Delta variants have a higher risk than the Alpha and Gamma variants. Notably, the random effects of Beta variant to the wild-type virus with respect to hospitalization rate, severe illness rate, and mortality rate are 2.16 (95% CI: 1.19-3.14), 2.23 (95% CI: 1.31-3.15), and 1.50 (95% CI: 1.26-1.74), respectively, and the random effects of Delta variant to the wild-type virus are 2.08 (95% CI: 1.77-2.39), 3.35 (95% CI: 2.5-4.2), and 2.33 (95% CI: 1.45-3.21), respectively. Although, the emergence of vaccines may reduce the threat posed by SARS-CoV-2 variants, these are still very important, especially the Beta and Delta variants.
Collapse
Affiliation(s)
- Lixin Lin
- Department of Applied Mathematics, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Ying Liu
- School of International Business, Xiamen University Tan Kah Kee College, Zhangzhou, China
| | - Xiujuan Tang
- Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Daihai He
- Department of Applied Mathematics, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| |
Collapse
|
1489
|
Dogra N, Ledesma-Feliciano C, Sen R. Developmental Aspects of SARS-CoV-2, Potential Role of Exosomes and Their Impact on the Human Transcriptome. J Dev Biol 2021; 9:54. [PMID: 34940501 PMCID: PMC8708617 DOI: 10.3390/jdb9040054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/12/2021] [Accepted: 11/23/2021] [Indexed: 12/21/2022] Open
Abstract
With over 4.8 million deaths within 2 years, time is of the essence in combating COVID-19. The infection now shows devastating impacts on the younger population, who were not previously predicted to be vulnerable, such as in the older population. COVID-19-related complications have been reported in neonates whose mothers were infected with SARS-CoV-2 during pregnancy, and in children who get infected. Hence, a deeper understanding of the pathophysiology of COVID-19 during various developmental stages and placental transmission is essential. Although a connection has not yet been established between exosomal trafficking and the placental transmission of COVID-19, reports indicate that SARS-CoV-2 components may be trafficked between cells through exosomes. As the infection spreads, the transcriptome of cells is drastically perturbed, e.g., through the severe upregulation of several immune-related genes. Consequently, a major outcome of COVID-19 is an elevated immune response and the detection of viral RNA transcripts in host tissue. In this direction, this review focuses on SARS-CoV-2 virology, its in utero transmission from infected pregnant mothers to fetuses, SARS-CoV-2 and exosomal cellular trafficking, transcriptomic impacts, and RNA-mediated therapeutics against COVID-19. Future research will establish stronger connections between the above processes to develop diagnostic and therapeutic solutions towards COVID-19 and similar viral outbreaks.
Collapse
Affiliation(s)
- Navneet Dogra
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Carmen Ledesma-Feliciano
- Division of Infectious Diseases, School of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA;
| | - Rwik Sen
- Active Motif, Incorporated, Carlsbad, CA 92008, USA
| |
Collapse
|
1490
|
dos Santos Ferreira CE, Gómez-Dantés H, Junqueira Bellei NC, López E, Nogales Crespo KA, O’Ryan M, Villegas J. The Role of Serology Testing in the Context of Immunization Policies for COVID-19 in Latin American Countries. Viruses 2021; 13:2391. [PMID: 34960660 PMCID: PMC8706237 DOI: 10.3390/v13122391] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/18/2021] [Accepted: 11/18/2021] [Indexed: 12/16/2022] Open
Abstract
This review aims to explore the role and value of serology testing in the context of COVID-19 immunization policies in Latin American countries and the barriers and challenges to the adequate use and uptake of this tool. It builds on a review of the academic literature, evidence, and existing policies, and includes a multistage process of discussion and feedback by a group of five experts. Regional and country-level evidence and resources from five focus countries-Argentina, Brazil, Chile, Colombia, and Mexico-were collected and analyzed. This review contains an overview of (1) the impact of the SARS-CoV-2 pandemic, the variants of concern and current testing strategies, (2) the introduction of COVID-19 vaccination, (3) the potential use of serology testing to support immunization initiatives, (4) the current frameworks for the use of serology testing in the region, and (5) the barriers and challenges to implementing serology testing in the context of COVID-19 immunization policies, including a discussion on the potential actions required to address these barriers and facilitate the uptake of this strategy in the region. Stakeholders can use elements of this document to guide timely decision-making, raise awareness, and inspire further studies.
Collapse
Affiliation(s)
- Carlos E. dos Santos Ferreira
- Clinical Pathology, Hospital Israelita Albert Einstein, São Paulo 05652-900, Brazil;
- Microbiology Sector, Federal University of São Paulo’s Central Laboratory Activities, São Paulo 04088-002, Brazil
- Brazilian Society of Clinical Pathology and Laboratory Medicine, Rio de Janeiro 22220-040, Brazil
| | | | | | - Eduardo López
- Department of Medicine, Hospital de Niños Gutiérrez, Buenos Aires C1425-EFD, Argentina;
- Pediatric Infectious Diseases Program, Faculty of Medicine, University of Buenos Aires, Buenos Aires C1121-ABG, Argentina
- Pediatrics and Vaccinology, Faculty of Medicine, University of Salvador, Buenos Aires C1055-AAG, Argentina
| | | | - Miguel O’Ryan
- Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago de Chile 8380000, Chile;
- Millennium Institute of Immunology and Immunotherapy, University of Chile, Santiago de Chile 8331150, Chile
- Chilean Academy of Medicine, Santiago de Chile 6500445, Chile
| | | |
Collapse
|
1491
|
Yap C, Ali A, Prabhakar A, Prabhakar A, Pal A, Lim YY, Kakodkar P. Comprehensive literature review on COVID-19 vaccines and role of SARS-CoV-2 variants in the pandemic. Ther Adv Vaccines Immunother 2021; 9:25151355211059791. [PMID: 34870090 PMCID: PMC8637774 DOI: 10.1177/25151355211059791] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 10/22/2021] [Indexed: 01/11/2023] Open
Abstract
Since the outbreak of the COVID-19 pandemic, there has been a rapid expansion in vaccine research focusing on exploiting the novel discoveries on the pathophysiology, genomics, and molecular biology of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Although the current preventive measures are primarily socially distancing by maintaining a 1 m distance, it is supplemented using facial masks and other personal hygiene measures. However, the induction of vaccines as primary prevention is crucial to eradicating the disease to attempt restoration to normalcy. This literature review aims to describe the physiology of the vaccines and how the spike protein is used as a target to elicit an antibody-dependent immune response in humans. Furthermore, the overview, dosing strategies, efficacy, and side effects will be discussed for the notable vaccines: BioNTech/Pfizer, Moderna, AstraZeneca, Janssen, Gamaleya, and SinoVac. In addition, the development of other prominent COVID-19 vaccines will be highlighted alongside the sustainability of the vaccine-mediated immune response and current contraindications. As the research is rapidly expanding, we have looked at the association between pregnancy and COVID-19 vaccinations, in addition to the current reviews on the mixing of vaccines. Finally, the prominent emerging variants of concern are described, and the efficacy of the notable vaccines toward these variants has been summarized.
Collapse
Affiliation(s)
- Charles Yap
- School of Medicine, National University of
Ireland, Galway, Ireland
| | - Abulhassan Ali
- School of Medicine, National University of
Ireland, Galway, Ireland
| | - Amogh Prabhakar
- School of Medicine, National University of
Ireland, Galway, Ireland
| | - Akul Prabhakar
- School of Medicine, National University of
Ireland, Galway, Ireland
| | - Aman Pal
- School of Medicine, National University of
Ireland, Galway, Ireland
| | - Ying Yi Lim
- School of Medicine, National University of
Ireland, Galway, Ireland
| | - Pramath Kakodkar
- School of Medicine, National University of
Ireland, Galway, University Road, Galway H91 TK33, Ireland
| |
Collapse
|
1492
|
Safety and Seroconversion of Immunotherapies against SARS-CoV-2 Infection: A Systematic Review and Meta-Analysis of Clinical Trials. Pathogens 2021; 10:pathogens10121537. [PMID: 34959492 PMCID: PMC8706687 DOI: 10.3390/pathogens10121537] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 11/17/2021] [Accepted: 11/19/2021] [Indexed: 02/06/2023] Open
Abstract
Clinical trials evaluating the safety and antibody response of strategies to manipulate prophylactic and therapeutic immunity have been launched. We aim to evaluate strategies for augmentation of host immunity against severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection. We searched clinical trials registered at the National Institutes of Health by 25 May 2021 and conducted analyses on inoculated populations, involved immunological processes, source of injected components, and trial phases. We then searched PubMed, Embase, Scopus, and the Cochrane Central Register of Controlled Trials for their corresponding reports published by 25 May 2021. A bivariate, random-effects meta-analysis was used to derive the pooled estimate of seroconversion and adverse events (AEs). A total of 929,359 participants were enrolled in 389 identified trials. The working mechanisms included heterologous immunity, active immunity, passive immunity, and immunotherapy, with 62.4% of the trials on vaccines. A total of 9072 healthy adults from 27 publications for 22 clinical trials on active immunity implementing vaccination were included for meta-analyses. The pooled odds ratios (ORs) of seroconversion were 13.94, 84.86, 106.03, and 451.04 (all p < 0.01) for vaccines based on protein, RNA, viral vector, and inactivated virus, compared with that of respective placebo/control treatment or pre-vaccination sera. The pooled ORs for safety, as defined by the inverse of systemic adverse events (AEs) were 0.53 (95% CI = 0.27–1.05; p = 0.07), 0.35 (95% CI = 0.16–0.75; p = 0.007), 0.32 (95% CI = 0.19–0.55; p < 0.0001), and 1.00 (95% CI = 0.73–1.36; p = 0.98) for vaccines based on protein, RNA, viral vector, and inactivated virus, compared with that of placebo/control treatment. A paradigm shift from all four immune-augmentative interventions to active immunity implementing vaccination was observed through clinical trials. The efficacy of immune responses to neutralize SARS-CoV-2 for these vaccines was promising, although systemic AEs were still evident for RNA-based and viral vector-based vaccines.
Collapse
|
1493
|
Khandker SS, Godman B, Jawad MI, Meghla BA, Tisha TA, Khondoker MU, Haq MA, Charan J, Talukder AA, Azmuda N, Sharmin S, Jamiruddin MR, Haque M, Adnan N. A Systematic Review on COVID-19 Vaccine Strategies, Their Effectiveness, and Issues. Vaccines (Basel) 2021; 9:1387. [PMID: 34960133 PMCID: PMC8708628 DOI: 10.3390/vaccines9121387] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/18/2021] [Accepted: 11/18/2021] [Indexed: 12/13/2022] Open
Abstract
COVID-19 vaccines are indispensable, with the number of cases and mortality still rising, and currently no medicines are routinely available for reducing morbidity and mortality, apart from dexamethasone, although others are being trialed and launched. To date, only a limited number of vaccines have been given emergency use authorization by the US Food and Drug Administration and the European Medicines Agency. There is a need to systematically review the existing vaccine candidates and investigate their safety, efficacy, immunogenicity, unwanted events, and limitations. The review was undertaken by searching online databases, i.e., Google Scholar, PubMed, and ScienceDirect, with finally 59 studies selected. Our findings showed several types of vaccine candidates with different strategies against SARS-CoV-2, including inactivated, mRNA-based, recombinant, and nanoparticle-based vaccines, are being developed and launched. We have compared these vaccines in terms of their efficacy, side effects, and seroconversion based on data reported in the literature. We found mRNA vaccines appeared to have better efficacy, and inactivated ones had fewer side effects and similar seroconversion in all types of vaccines. Overall, global variant surveillance and systematic tweaking of vaccines, coupled with the evaluation and administering vaccines with the same or different technology in successive doses along with homologous and heterologous prime-booster strategy, have become essential to impede the pandemic. Their effectiveness appreciably outweighs any concerns with any adverse events.
Collapse
Affiliation(s)
- Shahad Saif Khandker
- Gonoshasthaya-RNA Molecular Diagnostic & Research Center, Dhanmondi, Dhaka 1205, Bangladesh; (S.S.K.); (M.U.K.); (M.A.H.); (M.R.J.)
| | - Brian Godman
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G1 1XQ, UK;
- Division of Public Health Pharmacy and Management, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria 0204, South Africa
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman P.O. Box 346, United Arab Emirates
| | - Md. Irfan Jawad
- Department of Microbiology, Jahangirnagar University, Savar 1342, Bangladesh; (M.I.J.); (B.A.M.); (T.A.T.); (A.A.T.); (N.A.)
| | - Bushra Ayat Meghla
- Department of Microbiology, Jahangirnagar University, Savar 1342, Bangladesh; (M.I.J.); (B.A.M.); (T.A.T.); (A.A.T.); (N.A.)
| | - Taslima Akter Tisha
- Department of Microbiology, Jahangirnagar University, Savar 1342, Bangladesh; (M.I.J.); (B.A.M.); (T.A.T.); (A.A.T.); (N.A.)
| | - Mohib Ullah Khondoker
- Gonoshasthaya-RNA Molecular Diagnostic & Research Center, Dhanmondi, Dhaka 1205, Bangladesh; (S.S.K.); (M.U.K.); (M.A.H.); (M.R.J.)
- Department of Community Medicine, Gonoshasthaya Samaj Vittik Medical College, Savar 1344, Bangladesh
| | - Md. Ahsanul Haq
- Gonoshasthaya-RNA Molecular Diagnostic & Research Center, Dhanmondi, Dhaka 1205, Bangladesh; (S.S.K.); (M.U.K.); (M.A.H.); (M.R.J.)
| | - Jaykaran Charan
- Department of Pharmacology, All India Institute of Medical Sciences, Jodhpur 342005, India;
| | - Ali Azam Talukder
- Department of Microbiology, Jahangirnagar University, Savar 1342, Bangladesh; (M.I.J.); (B.A.M.); (T.A.T.); (A.A.T.); (N.A.)
| | - Nafisa Azmuda
- Department of Microbiology, Jahangirnagar University, Savar 1342, Bangladesh; (M.I.J.); (B.A.M.); (T.A.T.); (A.A.T.); (N.A.)
| | - Shahana Sharmin
- Department of Pharmacy, BRAC University, Dhaka 1212, Bangladesh;
| | - Mohd. Raeed Jamiruddin
- Gonoshasthaya-RNA Molecular Diagnostic & Research Center, Dhanmondi, Dhaka 1205, Bangladesh; (S.S.K.); (M.U.K.); (M.A.H.); (M.R.J.)
- Department of Pharmacy, BRAC University, Dhaka 1212, Bangladesh;
| | - Mainul Haque
- The Unit of Pharmacology, Faculty of Medicine and Defence Health, Universiti Pertahanan Nasional Malaysia (National Defence University of Malaysia), Kem Perdana Sugai Besi, Kuala Lumpur 57000, Malaysia
| | - Nihad Adnan
- Gonoshasthaya-RNA Molecular Diagnostic & Research Center, Dhanmondi, Dhaka 1205, Bangladesh; (S.S.K.); (M.U.K.); (M.A.H.); (M.R.J.)
- Department of Microbiology, Jahangirnagar University, Savar 1342, Bangladesh; (M.I.J.); (B.A.M.); (T.A.T.); (A.A.T.); (N.A.)
| |
Collapse
|
1494
|
Maeda K, Amano M, Uemura Y, Tsuchiya K, Matsushima T, Noda K, Shimizu Y, Fujiwara A, Takamatsu Y, Ichikawa Y, Nishimura H, Kinoshita M, Matsumoto S, Gatanaga H, Yoshimura K, Oka SI, Mikami A, Sugiura W, Sato T, Yoshida T, Shimada S, Mitsuya H. Correlates of neutralizing/SARS-CoV-2-S1-binding antibody response with adverse effects and immune kinetics in BNT162b2-vaccinated individuals. Sci Rep 2021; 11:22848. [PMID: 34819514 PMCID: PMC8613264 DOI: 10.1038/s41598-021-01930-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 11/01/2021] [Indexed: 01/08/2023] Open
Abstract
While mRNA vaccines against SARS-CoV-2 are exceedingly effective in preventing symptomatic infection, their immune response features remain to be clarified. In the present prospective study, 225 healthy individuals in Japan, who received two BNT162b2 doses, were enrolled. Correlates of BNT162b2-elicited SARS-CoV-2-neutralizing activity (50% neutralization titer: NT50; assessed using infectious virions) with various determinants were examined and the potency of sera against variants of concerns was determined. Significant rise in NT50s was seen in sera on day 28 post-1st dose. A moderate inverse correlation was seen between NT50s and ages, but no correlation seen between NT50s and adverse effects. NT50s and SARS-CoV-2-S1-binding-IgG levels on day 28 post-1st dose and pain scores following the 2nd dose were greater in women than in men. The average half-life of NT50s was ~ 68 days, and 23.6% (49 out of 208 individuals) failed to show detectable neutralizing activity on day 150. While sera from elite-responders (NT50s > 1,500: the top 4% among the participants) potently to moderately blocked all variants of concerns examined, some sera with low NT50s failed to block the B.1.351-beta strain. Since BNT162b2-elicited immunity against SARS-CoV-2 is short, an additional vaccine or other protective measures are needed.
Collapse
Affiliation(s)
- Kenji Maeda
- Department of Refractory Viral Infections, National Center for Global Health and Medicine (NCGM) Research Institute, Tokyo, Japan.
| | - Masayuki Amano
- Department of Clinical Sciences, Kumamoto University Hospital, Kumamoto, Japan
| | | | | | | | | | | | - Asuka Fujiwara
- Department of Refractory Viral Infections, National Center for Global Health and Medicine (NCGM) Research Institute, Tokyo, Japan
| | - Yuki Takamatsu
- Department of Refractory Viral Infections, National Center for Global Health and Medicine (NCGM) Research Institute, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | - Hiroaki Mitsuya
- Department of Refractory Viral Infections, National Center for Global Health and Medicine (NCGM) Research Institute, Tokyo, Japan.
- Department of Clinical Sciences, Kumamoto University Hospital, Kumamoto, Japan.
- Experimental Retrovirology Section, HIV and AIDS Malignancy Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
1495
|
Gronbeck C, Grant-Kels JM. Attention all anti-vaccinators: The cutaneous adverse events from the mRNA COVID-19 vaccines are not an excuse to avoid them! Clin Dermatol 2021; 39:674-687. [PMID: 34809772 PMCID: PMC8139532 DOI: 10.1016/j.clindermatol.2021.05.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Despite the growing availability of coronavirus disease 2019 (COVID-19) vaccines in the general population, a significant proportion of individuals demonstrate vaccine hesitancy. We sought to consolidate and update current evidence on cutaneous adverse events from COVID-19 vaccines to aid in the education and counseling of patients concerned about potential cutaneous side effects. We conducted a literature review of PubMed in May 2021 to identify reports of cutaneous events after vaccination with the Pfizer-BioNTech and Moderna vaccines (postauthorization clinical reports pertaining to the Johnson & Johnson and AstraZeneca vaccines were limited). Event reports in the Vaccine Adverse Event Reporting System were reviewed. Localized cutaneous reactions were common after the mRNA vaccines, consistent with clinical trial findings. Reported urticarial and morbilliform eruptions may reflect immediate hypersensitivity but have rarely been associated with anaphylaxis. There are infrequent reports of herpes zoster, dermatologic filler reactions, and immune thrombocytopenia, mainly occurring in high-risk patient groups. Ultimately, the identified cutaneous reactions are largely self-limited and should not discourage vaccination. Existing reports should reassure patients of the overall compelling safety profiles of the mRNA COVID-19 vaccines and benignity of skin reactions after vaccination.
Collapse
Affiliation(s)
- Christian Gronbeck
- University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Jane M Grant-Kels
- Department of Dermatology, University of Connecticut School of Medicine, Farmington, Connecticut, USA.
| |
Collapse
|
1496
|
Rotshild V, Hirsh-Raccah B, Miskin I, Muszkat M, Matok I. Comparing the clinical efficacy of COVID-19 vaccines: a systematic review and network meta-analysis. Sci Rep 2021; 11:22777. [PMID: 34815503 PMCID: PMC8611039 DOI: 10.1038/s41598-021-02321-z] [Citation(s) in RCA: 158] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 10/07/2021] [Indexed: 01/07/2023] Open
Abstract
New Coronavirus Disease 2019 (COVID-19) vaccines are available to prevent the ongoing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic. We compared the efficacy of new COVID-19 vaccines to prevent symptomatic and severe disease in the adult population and to prevent symptomatic COVID-19 among the elderly. Leading medical databases were searched until August 30, 2021. Published phase 3 randomized controlled trials (RCTs) evaluated efficacy of the vaccine to prevent symptomatic and sever COVID-19 in adults were included. Two reviewers independently evaluated the literature search results and independently extracted summary data. The risk of bias was evaluated using the Cochrane Risk of Bias Assessment Tool. We performed a network meta-analysis (NMA) according to PRISMA-NMA 2015 to pool indirect comparisons between different vaccines regarding their relative efficacy. The primary outcomes were the efficacy of the vaccine against symptomatic COVID-19 in adults (PROSPERO registration number: CRD42021235364). Above 200,000 adult participants from eight phase 3 RCTs were included in NMA, of whom 52% received the intervention (active COVID-19 vaccine). While each of nine vaccines was tested in the unique clinical trial as compared to control, based on indirect comparison, BNT162b2 and mRNA-1273 vaccines were ranked with the highest probability of efficacy against symptomatic COVID-19 (P-scores 0.952 and 0.843, respectively), followed by Gam-COVID-Vac (P-score 0.782), NVX-CoV23730 (P-score 0.700), CoronaVac (P-score 0.570), BN02 (P-score 0.428), WIV04 (P-score 0.327), and Ad26.COV2.S (P-score 0.198). No statistically significant difference was seen in the ability of the vaccines to prevent symptomatic disease in the elderly population. No vaccine was statistically significantly associated with a decreased risk for severe COVID-19 than other vaccines, although mRNA-1273 and Gam-COVID-Vac have the highest P-scores (0.899 and 0.816, respectively), indicating greater protection against severe disease than other vaccines. In our indirect comparison, the BNT162b2 and mRNA-1273 vaccines, which use mRNA technology, were associated with the highest efficacy to prevent symptomatic COVID-19 compared to other vaccines. This finding may have importance when deciding which vaccine to use, together with other important factors as availability of the vaccines, costs, logistics, side effects, and patient acceptability.
Collapse
Affiliation(s)
- Victoria Rotshild
- Pharmacoepidemiology Research Unit, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, P.O.B. 12272, 9112102, Jerusalem, Israel.
- Jerusalem Distric, Clalith Health Services Community Division, Jerusalem, Israel.
| | - Bruria Hirsh-Raccah
- Pharmacoepidemiology Research Unit, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, P.O.B. 12272, 9112102, Jerusalem, Israel
- Department of Cardiology, Hadassah University Hospital Ein Karem, Jerusalem, Israel
| | - Ian Miskin
- Jerusalem Distric, Clalith Health Services Community Division, Jerusalem, Israel
| | - Mordechai Muszkat
- Department of Medicine, Hadassah University Hospital Mt. Scopus, Jerusalem, Israel
| | - Ilan Matok
- Pharmacoepidemiology Research Unit, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, P.O.B. 12272, 9112102, Jerusalem, Israel
| |
Collapse
|
1497
|
Zhang Y, Banga Ndzouboukou JL, Gan M, Lin X, Fan X. Immune Evasive Effects of SARS-CoV-2 Variants to COVID-19 Emergency Used Vaccines. Front Immunol 2021; 12:771242. [PMID: 34880867 PMCID: PMC8645832 DOI: 10.3389/fimmu.2021.771242] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 10/29/2021] [Indexed: 12/30/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) pandemic is a serious threat to global public health and social and economic development. Various vaccine platforms have been developed rapidly and unprecedentedly, and at least 16 vaccines receive emergency use authorization (EUA). However, the causative pathogen severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has continued to evolve and mutate, emerging lots of viral variants. Several variants have successfully become the predominant strains and spread all over the world because of their ability to evade the pre-existing immunity obtained after previous infections with prototype strain or immunizations. Here, we summarized the prevalence and biological structure of these variants and the efficacy of currently used vaccines against the SARS-CoV-2 variants to provide guidance on how to design vaccines more rationally against the variants.
Collapse
Affiliation(s)
| | | | | | | | - Xionglin Fan
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
1498
|
Ambrosino D, Han HH, Hu B, Liang J, Clemens R, Johnson M, Siber G, Goldblatt D. Immunogenicity of SCB-2019 Coronavirus Disease 2019 Vaccine Compared With 4 Approved Vaccines. J Infect Dis 2021; 225:327-331. [PMID: 34888662 PMCID: PMC8763959 DOI: 10.1093/infdis/jiab574] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/15/2021] [Indexed: 11/17/2022] Open
Abstract
A significant correlation has been shown between the binding antibody responses against original severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein and vaccine efficacy of 4 approved coronavirus disease 2019 vaccines. We therefore assessed the immune response against original SARS-CoV-2 elicited by the adjuvanted S-Trimer vaccine, SCB-2019 + CpG/alum, in the same assay and laboratory. Responses to SCB-2019 were comparable or superior for antibody to original and Alpha variant when compared with 4 approved vaccines. The comparison accurately predicted success of the recently reported efficacy trial of SCB-2019 vaccine. Immunogenicity comparisons to original strain and variants of concern should be considered as a basis for authorization of vaccines.
Collapse
Affiliation(s)
| | | | - Branda Hu
- Clover Biopharmaceuticals, Chengdu, China
| | | | - Ralf Clemens
- Global Research in Infectious Diseases, Rio de Janeiro, Brazil
| | - Marina Johnson
- Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | | | - David Goldblatt
- Great Ormond Street Institute of Child Health, University College London, London, United Kingdom.,Great Ormond Street Children's Hospital NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
1499
|
Sriwastava S, Shrestha AK, Khalid SH, Colantonio MA, Nwafor D, Srivastava S. Spectrum of Neuroimaging Findings in Post-COVID-19 Vaccination: A Case Series and Review of Literature. Neurol Int 2021; 13:622-639. [PMID: 34842783 PMCID: PMC8628885 DOI: 10.3390/neurolint13040061] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/12/2021] [Accepted: 11/17/2021] [Indexed: 11/16/2022] Open
Abstract
Background and Purpose: The novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was first detected in Wuhan, China in December 2019. Symptoms range from mild flu-like symptoms to more severe presentations, including pneumonia, acute respiratory distress syndrome (ARDS), and even death. In response to the COVID-19 pandemic, the Emergency Use Authorization (EUA) approved the use of several vaccines. Because vaccines have been fast-tracked for emergency use, the short and long-term safety profile has been an area of concern. The aim of this paper is to extensively review published literature regarding post-COVID-19 vaccination neurological complications and characterize neuroimaging findings from three case presentations for early diagnosis and treatment. Methods: The analysis includes data from PubMed and Google Scholar. Articles included were retrieved from database inception beginning December 2020 with no language restrictions. Terms used include "SARS-CoV-2", "post Covid vaccination", "neurological complications", "Guillain-barre Syndrome", "Transverse-myelitis", "Cerebral Venous Sinus thrombosis", and "Cerebral hemorrhage". Results: The literature review yielded several neurological complications post vaccination, including cerebral sinus venous thrombosis, transverse myelitis, Guillain-Barré Syndrome and optic neuritis, to name a few. Patient case presentation findings were consistent with documented results in published literature. Conclusions: We present a case series with a thorough literature review documenting adverse neurological affects following COVID-19 vaccination. Our case presentations and literature review highlight the importance of neuroimaging when diagnosing post-COVID-19 vaccination adverse effects. MRI imaging study is an important tool to be considered in patients presenting with post-COVID-19 vaccination-related unexplained neurological symptoms for accurate diagnosis.
Collapse
Affiliation(s)
- Shitiz Sriwastava
- Department of Neurology, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA;
- School of Medicine, West Virginia University, Morgantown, WV 26506, USA; (M.A.C.); (D.N.)
- West Virginia Clinical and Translational Science Institute, Morgantown, WV 26506, USA
- Department of Neurology, Wayne State University, Detroit, MI 48201, USA;
- Correspondence:
| | | | - Syed Hassan Khalid
- Department of Neurology, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA;
| | - Mark A. Colantonio
- School of Medicine, West Virginia University, Morgantown, WV 26506, USA; (M.A.C.); (D.N.)
| | - Divine Nwafor
- School of Medicine, West Virginia University, Morgantown, WV 26506, USA; (M.A.C.); (D.N.)
| | | |
Collapse
|
1500
|
Tcheou J, Raskin A, Singh G, Kawabata H, Bielak D, Sun W, González-Domínguez I, Sather DN, García-Sastre A, Palese P, Krammer F, Carreño JM. Safety and Immunogenicity Analysis of a Newcastle Disease Virus (NDV-HXP-S) Expressing the Spike Protein of SARS-CoV-2 in Sprague Dawley Rats. Front Immunol 2021; 12:791764. [PMID: 34868082 PMCID: PMC8637447 DOI: 10.3389/fimmu.2021.791764] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 11/02/2021] [Indexed: 01/14/2023] Open
Abstract
Despite global vaccination efforts, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to evolve and spread globally. Relatively high vaccination rates have been achieved in most regions of the United States and several countries worldwide. However, access to vaccines in low- and mid-income countries (LMICs) is still suboptimal. Second generation vaccines that are universally affordable and induce systemic and mucosal immunity are needed. Here we performed an extended safety and immunogenicity analysis of a second-generation SARS-CoV-2 vaccine consisting of a live Newcastle disease virus vector expressing a pre-fusion stabilized version of the spike protein (NDV-HXP-S) administered intranasally (IN), intramuscularly (IM), or IN followed by IM in Sprague Dawley rats. Local reactogenicity, systemic toxicity, and post-mortem histopathology were assessed after the vaccine administration, with no indication of severe local or systemic reactions. Immunogenicity studies showed that the three vaccination regimens tested elicited high antibody titers against the wild type SARS-CoV-2 spike protein and the NDV vector. Moreover, high antibody titers were induced against the spike of B.1.1.7 (alpha), B.1.351 (beta) and B.1.617.2 (delta) variants of concern (VOCs). Importantly, robust levels of serum antibodies with neutralizing activity against the authentic SARS-CoV-2 USA-WA1/2020 isolate were detected after the boost. Overall, our study expands the pre-clinical safety and immunogenicity characterization of NDV-HXP-S and reinforces previous findings in other animal models about its high immunogenicity. Clinical testing of this vaccination approach is ongoing in different countries including Thailand, Vietnam, Brazil and Mexico.
Collapse
MESH Headings
- Administration, Intranasal
- Animals
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/immunology
- COVID-19/immunology
- COVID-19/prevention & control
- COVID-19 Vaccines/administration & dosage
- COVID-19 Vaccines/genetics
- COVID-19 Vaccines/immunology
- Immunogenicity, Vaccine
- Injections, Intramuscular
- Newcastle disease virus/genetics
- Newcastle disease virus/immunology
- Rats
- Rats, Sprague-Dawley
- SARS-CoV-2/genetics
- SARS-CoV-2/immunology
- Safety
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/immunology
- Vaccination
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- Johnstone Tcheou
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Ariel Raskin
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Gagandeep Singh
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Hisaaki Kawabata
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Dominika Bielak
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Weina Sun
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Irene González-Domínguez
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - D Noah Sather
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States
- Department of Pediatrics, University of Washington, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Peter Palese
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Juan Manuel Carreño
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|