1401
|
Jiang Y, Rubin L, Peng T, Liu L, Xing X, Lazarovici P, Zheng W. Cytokine storm in COVID-19: from viral infection to immune responses, diagnosis and therapy. Int J Biol Sci 2022; 18:459-472. [PMID: 35002503 PMCID: PMC8741849 DOI: 10.7150/ijbs.59272] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 08/05/2021] [Indexed: 12/15/2022] Open
Abstract
The COVID-19 outbreak is emerging as a significant public health challenge. Excessive production of proinflammatory cytokines, also known as cytokine storm, is a severe clinical syndrome known to develop as a complication of infectious or inflammatory diseases. Clinical evidence suggests that the occurrence of cytokine storm in severe acute respiratory syndrome secondary to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is closely associated with the rapid deterioration and high mortality of severe cases. In this review, we aim to summarize the mechanism of SARS-CoV-2 infection and the subsequent immunological events related to excessive cytokine production and inflammatory responses associated with ACE2-AngII signaling. An overview of the diagnosis and an update on current therapeutic regimens and vaccinations is also provided.
Collapse
Affiliation(s)
- Yizhou Jiang
- Center of Reproduction, Development & Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Limor Rubin
- Allergy and Clinical Immunology Unit, Department of Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Tangming Peng
- Center of Reproduction, Development & Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Linlin Liu
- Center of Reproduction, Development & Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Xingan Xing
- Center of Reproduction, Development & Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Philip Lazarovici
- School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Wenhua Zheng
- Center of Reproduction, Development & Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| |
Collapse
|
1402
|
Zheng C, Shao W, Chen X, Zhang B, Wang G, Zhang W. Real-world effectiveness of COVID-19 vaccines: a literature review and meta-analysis. Int J Infect Dis 2022; 114:252-260. [PMID: 34800687 PMCID: PMC8595975 DOI: 10.1016/j.ijid.2021.11.009] [Citation(s) in RCA: 399] [Impact Index Per Article: 133.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 10/21/2021] [Accepted: 11/05/2021] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE To estimate the coronavirus disease 2019 (COVID-19) vaccine effectiveness (VE) against concerned outcomes in real-world settings. METHODS Studies reporting COVID-19 VE from August 6, 2020 to October 6, 2021 were included. The summary VE (with 95% confidence intervals (95% CI)) against disease related to COVID-19 was estimated. The results were presented in forest plots. Predefined subgroup analyses and sensitivity analyses were also performed. RESULTS A total of 51 records were included in this meta-analysis. In fully vaccinated populations, the VE against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, COVID-19-related hospitalization, admission to the intensive care unit, and death was 89.1% (95% CI 85.6-92.6%), 97.2% (95% CI 96.1-98.3%), 97.4% (95% CI 96.0-98.8%), and 99.0% (95% CI 98.5-99.6%), respectively. The VE against infection in the general population aged ≥16 years, the elderly, and healthcare workers was 86.1% (95% CI 77.8-94.4%), 83.8% (95% CI 77.1-90.6%), and 95.3% (95% CI 92.0-98.6%), respectively. For those fully vaccinated against infection, the observed effectiveness of the Pfizer-BioNTech vaccine was 91.2% and of the Moderna vaccine was 98.1%, while the effectiveness of the CoronaVac vaccine was found to be 65.7%. CONCLUSIONS The COVID-19 vaccines are highly protective against SARS-CoV-2-related diseases in real-world settings.
Collapse
Affiliation(s)
| | | | | | | | | | - Weidong Zhang
- Corresponding author: Weidong Zhang, Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou, Henan, 450001, China. Tel: +86-0371-67781964
| |
Collapse
|
1403
|
Giri S, Sen S, Singh R, Paul P, Sahu R, Nandi G, Dua TK. Current challenges in different approaches to control COVID-19: a comprehensive review. BULLETIN OF THE NATIONAL RESEARCH CENTRE 2022; 46:47. [PMID: 35261539 PMCID: PMC8892405 DOI: 10.1186/s42269-022-00730-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 02/13/2022] [Indexed: 05/11/2023]
Abstract
BACKGROUND The World Health Organization declared the outbreak of the novel coronavirus (COVID-19) as a global health emergency on January 30, 2020, and as a pandemic disease on March 11, 2020. This review highlights the international situation, risk factors, and related protections to be taken as prerequisite measures and probable treatment options for the COVID-19-infected population in the current scenario. MAIN TEXT The SARS-CoV-2 viruses and their variants caused mild-to-severe respiratory tract infection and used airborne pathways as a way of contagion. Human-to-human transmission led to an exponential growth in the rise in the number of cases making it a real burden to immobilize the rapid spread of the virus while asymptomatic patients created ambiguity for confirmation in the community. It was clear from the case studies of patients that most of them were asymptomatic but still vulnerable to the people around, and hence, in a flash, many countries around the globe went into a complete lockdown, influencing the economy and thrashing industrial outputs. On the other hand, numerous researches were made to counteract the spread through studies in antiviral therapy, immune-based therapy, vaccination development, and natural remedies. CONCLUSION Although exploration for a specific drug required for the COVID-19 treatment is under extensive research worldwide and some of them are in clinical trial now. Virtual drug library screening is one of the current techniques for repurposing accessible compounds. This review could provide beneficial information about the potential current and future treatment strategies to treat the pandemic COVID-19 infection.
Collapse
Affiliation(s)
- Simran Giri
- Department of Pharmaceutical Technology, University of North Bengal, Raja Rammohunpur, P.O.- NBU, District- Darjeeling, West Bengal 734013 India
| | - Sanjukta Sen
- Department of Pharmaceutical Technology, University of North Bengal, Raja Rammohunpur, P.O.- NBU, District- Darjeeling, West Bengal 734013 India
| | - Rohan Singh
- Department of Pharmaceutical Technology, University of North Bengal, Raja Rammohunpur, P.O.- NBU, District- Darjeeling, West Bengal 734013 India
| | - Paramita Paul
- Department of Pharmaceutical Technology, University of North Bengal, Raja Rammohunpur, P.O.- NBU, District- Darjeeling, West Bengal 734013 India
| | - Ranabir Sahu
- Department of Pharmaceutical Technology, University of North Bengal, Raja Rammohunpur, P.O.- NBU, District- Darjeeling, West Bengal 734013 India
| | - Gouranga Nandi
- Department of Pharmaceutical Technology, University of North Bengal, Raja Rammohunpur, P.O.- NBU, District- Darjeeling, West Bengal 734013 India
| | - Tarun Kumar Dua
- Department of Pharmaceutical Technology, University of North Bengal, Raja Rammohunpur, P.O.- NBU, District- Darjeeling, West Bengal 734013 India
| |
Collapse
|
1404
|
Alsaffar WA, Alwesaibi AA, Alhaddad MJ, Alsenan ZK, Alsheef HJ, Alramadan SH, Aljassas HA, Alsaghirat MA, Alzahrani HJ. The Effectiveness of COVID-19 Vaccines in Improving the Outcomes of Hospitalized COVID-19 Patients. Cureus 2022; 14:e21485. [PMID: 35103227 PMCID: PMC8783154 DOI: 10.7759/cureus.21485] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2022] [Indexed: 12/28/2022] Open
Abstract
Background With the rapid spread of coronavirus disease 2019 (COVID-19), most countries took extreme measures to control the disease. Equitable access to safe and effective vaccines is critical to ending the COVID-19 pandemic. The Ministry of Health program in Saudi Arabia aimed to cover 17.4 million adults (70% of the adult population of Saudi Arabia) by the third quarter of 2021. We investigated the impact of the COVID-19 vaccine on the clinical course and outcomes of the admitted confirmed COVID-19 patients, in comparison to non-vaccinated patients. Methodology A retrospective cross-sectional record review was conducted for all hospitalized confirmed COVID-19 patients at Dammam Medical Complex (Eastern Province, Saudi Arabia) from June to July 2021. Two groups were studied according to the vaccination status (i.e., vaccinated and non-vaccinated). Information regarding comorbidities, length of stay, high oxygen requirements, ICU admission, and mortality data were collected and analyzed using the Python programming language (version 3.7.6) with the use of SciPy library (1.4.1) and Statsmodels module (v0.11.1). Results The sample included a total of 260 admitted confirmed COVID-19 cases. The mortality was significantly lower in the vaccinated group versus the non-vaccinated group with an odds ratio (OR) of 0.378 (CI 0.154-0.928). In addition, the OR of ICU admission was 0.476 (CI 0.218-1.042) and OR of endotracheal intubation was 0.561 (CI 0.249-1.265), but these did not reach statistical significance. We also detected a statistically significant relationship between mortality - regardless of vaccination status - and age ≥ 65 years (P=0.000, OR=7.51; 95%CI 3.13 to 18.04), chronic kidney disease (P=0.010, OR=5.62; CI 1.52 to 20.79), and renal transplant (P=0.037, OR=10.17; CI 1.15 to 89.76). Of note, most of our vaccinated patients received only a single dose (85%). Conclusion There is a significant reduction in mortality cases as well as less complicated hospital courses among the vaccinated group, in spite of the fact that most of our admitted patients had only a single vaccine shot. Suboptimal response to the vaccines was observed in the elder, chronic kidney disease, and renal transplant patients, hence the poorer outcomes in comparison to other patients.
Collapse
|
1405
|
Zhang HL, Li YX, Zhou AF, Li Y. New Frontier in Antiviral Drugs for Disorders of the Respiratory System. RECENT ADVANCES IN ANTI-INFECTIVE DRUG DISCOVERY 2022; 17:2-12. [PMID: 35430978 DOI: 10.2174/1574891x16666220416164740] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/25/2021] [Accepted: 01/26/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND COVID-19 is still soaring, and the new delta COVID-19 variant is on the rise and spreading around the world. OBJECTIVE We conducted a patent analysis to better understand the therapeutic strategy developed for antivirals available for the disorders of the respiratory system. MATERIALS AND METHODS European granted patents filed from January 2002 to June 2021 were analyzed. We used a combination of International patent classification (IPC) "A61p31/12" and "A61p11/00" to search the relevant documents. RESULTS Our study showed R&D of antiviral drugs for disorders of the respiratory system to be decreasing over the past 20 years. Chemical drugs showed various chemical structures. The development of chemical drugs or herbal medicines appeared to commence earlier than the biological products. Also, the results indicated that large global companies play a leading role in developing kinase inhibitors as chemical drugs. CONCLUSION There are three strategies for developing antiviral drugs for the disorders of the respiratory system, including chemical drugs, herbal medicines or natural products, and biological products. Herbal medicines may provide a new insight and approach to developing antiviral drugs for disorders of the respiratory system. A combination of chemical drugs and natural products may be a promising therapeutic method for treating patients with COVID- 19.
Collapse
Affiliation(s)
- Hai-Long Zhang
- Central International Intellectual Property (Baotou) Co., Ltd., Baotou, China
| | - Yong-Xia Li
- Central International Intellectual Property (Baotou) Co., Ltd., Baotou ,China
| | - Ai-Feng Zhou
- Central International Intellectual Property (Baotou) Co., Ltd., Baotou ,China
| | - Yiqian Li
- Department of Building Services Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| |
Collapse
|
1406
|
Kantarcioglu B, Iqbal O, Lewis J, Carter CA, Singh M, Lievano F, Ligocki M, Jeske W, Adiguzel C, Gerotziafas GT, Fareed J. An Update on the Status of Vaccine Development for SARS-CoV-2 Including Variants. Practical Considerations for COVID-19 Special Populations. Clin Appl Thromb Hemost 2022; 28:10760296211056648. [PMID: 35167393 PMCID: PMC8851053 DOI: 10.1177/10760296211056648] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/25/2021] [Accepted: 10/13/2021] [Indexed: 01/09/2023] Open
Abstract
The progress in the development of various vaccine platforms against SARS-CoV-2 have been rather remarkable owing to advancement in molecular and biologic sciences. Most of the current vaccines and those in development focus on targeting the viral spike proteins by generating antibodies of varying spectrum. These vaccines represent a variety of platforms including whole virus vaccines, viral vector vaccines, nucleic acid vaccines representing RNA, DNA, and their hybrid forms.The therapeutic efficacy of these vaccines varies owing to their pharmacodynamic individualities. COVID-19 variants are capable of inducing different pathologic responses and some of which may be resistant to antibodies generated by current vaccines. The current clinical use of these vaccines has been through emergency use authorization until recently. Moreover, the efficacy and safety of these vaccines have been tested in substantial numbers of individuals but studies in special populations that better reflect the global population are pending results. These specialized populations include young children, immunocompromised patients, pregnant individuals, and other specialized groups. Combination approaches, molecularly modified vaccination approaches, and vaccines conferring longer periods of immunity are being currently being investigated, as well as pharmacovigilance studies.The continual transformation of SARS-CoV-2 and its variants are of concern along with the breakthrough infections. These considerations pose new challenges for the development of vaccination platforms. For this purpose, booster doses, combination vaccine approaches, and other modalities are being discussed. This review provides an updated account of currently available vaccines and those in advanced development with reference to their composition and mechanisms of action.A discussion on the use of vaccines in special populations including immunocompromised patients, pregnant women and other specialized populations are also included.
Collapse
Affiliation(s)
- Bulent Kantarcioglu
- Cardiovascular Research Institute, Loyola University Chicago, Health Sciences Division, Maywood, IL, USA
| | - Omer Iqbal
- Cardiovascular Research Institute, Loyola University Chicago, Health Sciences Division, Maywood, IL, USA
| | - Joseph Lewis
- Cardiovascular Research Institute, Loyola University Chicago, Health Sciences Division, Maywood, IL, USA
| | - Charles A. Carter
- Campbell University College of Pharmacy and Health Sciences, Campbell University, Buies Creek, NC, USA
| | - Meharvan Singh
- Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
| | | | | | - Walter Jeske
- Cardiovascular Research Institute, Loyola University Chicago, Health Sciences Division, Maywood, IL, USA
| | | | - Grigoris T. Gerotziafas
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Thrombosis Center, Service D’Hématologie Biologique Hôpital Tenon, Paris, France
| | - Jawed Fareed
- Cardiovascular Research Institute, Loyola University Chicago, Health Sciences Division, Maywood, IL, USA
| |
Collapse
|
1407
|
Dahal A, Sonju JJ, Kousoulas KG, Jois SD. Peptides and peptidomimetics as therapeutic agents for Covid-19. Pept Sci (Hoboken) 2022; 114:e24245. [PMID: 34901700 PMCID: PMC8646791 DOI: 10.1002/pep2.24245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/24/2021] [Accepted: 08/26/2021] [Indexed: 12/27/2022]
Abstract
The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) Covid-19 pandemic has caused high morbidity and mortality rates worldwide. Virus entry into cells can be blocked using several strategies, including inhibition of protein-protein interactions (PPIs) between the viral spike glycoprotein and cellular receptors, as well as blocking of spike protein conformational changes that are required for cleavage/activation and fusogenicity. The spike-mediated viral attachment and entry into cells via fusion of the viral envelope with cellular membranes involve PPIs mediated by short peptide fragments exhibiting particular secondary structures. Thus, peptides that can inhibit these PPIs may be used as potential antiviral agents preventing virus entry and spread. This review is focused on peptides and peptidomimetics as PPI modulators and protease inhibitors against SARS-CoV-2.
Collapse
Affiliation(s)
- Achyut Dahal
- School of Basic Pharmaceutical and Toxicological Sciences, College of PharmacyUniversity of Louisiana at MonroeMonroeLouisianaUSA
| | - Jafrin Jobayer Sonju
- School of Basic Pharmaceutical and Toxicological Sciences, College of PharmacyUniversity of Louisiana at MonroeMonroeLouisianaUSA
| | - Konstantin G. Kousoulas
- Department of Pathobiological Sciences, School of Veterinary MedicineLouisiana State UniversityBaton RougeLouisianaUSA
| | - Seetharama D. Jois
- School of Basic Pharmaceutical and Toxicological Sciences, College of PharmacyUniversity of Louisiana at MonroeMonroeLouisianaUSA
| |
Collapse
|
1408
|
Ranjbar S, Fatahi Y, Atyabi F. The quest for a better fight: How can nanomaterials address the current therapeutic and diagnostic obstacles in the fight against COVID-19? J Drug Deliv Sci Technol 2022; 67:102899. [PMID: 34630635 PMCID: PMC8489264 DOI: 10.1016/j.jddst.2021.102899] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 09/19/2021] [Accepted: 10/01/2021] [Indexed: 01/18/2023]
Abstract
The inexorable coronavirus disease 2019 (COVID-19) pandemic with around 226 million people diagnosed and approximately 4.6 million deaths, is still moving toward more frightening statistics, calling for the urgent need to explore solutions for the current challenges in therapeutic and diagnostic approaches. The challenges associated with existing therapeutics in COVID-19 include lack of in vivo stability, efficacy, and safety. Nanoparticles (NPs) can offer a handful of tools to tackle these problems by enabling efficacious and safe delivery of both virus- and host-directed therapeutics. Furthermore, they can enable maximized clinical outcome while eliminating the chance of resistance to therapy by tissue-targeting and concomitant delivery of multiple therapeutics. The promising application of NPs as vaccine platforms is reflected by the major advances in developing novel COVID-19 vaccines. Two of the most critical COVID-19 vaccines are mRNA-based vaccines delivered via NPs, making them the first FDA-approved mRNA vaccines. Besides, NPs have been deployed as simple, rapid, and precise tools for point of care disease diagnosis. Not enough said NPs can also be exploited in novel ways to expedite the drug discovery process. In light of the above, this review discusses how NPs can overcome the hurdles associated with therapeutic and diagnostic approaches against COVID-19.
Collapse
Affiliation(s)
- Sheyda Ranjbar
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, 1417614411, Iran,Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, 1417614411, Iran
| | - Yousef Fatahi
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, 1417614411, Iran
| | - Fatemeh Atyabi
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, 1417614411, Iran,Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, 1417614411, Iran,Corresponding author. Faculty of Pharmacy, Tehran University of Medical Sciences Tehran, PO Box 14155-6451, 1417614411, Iran
| |
Collapse
|
1409
|
Singh DD, Parveen A, Yadav DK. SARS-CoV-2: Emergence of New Variants and Effectiveness of Vaccines. Front Cell Infect Microbiol 2022; 11:777212. [PMID: 34970509 PMCID: PMC8713083 DOI: 10.3389/fcimb.2021.777212] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/09/2021] [Indexed: 01/14/2023] Open
Abstract
The emergence of SARS-CoV-2 variants may cause resistance at the immunity level against current vaccines. Some emergent new variants have increased transmissibility, infectivity, hospitalization, and mortality. Since the administration of the first SARS-CoV-2 vaccine to a human in March 2020, there is an ongoing global race against SARS-CoV-2 to control the current pandemic situation. Spike (S) glycoprotein of SARS-CoV-2 is the main target for current vaccine development, which can neutralize the infection. Companies and academic institutions have developed vaccines based on the S glycoprotein, as well as its antigenic domains and epitopes, which have been proven effective in generating neutralizing antibodies. The effectiveness of SARS-CoV-2 vaccines and other therapeutics developments are limited by the new emergent variants at the global level. We have discussed the emergent variants of SARS-CoV-2 on the efficacy of developed vaccines. Presently, most of the vaccines have been tremendously effective in severe diseases. However, there are still noteworthy challenges in certifying impartial vaccines; the stories of re-infections are generating more stressful conditions, and this needs further clinical evaluation.
Collapse
Affiliation(s)
- Desh Deepak Singh
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, India
| | - Amna Parveen
- Gachon Institute of Pharmaceutical Science and Department of Pharmacy, College of Pharmacy, Gachon University, Incheon, South Korea
| | - Dharmendra Kumar Yadav
- Gachon Institute of Pharmaceutical Science and Department of Pharmacy, College of Pharmacy, Gachon University, Incheon, South Korea
| |
Collapse
|
1410
|
Zhao Z, Anselmo AC, Mitragotri S. Viral vector-based gene therapies in the clinic. Bioeng Transl Med 2022; 7:e10258. [PMID: 35079633 PMCID: PMC8780015 DOI: 10.1002/btm2.10258] [Citation(s) in RCA: 136] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 08/04/2021] [Accepted: 08/11/2021] [Indexed: 02/06/2023] Open
Abstract
Gene therapies are currently one of the most investigated therapeutic modalities in both the preclinical and clinical settings and have shown promise in treating a diverse spectrum of diseases. Gene therapies aim at introducing a gene material in target cells and represent a promising approach to cure diseases that were thought to be incurable by conventional modalities. In many cases, a gene therapy requires a vector to deliver gene therapeutics into target cells; viral vectors are among the most widely studied vectors owing to their distinguished advantages such as outstanding transduction efficiency. With decades of development, viral vector-based gene therapies have achieved promising clinical outcomes with many products approved for treating a range of diseases including cancer, infectious diseases and monogenic diseases. In addition, a number of active clinical trials are underway to further expand their therapeutic potential. In this review, we highlight the diversity of viral vectors, review approved products, and discuss the current clinical landscape of in vivo viral vector-based gene therapies. We have reviewed 13 approved products and their clinical applications. We have also analyzed more than 200 active trials based on various viral vectors and discussed their respective therapeutic applications. Moreover, we provide a critical analysis of the major translational challenges for in vivo viral vector-based gene therapies and discuss possible strategies to address the same.
Collapse
Affiliation(s)
- Zongmin Zhao
- Department of Pharmaceutical Sciences, College of PharmacyUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Aaron C. Anselmo
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of PharmacyUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMassachusettsUSA
| |
Collapse
|
1411
|
Ciotti M, Ciccozzi M, Pieri M, Bernardini S. The COVID-19 pandemic: viral variants and vaccine efficacy. Crit Rev Clin Lab Sci 2022; 59:66-75. [PMID: 34598660 DOI: 10.1080/10408363.2021.1979462] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/15/2021] [Accepted: 09/08/2021] [Indexed: 12/22/2022]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has prompted the scientific community and the pharmaceutical companies to put maximum efforts into developing vaccines to contain the spread of this disease. Presently, many vaccines have been developed and authorized for use in human beings in different countries. In particular, in Europe to date, the Pfizer-BioNTech, Moderna, AstraZeneca and Janssen COVID-19 vaccines have been authorized. All of them are based on a version of the spike (S) glycoprotein characterized at the beginning of the pandemic. However, they differ by their level of efficacy against COVID-19. SARS-COV-2, like other RNA viruses, mutates continually. Genome sequencing analysis shows a nucleotide substitution rate of about 1 × 10-3 substitutions per year that leads to the emergence of variants through point mutations, insertions, deletions and recombination. There is concern about the ability of the current vaccines to protect against emerging viral variants. Mutations in the S-glycoprotein may affect transmission dynamics and the risk of immune escape. In this review, we address the different technological platforms in use for developing COVID-19 vaccines, the impact of emerging viral variants on virus transmission, hospitalization, and response to current vaccines, as well as rare but important adverse reactions to them. Finally, different methods for measuring antibody response to the vaccines, including the importance of using the WHO International Standard to calibrate immunoassays accurately to an arbitrary unit, to reduce interlaboratory variation and to create a common language for reporting results, are reported.
Collapse
Affiliation(s)
- Marco Ciotti
- Virology Unit, Polyclinic Tor Vergata Foundation, Rome, Italy
| | - Massimo Ciccozzi
- Unit of Medical Statistics and Molecular Epidemiology, Campus Bio-Medico of Rome, Rome, Italy
| | - Massimo Pieri
- Department of Experimental Medicine, University of Tor Vergata, Rome, Italy
- Department of Laboratory Medicine, Polyclinic Tor Vergata Foundation, Viale Oxford, Rome, Italy
| | - Sergio Bernardini
- Department of Experimental Medicine, University of Tor Vergata, Rome, Italy
- Emerging Technologies Division (ETD) of the International Federation Clinical Chemistry and Laboratory Medicine (IFCC), Milan, Italy
| |
Collapse
|
1412
|
Garces KN, Cocores AN, Goadsby PJ, Monteith TS. Headache After Vaccination: An Update on Recent Clinical Trials and Real-World Reporting. Curr Pain Headache Rep 2022; 26:895-918. [PMID: 36418848 PMCID: PMC9685066 DOI: 10.1007/s11916-022-01094-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2022] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW The aim of this review is to characterize headache as a vaccine adverse event (VAE) in clinical trials. RECENT FINDINGS Of the recent phase III vaccine RCTs (non-COVID-19), 53 studies reported on headache (13 infectious agents). The median rate (interquartile range) of headache was 15.6% (IQR: 9.6-37.6%). Of these, 24.5% of the RCTs reported headache greater in the vaccine group compared to the placebo/control group. In the herpes zoster vaccination trials, headache was more common in all active groups: median rate 33.9% (IQR: 29.7-40.5%) as compared to placebo: median rate 17.7% (IQR: 15.4-23.8%). Influenza and HPV vaccination trials were the 2nd and 3rd most common to have headache as a VAE. Of the 6 widely distributed COVID-19 vaccinations, median rate of post-vaccination headache was 39% (IQR: 28-50%). Headache is a common VAE in vaccine trials. Standardized grading methods, predictors of persistence, and treatment regimens are warranted.
Collapse
Affiliation(s)
- Kimberly N. Garces
- Division of Headache, Department of Neurology, University of Miami, Miller School of Medicine, Miami, FL USA
| | - Alexandra N. Cocores
- Division of Headache, Department of Neurology, University of Miami, Miller School of Medicine, Miami, FL USA
| | - Peter J. Goadsby
- NIHR King’s Clinical Research Facility & Headache Group-Wolfson CARD, King’s College London, London, UK ,Department of Neurology, University of California, Los Angeles, Los Angeles, CA USA
| | - Teshamae S. Monteith
- Division of Headache, Department of Neurology, University of Miami, Miller School of Medicine, Miami, FL USA
| |
Collapse
|
1413
|
Lipsitch M, Krammer F, Regev-Yochay G, Lustig Y, Balicer RD. SARS-CoV-2 breakthrough infections in vaccinated individuals: measurement, causes and impact. Nat Rev Immunol 2022; 22:57-65. [PMID: 34876702 PMCID: PMC8649989 DOI: 10.1038/s41577-021-00662-4] [Citation(s) in RCA: 222] [Impact Index Per Article: 74.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/16/2021] [Indexed: 02/04/2023]
Abstract
Breakthrough infections with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in fully vaccinated individuals are receiving intense scrutiny because of their importance in determining how long restrictions to control virus transmission will need to remain in place in highly vaccinated populations as well as in determining the need for additional vaccine doses or changes to the vaccine formulations and/or dosing intervals. Measurement of breakthrough infections is challenging outside of randomized, placebo-controlled, double-blind field trials. However, laboratory and observational studies are necessary to understand the impact of waning immunity, viral variants and other determinants of changing vaccine effectiveness against various levels of coronavirus disease 2019 (COVID-19) severity. Here, we describe the approaches being used to measure vaccine effectiveness and provide a synthesis of the burgeoning literature on the determinants of vaccine effectiveness and breakthrough rates. We argue that, rather than trying to tease apart the contributions of factors such as age, viral variants and time since vaccination, the rates of breakthrough infection are best seen as a consequence of the level of immunity at any moment in an individual, the variant to which that individual is exposed and the severity of disease being considered. We also address key open questions concerning the transition to endemicity, the potential need for altered vaccine formulations to track viral variants, the need to identify immune correlates of protection, and the public health challenges of using various tools to counter breakthrough infections, including boosters in an era of global vaccine shortages.
Collapse
Affiliation(s)
- Marc Lipsitch
- Center for Communicable Disease Dynamics, Department of Epidemiology and Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gili Regev-Yochay
- Infection Prevention & Control Unit, Sheba Medical Center, Ramat-Gan, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yaniv Lustig
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- The Central Virology Laboratory, Public Health Services, Ministry of Health, Sheba Medical Center, Tel-Hashomer, Israel
| | - Ran D Balicer
- Clalit Research Institute, Innovation Division, Clalit Health Services, Tel Aviv, Israel
- The School of Public Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| |
Collapse
|
1414
|
Singh A, Khillan R, Mishra Y, Khurana S. The safety profile of COVID-19 vaccinations in the United States. Am J Infect Control 2022; 50:15-19. [PMID: 34699960 PMCID: PMC8539830 DOI: 10.1016/j.ajic.2021.10.015] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/13/2021] [Accepted: 10/16/2021] [Indexed: 12/27/2022]
Abstract
Background/Aim Pfizer-BioNTech, Moderna, and Johnson & Johnson's Janssen are the 3 COVID-19 vaccines authorized for emergency use in the United States. This study aims to analyze and compare adverse events following immunization associated with these COVID-19 vaccines based on Vaccine Adverse Effect Reporting System data. Methods We utilized Vaccine Adverse Effect Reporting System data from January 1, 2021 to April 30, 2021 to analyze and characterize adverse effects postvaccination with these authorized COVID-19 vaccines in the US population. Results A total of 141,208 individuals suffered at least one adverse events following immunization following 239.97 million doses of COVID-19 vaccination. The frequency of side effects was 0.04%, 0.06%, and 0.35% following administration of Pfizer-BioNTech, Moderna, and Johnson & Johnson's Janssen vaccines, respectively. Most of the patients had mild systemic side effects, the most common being headache (0.01%) and fever (0.01%). The frequency of serious side effects including anaphylaxis (0.0003%) and death (0.002%) was extremely low. Conclusions The three COVID 19 vaccines have a wide safety profile with only minor and self-limiting adverse effects. However, continued monitoring and surveillance is required to review any unexpected serious adverse effects.
Collapse
|
1415
|
Syahniar R, Kharisma DS. SARS-CoV-2 vaccine challenge based on spike glycoprotein against several new variants. Clin Exp Vaccine Res 2022; 11:173-183. [PMID: 35799876 PMCID: PMC9200648 DOI: 10.7774/cevr.2022.11.2.173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 04/30/2022] [Indexed: 11/15/2022] Open
Abstract
The coronavirus disease 2019 pandemic has not ended, and several variants of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus continue to emerge. The emergence of new variants is worrying because higher transmission leads to spikes in infections, vaccine efforts, and other therapeutic developments. Existing literature reports that with new variants affecting vaccine efficacy, hospitalization and risk of a recurrent infection increase. In this review article, we describe the latest variants of SARS-CoV-2, and the impact of each new variant on the efficacy of the developed vaccines reported in the literature and findings. The report concludes that the emergence of a variant that completely evades the immune response and reduces neutralizing antibodies.
Collapse
Affiliation(s)
- Rike Syahniar
- Department of Microbiology, Faculty of Medicine and Health, Universitas Muhammadiyah Jakarta, South Jakarta, Indonesia
| | - Dayu Swasti Kharisma
- Department of Microbiology, Faculty of Medicine and Health, Universitas Muhammadiyah Jakarta, South Jakarta, Indonesia
| |
Collapse
|
1416
|
Abstract
INTRODUCTION Appearances of SARS-CoV-2 variants have created havoc and additional challenges for the ongoing vaccination drive against pandemic COVID-19. Interestingly, several vaccine platforms are showing great potential to produce successful vaccines against SARS-CoV-2 and its variants. Billions of COVID-19 vaccine doses have been administered worldwide. Mix-and-Match COVID-19 vaccines involving the mixing of the same platform vaccines and also two different vaccine platforms may provide greater protection against SARS-CoV-2 and its variants. COVID-19 vaccines have become one of the most important tools to mitigate the ongoing pandemic COVID-19. AREAS COVERED We describe SARS-Cov-2 variants, their impact on the population, COVID-19 vaccines, diverse vaccine platforms, doses of vaccines, the efficacy of vaccines against SARS-CoV-2 and its variants, mitigation of the COVID-19 transmission- alternatives to vaccines. EXPERT OPINION Diverse vaccine platforms may safeguard against ongoing, deadly SARS-CoV-2 and its infectious variants. The efficacies of COVID-19 vaccines are significantly high after the administration of the second dose. Further, it protects individuals including vulnerable patients with co-morbidities from SARS-CoV-2 and its variants. The hospitalizations and deaths of the individuals may be prevented by COVID-19 vaccines.
Collapse
Affiliation(s)
- Bhaswati Chatterjee
- Chemical Science, National Institute of Pharmaceutical Education and Research, Hyderabad, India,CONTACT Bhaswati Chatterjee Chemical Science National Institute of Pharmaceutical Education and Research, India
| | - Suman S. Thakur
- Proteomics and Cell Signaling, Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad, India,Suman S. Thakur Principal Scientist, Proteomics and Cell Signaling, Lab W110 Centre for Cellular and Molecular Biology Uppal Road, Hyderabad-500007, India
| |
Collapse
|
1417
|
Aravind S, Mathew KA, Madathil BK, Mini S, John A. Current strategies and future perspectives in COVID-19 therapy. STEM CELLS AND COVID-19 2022:169-227. [DOI: 10.1016/b978-0-323-89972-7.00011-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
1418
|
Abdulla M. Cutaneous leukocytoclastic vasculitis after ChAdOx1 nCoV-19 vaccine. INDIAN JOURNAL OF MEDICAL SPECIALITIES 2022. [DOI: 10.4103/injms.injms_87_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
1419
|
Heesen G, Heinemann S, Müller F, Dopfer-Jablonka A, Mikuteit M, Niewolik J, Klawonn F, Vahldiek K, Hummers E, Schröder D. Social participation and mental health of immunocompromised individuals before and after COVID-19 vaccination-Results of a longitudinal observational study over three time points. Front Psychiatry 2022; 13:1080106. [PMID: 36590634 PMCID: PMC9795223 DOI: 10.3389/fpsyt.2022.1080106] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 11/29/2022] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION The coronavirus disease 2019 (COVID-19) pandemic impacted how people perform their daily lives in manifold and sometimes massive ways. Particularly, individuals who are at high risk for a severe disease progression, like immunocompromised people, may have experienced drastic changes in social participation during the pandemic. A COVID-19 basic vaccination may have changed the safety behavior of immunocompromised individuals in terms of infection risk and thereby influence social participation and mental wellbeing. METHODS This study aims to investigate self-perceived social participation at baseline before and at follow-up 1 and 6 months after basic vaccination. Beginning in March 2021, 274 immunocompromised persons 18 years or older were enrolled in the COVID-19 Contact Immune study (CoCo study) in Lower Saxony, Germany. Measurements were performed at three time points regarding social participation [Index for the Assessment of Health Impairments (IMET)], mental health [Patient Health Questionnaire-4 (PHQ-4)], subjective health status (five-point Likert-scale) and quality of life (five-point Likert-scale). RESULTS In total, 126 participants were included in the final analysis. About 60% of the participants showed increasing social participation over time. The greatest increase in social participation was observed within the first month after basic vaccination (p < 0.001). During the following 5 months, social participation remained stable. The domains "social activities," "recreation and leisure" and "close personal relationships" were responsible for the overall change in social participation. No association was found between social participation and mental health, sociodemographic or medical factors (except hypertension). DISCUSSION It is unclear why social participation increased after basic vaccination. Perceived vaccine efficacy and a feeling of being protected by the vaccine may have caused relaxed social distancing behaviors. Reducing safety behaviors may, however, increase the risk of a COVID-19 infection for immunocompromised individuals. Further investigations are needed to explore the health-related consequences of more social participation among immunocompromised persons.
Collapse
Affiliation(s)
- Gloria Heesen
- Department of General Practice, University Medical Center, Göttingen, Germany
| | - Stephanie Heinemann
- Department of General Practice, University Medical Center, Göttingen, Germany
| | - Frank Müller
- Department of General Practice, University Medical Center, Göttingen, Germany
| | - Alexandra Dopfer-Jablonka
- Department of Rheumatology and Immunology, Hannover Medical School, Hanover, Germany.,German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Brunswick, Germany
| | - Marie Mikuteit
- Department of Rheumatology and Immunology, Hannover Medical School, Hanover, Germany
| | - Jacqueline Niewolik
- Department of Rheumatology and Immunology, Hannover Medical School, Hanover, Germany
| | - Frank Klawonn
- Department of Computer Science, Ostfalia University of Applied Sciences, Wolfenbüttel, Germany.,Biostatistics Group, Helmholtz Centre for Infection Research, Brunswick, Germany
| | - Kai Vahldiek
- Department of Computer Science, Ostfalia University of Applied Sciences, Wolfenbüttel, Germany
| | - Eva Hummers
- Department of General Practice, University Medical Center, Göttingen, Germany
| | - Dominik Schröder
- Department of General Practice, University Medical Center, Göttingen, Germany
| |
Collapse
|
1420
|
Abstract
This article is a narrative review of the rapidly moving coronavirus disease 2019 vaccine field with an emphasis on clinical efficacy established in both randomized trials and postmarketing surveillance of clinically available vaccines. We review the major clinical trials that supported authorization for general use of the Janssen (Ad.26.CoV2), Pfizer-BioNTech (BNT162b2), and Moderna (mRNA-1273) vaccines and the publicly available postmarketing information with the goal of providing a broad, clinically relevant comparison of efficacy and safety. This review is primarily focused on the US market.
Collapse
Affiliation(s)
- William O Hahn
- Division of Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA, USA.
| | - Zanthia Wiley
- Division of Infectious Diseases, Department of Medicine, Emory University, Infectious Diseases Clinic, 550 Peachtree Street NE, 7th Floor - Medical Office Tower, Atlanta, GA 30308, USA
| |
Collapse
|
1421
|
Balakrishna K, Randhi U, Tammina B, Jangal S, Chandu K. A layman approach to adopt COVID-19 appropriate behaviour and vaccination: A narrative review. ASIAN JOURNAL OF PHARMACEUTICAL RESEARCH AND HEALTH CARE 2022. [DOI: 10.4103/ajprhc.ajprhc_12_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
1422
|
Naranbhai V, Pernat CA, Gavralidis A, St Denis KJ, Lam EC, Spring LM, Isakoff SJ, Farmer JR, Zubiri L, Hobbs GS, How J, Brunner AM, Fathi AT, Peterson JL, Sakhi M, Hambelton G, Denault EN, Mortensen LJ, Perriello LA, Bruno MN, Bertaux BY, Lawless AR, Jackson MA, Niehoff E, Barabell C, Nambu CN, Nakajima E, Reinicke T, Bowes C, Berrios-Mairena CJ, Ofoman O, Kirkpatrick GE, Thierauf JC, Reynolds K, Willers H, Beltran WG, Dighe AS, Saff R, Blumenthal K, Sullivan RJ, Chen YB, Kim A, Bardia A, Balazs AB, Iafrate AJ, Gainor JF. Immunogenicity and Reactogenicity of SARS-CoV-2 Vaccines in Patients With Cancer: The CANVAX Cohort Study. J Clin Oncol 2022; 40:12-23. [PMID: 34752147 PMCID: PMC8683230 DOI: 10.1200/jco.21.01891] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/24/2021] [Accepted: 10/13/2021] [Indexed: 01/03/2023] Open
Abstract
PURPOSE The immunogenicity and reactogenicity of SARS-CoV-2 vaccines in patients with cancer are poorly understood. METHODS We performed a prospective cohort study of adults with solid-organ or hematologic cancers to evaluate anti-SARS-CoV-2 immunoglobulin A/M/G spike antibodies, neutralization, and reactogenicity ≥ 7 days following two doses of mRNA-1273, BNT162b2, or one dose of Ad26.COV2.S. We analyzed responses by multivariate regression and included data from 1,638 healthy controls, previously reported, for comparison. RESULTS Between April and July 2021, we enrolled 1,001 patients; 762 were eligible for analysis (656 had neutralization measured). mRNA-1273 was the most immunogenic (log10 geometric mean concentration [GMC] 2.9, log10 geometric mean neutralization titer [GMT] 2.3), followed by BNT162b2 (GMC 2.4; GMT 1.9) and Ad26.COV2.S (GMC 1.5; GMT 1.4; P < .001). The proportion of low neutralization (< 20% of convalescent titers) among Ad26.COV2.S recipients was 69.9%. Prior COVID-19 infection (in 7.1% of the cohort) was associated with higher responses (P < .001). Antibody titers and neutralization were quantitatively lower in patients with cancer than in comparable healthy controls, regardless of vaccine type (P < .001). Receipt of chemotherapy in the prior year or current steroids were associated with lower antibody levels and immune checkpoint blockade with higher neutralization. Systemic reactogenicity varied by vaccine and correlated with immune responses (P = .002 for concentration, P = .016 for neutralization). In 32 patients who received an additional vaccine dose, side effects were similar to prior doses, and 30 of 32 demonstrated increased antibody titers (GMC 1.05 before additional dose, 3.17 after dose). CONCLUSION Immune responses to SARS-CoV-2 vaccines are modestly impaired in patients with cancer. These data suggest utility of antibody testing to identify patients for whom additional vaccine doses may be effective and appropriate, although larger prospective studies are needed.
Collapse
Affiliation(s)
- Vivek Naranbhai
- Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
- Dana-Farber Cancer Institute, Boston, MA
- Center for the AIDS Programme of Research in South Africa, Durban, South Africa
| | - Claire A. Pernat
- Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Alexander Gavralidis
- Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
- Salem Hospital, Salem, MA
| | | | - Evan C. Lam
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA
| | - Laura M. Spring
- Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Steven J. Isakoff
- Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Jocelyn R. Farmer
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Leyre Zubiri
- Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Gabriela S. Hobbs
- Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Joan How
- Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
- Division of Hematology, Brigham and Women's Hospital, Boston, MA
| | - Andrew M. Brunner
- Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Amir T. Fathi
- Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Jennifer L. Peterson
- Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Mustafa Sakhi
- Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Grace Hambelton
- Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Elyssa N. Denault
- Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Lindsey J. Mortensen
- Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Lailoo A. Perriello
- Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Marissa N. Bruno
- Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Brittany Y. Bertaux
- Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Aleigha R. Lawless
- Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Monica A. Jackson
- Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Elizabeth Niehoff
- Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Caroline Barabell
- Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Christian N. Nambu
- Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Erika Nakajima
- Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Trenton Reinicke
- Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Cynthia Bowes
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA
| | | | - Onosereme Ofoman
- Department of Pathology, Massachusetts General Hospital, Boston, MA
| | | | | | - Kerry Reynolds
- Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Henning Willers
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA
| | - Wilfredo-Garcia Beltran
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA
- Department of Pathology, Massachusetts General Hospital, Boston, MA
| | - Anand S. Dighe
- Department of Pathology, Massachusetts General Hospital, Boston, MA
| | - Rebecca Saff
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Kimberly Blumenthal
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Ryan J. Sullivan
- Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Yi-Bin Chen
- Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Arthur Kim
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Aditya Bardia
- Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | | | - A. John Iafrate
- Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
- Department of Pathology, Massachusetts General Hospital, Boston, MA
| | - Justin F. Gainor
- Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
1423
|
Faour WH, Choaib A, Issa E, Choueiry FE, Shbaklo K, Alhajj M, Sawaya RT, Harhous Z, Alefishat E, Nader M. Mechanisms of COVID-19-induced kidney injury and current pharmacotherapies. Inflamm Res 2022; 71:39-56. [PMID: 34802072 PMCID: PMC8606168 DOI: 10.1007/s00011-021-01520-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 10/31/2021] [Accepted: 11/01/2021] [Indexed: 01/08/2023] Open
Abstract
The COVID-19 pandemic created a worldwide debilitating health crisis with the entire humanity suffering from the deleterious effects associated with the high infectivity and mortality rates. While significant evidence is currently available online and targets various aspects of the disease, both inflammatory and noninflammatory kidney manifestations secondary to COVID-19 infection are still largely underrepresented. In this review, we summarized current knowledge about COVID-19-related kidney manifestations, their pathologic mechanisms as well as various pharmacotherapies used to treat patients with COVID-19. We also shed light on the effect of these medications on kidney functions that can further enhance renal damage secondary to the illness.
Collapse
Affiliation(s)
- Wissam H Faour
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos, Lebanon.
| | - Ali Choaib
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos, Lebanon
| | - Elio Issa
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos, Lebanon
| | - Francesca El Choueiry
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos, Lebanon
| | - Khodor Shbaklo
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos, Lebanon
| | - Maryline Alhajj
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos, Lebanon
| | - Ramy Touma Sawaya
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos, Lebanon
| | - Zeina Harhous
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos, Lebanon
| | - Eman Alefishat
- Department of Pharmacology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, P.O. Box 127788, Abu Dhabi, United Arab Emirates
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Department of Biopharmaceutics and Clinical Pharmacy, School of Pharmacy, The University of Jordan, Amman, Jordan
| | - Moni Nader
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, P.O. Box 127788, Abu Dhabi, United Arab Emirates.
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
1424
|
Testino G, Vignoli T, Patussi V, Allosio P, Amendola MF, Aricò S, Baselice A, Balbinot P, Campanile V, Fanucchi T, Macciò L, Meneguzzi C, Mioni D, Parisi M, Renzetti D, Rossin R, Gandin C, Bottaro LC, Caio G, Lungaro L, Zoli G, Scafato E, Caputo F. Alcohol use disorder in the COVID-19 era: Position paper of the Italian Society on Alcohol (SIA). Addict Biol 2022; 27:e13090. [PMID: 34532923 PMCID: PMC8646667 DOI: 10.1111/adb.13090] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 12/11/2022]
Abstract
Coronavirus disease 2019 (COVID-19) first emerged in China in November 2019. Most governments have responded to the COVID-19 pandemic by imposing a lockdown. Some evidence suggests that a period of isolation might have led to a spike in alcohol misuse, and in the case of patients with alcohol use disorder (AUD), social isolation can favour lapse and relapse. The aim of our position paper is to provide specialists in the alcohol addiction field, in psychopharmacology, gastroenterology and in internal medicine, with appropriate tools to better manage patients with AUD and COVID-19,considering some important topics: (a) the susceptibility of AUD patients to infection; (b) the pharmacological interaction between medications used to treat AUD and to treat COVID-19; (c) the reorganization of the Centre for Alcohol Addiction Treatment for the management of AUD patients in the COVID-19 era (group activities, telemedicine, outpatients treatment, alcohol-related liver disease and liver transplantation, collecting samples); (d) AUD and SARS-CoV-2 vaccination. Telemedicine/telehealth will undoubtedly be useful/practical tools even though it remains at an elementary level; the contribution of the family and of caregivers in the management of AUD patients will play a significant role; the multidisciplinary intervention involving experts in the treatment of AUD with specialists in the treatment of COVID-19 disease will need implementation. Thus, the COVID-19 pandemic is rapidly leading addiction specialists towards a new governance scenario of AUD, which necessarily needs an in-depth reconsideration, focusing attention on a safe approach in combination with the efficacy of treatment.
Collapse
Affiliation(s)
- Gianni Testino
- Unit of Addiction and Hepatology, Regional Centre on AlcoholASL3 San Martino HospitalGenoaItaly
| | - Teo Vignoli
- Unit of Addiction TreatmentLugo (Ravenna)Italy
| | | | | | | | - Sarino Aricò
- Gastroenterology UnitMauriziano HospitalTorinoItaly
| | | | - Patrizia Balbinot
- Unit of Addiction and Hepatology, Regional Centre on AlcoholASL3 San Martino HospitalGenoaItaly
| | | | | | | | | | | | | | - Doda Renzetti
- Department of Internal MedicineMater Dei HospitalBariItaly
| | | | - Claudia Gandin
- National Observatory on AlcoholNational Institute of HealthRomeItaly
| | | | - Giacomo Caio
- Centre for the Study and Treatment of Alcohol‐Related DiseasesDepartment of Translational Medicine, University of FerraraFerraraItaly
| | - Lisa Lungaro
- Centre for the Study and Treatment of Alcohol‐Related DiseasesDepartment of Translational Medicine, University of FerraraFerraraItaly
| | - Giorgio Zoli
- Centre for the Study and Treatment of Alcohol‐Related DiseasesDepartment of Translational Medicine, University of FerraraFerraraItaly
- Department of Internal Medicine, SS Annunziata Hospital, Cento (Ferrara)University of FerraraFerraraItaly
| | - Emanuele Scafato
- National Observatory on AlcoholNational Institute of HealthRomeItaly
| | - Fabio Caputo
- Centre for the Study and Treatment of Alcohol‐Related DiseasesDepartment of Translational Medicine, University of FerraraFerraraItaly
- Department of Internal Medicine, SS Annunziata Hospital, Cento (Ferrara)University of FerraraFerraraItaly
| |
Collapse
|
1425
|
Verma M, Sharma S, Kumar A, Hakim A, Bhansali S, Meena R. Comorbidities and Vaccination Status of COVID-19 All-Cause Mortality at a Tertiary Care Center of Western India. Cureus 2022; 14:e21721. [PMID: 35251795 PMCID: PMC8886330 DOI: 10.7759/cureus.21721] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/29/2022] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION COVID-19 vaccines have been found to be efficacious for preventing severe disease, yet breakthrough infections and deaths have occurred in a small proportion of vaccinated individuals. This study aimed to describe the vaccination status and comorbidities of COVID-19 all-cause deaths. METHODS This descriptive observational study was conducted at a tertiary care center in western India. A total of 310, RT-PCR positive COVID-19 deaths, aged 45 years and above irrespective of the cause of death (all-cause mortality), were included in the study. Death after breakthrough infection was defined as death in patient with disease onset after 14 days of the second dose of vaccine. RESULTS Diabetes was the most common comorbidity found in 17.1% of the deaths, followed by hypertension. Cardiovascular disease and renal disease were other common comorbidities seen in 8.7% and 4.83% deaths respectively. Other less common comorbidities include neurological disorders, HIV, autoimmune disorders. Out of these 310 deaths, 21.4% of patients developed disease within 14 days of the first dose. Death after true breakthrough infection (after 14 days of both doses) was seen in only two patients (0.6%). One of these two patients was aged 60 years and had diabetes, while the other was aged 72 years and had a history of smoking. CONCLUSION Diabetes and hypertension were the most common comorbidities, indicating a higher risk of mortality among comorbid patients. Only a small proportion of deaths (0.6%) occurred after breakthrough infection beyond 14 days of two doses. COVID-19 vaccines have shown promising efficacy against severe disease, thus high vaccination coverage needs to be achieved to prevent morbidity and mortality.
Collapse
Affiliation(s)
- Manoj Verma
- Community Medicine, Dr. Sampurnanand (S. N. Medical College), Jodhpur, IND
| | - Savitri Sharma
- Community Medicine, Dr. Sampurnanand (S. N. Medical College), Jodhpur, IND
| | - Arun Kumar
- Community Medicine, Dr. Sampurnanand (S. N. Medical College), Jodhpur, IND
| | - Afzal Hakim
- Community Medicine, Dr. Sampurnanand (S. N. Medical College), Jodhpur, IND
| | - Suman Bhansali
- Community Medicine, Dr. Sampurnanand (S. N. Medical College), Jodhpur, IND
| | - Rita Meena
- Community Medicine, Dr. Sampurnanand (S. N. Medical College), Jodhpur, IND
| |
Collapse
|
1426
|
Vaccines. SIDE EFFECTS OF DRUGS ANNUAL 2022. [PMCID: PMC9646283 DOI: 10.1016/bs.seda.2022.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The safety of COVID-19 vaccines, as was the case last year, remains a large part of the focus in this volume. COVID-19 placed a large magnifying glass on both vaccines, specifically vaccine safety. This was most readily apparent as the number of records in VAERS ballooned to about 10 times the size from 2020 to 2021 (Vaccine Adverse Event Reporting System (VAERS), 2022) [S]. While we have added and/or improved VAERS during COVID-19, including adding or improving other vaccine safety surveillance tools like v-safe and vaccine safety datalink (Blumenthal, Phadke, et al., 2021) [MC], there is still room for improvement in these pharmacovigilance tools (Rizk et al., 2021) [r]. A major global initiative in this realm is the Global Vaccines Safety Blueprint 2.0 (GVSB2.0) (Organization, 2021, pp. 2021–2023) [S]. We wholeheartedly endorse these initiatives, which could significantly improve vaccine safety. As noted in past SEDA issues, clinicians should be mindful of the risks of AEs and SAEs associated with each individual vaccine.
Collapse
|
1427
|
López F, Català M, Prats C, Estrada O, Oliva I, Prat N, Isnard M, Vallès R, Vilar M, Clotet B, Argimon JM, Aran A, Ara J. A Cost-Benefit Analysis of COVID-19 Vaccination in Catalonia. Vaccines (Basel) 2021; 10:59. [PMID: 35062719 PMCID: PMC8780175 DOI: 10.3390/vaccines10010059] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/21/2021] [Accepted: 12/29/2021] [Indexed: 11/17/2022] Open
Abstract
(1) Background: In epidemiological terms, it has been possible to calculate the savings in health resources and the reduction in the health effects of COVID vaccines. Conducting an economic evaluation, some studies have estimated its cost-effectiveness; the vaccination shows highly favorable results, cost-saving in some cases. (2) Methods: Cost-benefit analysis of the vaccination campaign in the North Metropolitan Health Region (Catalonia). An epidemiological model based on observational data and before and after comparison is used. The information on the doses used and the assigned resources (conventional hospital beds, ICU, number of tests) was extracted from administrative data from the largest primary care provider in the region (Catalan Institute of Health). A distinction was made between the social perspective and the health system. (3) Results: the costs of vaccination are estimated at 137 million euros (€48.05/dose administered). This figure is significantly lower than the positive impacts of the vaccination campaign, which are estimated at 470 million euros (€164/dose administered). Of these, 18% corresponds to the reduction in ICU discharges, 16% to the reduction in conventional hospital discharges, 5% to the reduction in PCR tests and 1% to the reduction in RAT tests. The monetization of deaths and cases that avoid sequelae account for 53% and 5% of total savings, respectively. The benefit/cost ratio is estimated at 3.4 from a social perspective and 1.4 from a health system perspective. The social benefits of vaccination are estimated at €116.67 per vaccine dose (€19.93 from the perspective of the health system). (4) Conclusions: The mass vaccination campaign against COVID is cost-saving. From a social perspective, most of these savings come from the monetization of the reduction in mortality and cases with sequelae, although the intervention is equally widely cost-effective from the health system perspective thanks to the reduction in the use of resources. It is concluded that, from an economic perspective, the vaccination campaign has high social returns.
Collapse
Affiliation(s)
- Francesc López
- Directorate for Innovation and Interdisciplinary Cooperation, North Metropolitan Territorial Authority, Catalan Institute of Health, 08006 Barcelona, Spain; (O.E.); (I.O.); (J.A.)
- Centre for Research in Health and Economics, Pompeu Fabra University, 08002 Barcelona, Spain
- Fight AIDS and Infectious Diseases Foundation, 08916 Barcelona, Spain;
| | - Martí Català
- Comparative Medicine and Bioimage Centre of Catalonia (CMCiB), Fundació Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, 08916 Barcelona, Spain; (M.C.); (C.P.)
| | - Clara Prats
- Comparative Medicine and Bioimage Centre of Catalonia (CMCiB), Fundació Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, 08916 Barcelona, Spain; (M.C.); (C.P.)
- BIOCOM-SC, Physics Department, Universitat Politècnica de Catalunya, 08034 Barcelona, Spain
| | - Oriol Estrada
- Directorate for Innovation and Interdisciplinary Cooperation, North Metropolitan Territorial Authority, Catalan Institute of Health, 08006 Barcelona, Spain; (O.E.); (I.O.); (J.A.)
| | - Irene Oliva
- Directorate for Innovation and Interdisciplinary Cooperation, North Metropolitan Territorial Authority, Catalan Institute of Health, 08006 Barcelona, Spain; (O.E.); (I.O.); (J.A.)
- Centre for Research in Health and Economics, Pompeu Fabra University, 08002 Barcelona, Spain
| | - Núria Prat
- North Metropolitan Primary Care Directorate, Catalan Institute of Health, 08006 Barcelona, Spain; (N.P.); (M.I.); (R.V.); (M.V.)
| | - Mar Isnard
- North Metropolitan Primary Care Directorate, Catalan Institute of Health, 08006 Barcelona, Spain; (N.P.); (M.I.); (R.V.); (M.V.)
| | - Roser Vallès
- North Metropolitan Primary Care Directorate, Catalan Institute of Health, 08006 Barcelona, Spain; (N.P.); (M.I.); (R.V.); (M.V.)
| | - Marc Vilar
- North Metropolitan Primary Care Directorate, Catalan Institute of Health, 08006 Barcelona, Spain; (N.P.); (M.I.); (R.V.); (M.V.)
| | - Bonaventura Clotet
- Fight AIDS and Infectious Diseases Foundation, 08916 Barcelona, Spain;
- IrsiCaixa AIDS Research Institute, University Hospital Germans Trias i Pujol, 08916 Barcelona, Spain
| | | | - Anna Aran
- Catalan Health Service, Ministry of Health, 08007 Barcelona, Spain;
| | - Jordi Ara
- Directorate for Innovation and Interdisciplinary Cooperation, North Metropolitan Territorial Authority, Catalan Institute of Health, 08006 Barcelona, Spain; (O.E.); (I.O.); (J.A.)
| |
Collapse
|
1428
|
Antibody responses induced by the BNT162b2 mRNA COVID-19 vaccine in healthcare workers in a single community hospital in Japan. J Infect Chemother 2021; 28:539-542. [PMID: 35016824 PMCID: PMC8716179 DOI: 10.1016/j.jiac.2021.12.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/30/2021] [Accepted: 12/22/2021] [Indexed: 12/27/2022]
Abstract
Introduction The effectiveness of several vaccines against coronavirus disease (COVID-19) has been reported in the real-world setting. However, it is still unknown how long antibodies persist following vaccination and whether or not the persistence of antibodies has a protective effect against COVID-19. Methods Healthcare workers who had received two doses of the BNT162b2 mRNA COVID-19 vaccine were enrolled, and a single-center study was conducted at the National Hospital Organization Hakodate National Hospital. Serum samples from all participants were collected 13–21 weeks (median: 20 weeks) after the second dose of vaccination. The antibody titers were measured using an electrochemiluminescence immunoassay (Elecsys® Anti-SARS-CoV-2 S). Data on characteristics of the participants were gathered from patient records and interview sheets. Results A total of 401 participants, among whom 70.1% were women and the median age was 42 years, were evaluated in this study. None of the participants had a definite COVID-19 history, and all participants who received complete vaccination showed positive antibody titers. The antibody titer was observed to be higher in participants with younger age (p < 0.001) and those who were females (p = 0.028). Despite the higher risk of infection than that of the general public, no vaccinated staff developed breakthrough infections. Conclusions This study demonstrates the significant contribution of the BNT162b2 vaccine in the acquisition of anti-SARS-CoV-2S antibodies; therefore, the general population should benefit from these two vaccine doses, which are expected to be protective for at least five months.
Collapse
|
1429
|
Vaezi A, Meysamie A. COVID-19 Vaccines Cost-Effectiveness Analysis: A Scenario for Iran. Vaccines (Basel) 2021; 10:37. [PMID: 35062698 PMCID: PMC8777749 DOI: 10.3390/vaccines10010037] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 11/15/2021] [Accepted: 11/22/2021] [Indexed: 12/18/2022] Open
Abstract
COVID-19 vaccines are supposed to be critical measure for ending the pandemic. Governments had to decide on the type of vaccine to provide for their population. In this decision-making process, cost-effectiveness analysis is considered a helpful tool. This study is a cost-effectiveness analysis utilized to calculate the incremental cost per averted disability-adjusted life year (DALY) by vaccination compared to no vaccination for different COVID-19 vaccines. The incremental cost-effectiveness ratio (ICER) for a vaccination with COVID-19 vaccines was estimated at 6.2 to 121.2 USD to avert one DALY and 566.8 to 10,957.7 USD per one death. The lowest and highest ICERs belong to Ad26.COV2.S and CoronaVac, respectively. Considering the scenario of Iran, vaccines that are recommended include ad26.cov2.s, chadox1-S, rAd26-S + rAd5-S, and BNT162b2 in the order of recommendation.
Collapse
Affiliation(s)
- Atefeh Vaezi
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 8174673461, Iran
| | - Alipasha Meysamie
- Department of Community Medicine, School of Medicine, Tehran University of Medical Sciences, Tehran 1416753955, Iran
- Community Based Participatory Research Center, Iranian Institute for Reduction of High Risk Behaviors, Tehran University of Medical Sciences, Tehran 1416753955, Iran
| |
Collapse
|
1430
|
Nesterenko PA, McLaughlin J, Tsai BL, Burton Sojo G, Cheng D, Zhao D, Mao Z, Bangayan NJ, Obusan MB, Su Y, Ng RH, Chour W, Xie J, Li YR, Lee D, Noguchi M, Carmona C, Phillips JW, Kim JT, Yang L, Heath JR, Boutros PC, Witte ON. HLA-A ∗02:01 restricted T cell receptors against the highly conserved SARS-CoV-2 polymerase cross-react with human coronaviruses. Cell Rep 2021; 37:110167. [PMID: 34919800 PMCID: PMC8660260 DOI: 10.1016/j.celrep.2021.110167] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 10/15/2021] [Accepted: 12/02/2021] [Indexed: 12/14/2022] Open
Abstract
Cross-reactivity and direct killing of target cells remain underexplored for severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2)-specific CD8+ T cells. Isolation of T cell receptors (TCRs) and overexpression in allogeneic cells allows for extensive T cell reactivity profiling. We identify SARS-CoV-2 RNA-dependent RNA polymerase (RdRp/NSP12) as highly conserved, likely due to its critical role in the virus life cycle. We perform single-cell TCRαβ sequencing in human leukocyte antigen (HLA)-A∗02:01-restricted, RdRp-specific T cells from SARS-CoV-2-unexposed individuals. Human T cells expressing these TCRαβ constructs kill target cell lines engineered to express full-length RdRp. Three TCR constructs recognize homologous epitopes from common cold coronaviruses, indicating CD8+ T cells can recognize evolutionarily diverse coronaviruses. Analysis of individual TCR clones may help define vaccine epitopes that can induce long-term immunity against SARS-CoV-2 and other coronaviruses.
Collapse
Affiliation(s)
- Pavlo A Nesterenko
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jami McLaughlin
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Brandon L Tsai
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Urology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Institute for Precision Health, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Giselle Burton Sojo
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Donghui Cheng
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Daniel Zhao
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Urology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Institute for Precision Health, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Zhiyuan Mao
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Nathanael J Bangayan
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Matthew B Obusan
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yapeng Su
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Rachel H Ng
- Institute for Systems Biology, Seattle, WA 98109, USA; Department of Bioengineering, University of Washington, Seattle, WA 98105, USA
| | - William Chour
- Institute for Systems Biology, Seattle, WA 98109, USA; Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA; Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Jingyi Xie
- Institute for Systems Biology, Seattle, WA 98109, USA; Department of Bioengineering, University of Washington, Seattle, WA 98105, USA
| | - Yan-Ruide Li
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Derek Lee
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Miyako Noguchi
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Camille Carmona
- Division of Infectious Diseases, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - John W Phillips
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jocelyn T Kim
- Division of Infectious Diseases, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Lili Yang
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - James R Heath
- Institute for Systems Biology, Seattle, WA 98109, USA; Parker Institute for Cancer Immunotherapy, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Paul C Boutros
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Urology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Institute for Precision Health, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Owen N Witte
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Parker Institute for Cancer Immunotherapy, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
1431
|
Brüssow H, Zuber S. Can a combination of vaccination and face mask wearing contain the COVID-19 pandemic? Microb Biotechnol 2021; 15:721-737. [PMID: 34962710 PMCID: PMC8913850 DOI: 10.1111/1751-7915.13997] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 12/16/2021] [Indexed: 12/13/2022] Open
Abstract
The COVID‐19 pandemic is going into its third year with Europe again being the focus of major epidemic activity. The present review tries to answer the question whether one can come to grip with the pandemic by a combination of vaccinations and non‐pharmaceutical interventions (NPIs). Several COVID‐19 vaccines are of remarkable efficacy and achieve high protection rates against symptomatic disease, especially severe disease, but mathematical models suggest that the current vaccination coverage in many countries is insufficient to achieve pandemic control. NPIs are needed as complementary measures because recent research has also revealed the limits of vaccination alone. Here, we review the evidence for efficacy of face mask wearing in various settings. Overall pooled analysis showed significant reduction in COVID‐19 incidence with mask wearing, although heterogeneity between studies was substantial. Controlled trials of mask wearing are difficult to conduct, separating mask wearing effects in population studies from the impact of other NPIs is challenging and the efficacy of masks depend on mask material and mask fit. The combination of vaccination and mask wearing is potentially synergistic since vaccination protects so far well from disease development (the omicron variant is currently an unknown) but immunity from infection wanes over few months after vaccination. In comparison, masks interfere with the virus transmission process at a level of a physical barrier independent of coronavirus variant. Vaccination and masks are much less costly to apply than other NPI measures which are associated with high economic and social costs, but paradoxically both measures are the target of a vocal opposition by a sizable minority of the society. In parallel with biomedical research, we need more social science research into this opposition to guide political decisions on how to end the pandemic. The present review tries to answer the question whether one can control the pandemic by a combination of vaccinations and non‐pharmaceutical interventions. The combination of vaccination and mask wearing is potentially synergistic since vaccination protects so far well from disease while masks interfere with the virus transmission process as a physical barrier against any type of coronavirus variant.
Collapse
Affiliation(s)
- Harald Brüssow
- Laboratory of Gene Technology, Department of Biosystems, KU Leuven, Leuven, Belgium
| | - Sophie Zuber
- Institute of Food Safety and Analytical Science, Nestlé Research, Lausanne 26, 1000, Switzerland
| |
Collapse
|
1432
|
Ostropolets A, Hripcsak G. COVID-19 vaccination effectiveness rates by week and sources of bias.. [PMID: 34981073 PMCID: PMC8722616 DOI: 10.1101/2021.12.22.21268253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Importance Randomized clinical trials and observational studies have demonstrated high overall effectiveness for the three US-authorized COVID-19 vaccines against symptomatic COVID-19 infection. Nevertheless, the challenges associated with the use of observational data can undermine the results of the studies. Objective To assess the feasibility of using observational data for vaccine effectiveness studies by examining granular weekly effectiveness. Design, Settings and Participants In this retrospective cohort study, we used Columbia University Medical Center data linked to State and City Immunization Registries to assess the weekly effectiveness of mRNA COVID-19 vaccines. We conducted manual chart review of cases in week one in both groups along with a set of sensitivity analyses for Pfizer- BioNTech, Moderna and Janssen vaccines. Main Outcomes and Measures We used propensity score matching with up to 54,987 covariates and fitted Cox proportional hazards models to estimate hazard ratios and constructed Kaplan-Meier plots for two main outcomes (COVID-19 infection and COVID-19-associated hospitalization). Results The study included 179,666 patients. We observed increasing effectiveness after the first dose of mRNA vaccines with week 6 effectiveness approximating 84% (95% CI 72–91%) for COVID-19 infection and 86% (95% CI 69–95) for COVID-19-associated hospitalization. When analyzing unexpectedly high effectiveness in week one, chart review revealed that vaccinated patients are less likely to seek care after vaccination and are more likely to be diagnosed with COVID-19 during the encounters for other conditions. Sensitivity analyses showed potential outcome misclassification for COVID-19 ICD10-CM diagnosis and the influence of excluding patients with prior COVID-19 infection and anchoring in the unexposed group. Overall vaccine effectiveness analysis in fully vaccinated patients matched the results of the randomized trials. Conclusions and Relevance Observational data can be used to ascertain vaccine effectiveness if potential biases are accounted for. The data need to be scrutinized to ensure that compared groups exhibit similar health seeking behavior and are equally likely to be captured in the data. Given the difference in temporal trends of vaccine exposure and baseline characteristics, indirect comparison of vaccines may produce biased results.
Collapse
|
1433
|
Efficacy and safety of the BNT162b2 mRNA COVID-19 vaccine in participants with a history of cancer: subgroup analysis of a global phase 3 randomized clinical trial. Vaccine 2021; 40:1483-1492. [PMID: 35131133 PMCID: PMC8702495 DOI: 10.1016/j.vaccine.2021.12.046] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 12/17/2021] [Accepted: 12/19/2021] [Indexed: 12/12/2022]
Abstract
Introduction Individuals with an underlying malignancy have high risk of poor COVID-19 outcomes. In clinical trials, COVID-19 vaccines were safe and efficacious against infection, hospitalization, and death, but most trials excluded participants with cancer. We report results from participants with a history of past or active neoplasm (malignant or benign/unknown) and up to 6 months’ follow-up post-dose 2 from the placebo-controlled, observer-blinded trial of the 2-dose BNT162b2 mRNA COVID-19 vaccine. Patients and methods Between July 2020–January 2021, 46,429 participants aged ≥ 12 years were randomized at 152 sites in 6 countries. Healthy participants with pre-existing stable neoplasm could participate; those receiving immunosuppressive therapy were excluded. Data are reported for participants, aged ≥ 16 years for safety and ≥ 12 years for efficacy, who had any history of neoplasm at baseline (data cut-off: March 13, 2021). Adverse-event (AE) data are controlled for follow-up time before unblinding and reported as incidence rates (IRs) per 100 person-years follow-up. Results At baseline, 3813 participants had a history of neoplasm; most common malignancies were breast (n = 460), prostate (n = 362), and melanoma (n = 223). Four BNT162b2 and 71 placebo recipients developed COVID-19 from 7 days post-dose 2; vaccine efficacy was 94.4% (95% CI: 85.2, 98.5) after up to 6 months’ follow-up post-dose 2. This compares favorably with vaccine efficacy of 91.1% in the overall trial population after the same follow-up. AEs were reported at IRs of 95.4 (BNT162b2) and 48.3 (placebo) per 100 person-years. Most common AEs were reactogenicity events (injection-site pain, fatigue, pyrexia). Three BNT162b2 and 1 placebo recipients withdrew because of vaccine-related AEs. No vaccine-related deaths were reported. Conclusion In participants with past or active neoplasms, BNT162b2 vaccine has a similar efficacy and safety profile as in the overall trial population. These results can inform BNT162b2 use during the COVID-19 pandemic and future trials in participants with cancer. Clinical trial number: NCT04368728.
Collapse
|
1434
|
Kalaij AGI, Dirjayanto VJ, Yusuf SM, Nelwan EJ. Immunogenicity and safety of adenovirus-based vector vaccines for COVID-19: a systematic review and meta-analysis. MEDICAL JOURNAL OF INDONESIA 2021. [DOI: 10.13181/mji.oa.215559] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
BACKGROUND Despite various research on vaccine development, severe acute respiratory syndrome coronavirus 2 infection continues to spread. Thus, developing a more effective vaccine for production and clinical efficacy is still in high demand. This review aimed to assess the immunogenicity and safety of adenovirus-based vector vaccine (Ad-vaccines) including Ad5-vectored, ChAdOx1 nCoV-19, rAd26-S or rAd5-S, and Ad26.COV2.S as the promising solutions for COVID-19.
METHODS We conducted a systematic review and meta analysis of clinical trials based on the preferred reporting items for systematic reviews and meta-analyses guidelines through PubMed, Scopus, Cochrane, and EBSCOhost until August 17, 2021. We implemented inclusion and exclusion criteria and assessed the studies using OHAT risk of bias rating tool for human and animal studies. Pooled estimates of odds ratio (OR) were analyzed using fixed-effect model.
RESULTS This systematic review yielded 12 clinical studies with a total of 75,105 subjects. Although the studies were heterogeneous, this meta-analysis showed that Ad-vaccine significantly increased protection and immune response against COVID-19 with a pooled efficacy of 84.68% compared to placebo (p<0.00001). Forest plot also indicated that Ad-vaccine conferred protection against moderate to severe COVID-19 with a pooled OR of 0.26 (p<0.00001). Ad-vaccine had also shown a good safety profile with local site pain and fever as the most common side effects.
CONCLUSIONS Ad-vaccine had shown a good immunogenicity for COVID-19 with a good pooled efficacy and was proven safe for COVID-19 patients.
Collapse
|
1435
|
Islam KU, A-Elgadir TME, Afaq S, Ahmad T, Iqbal J. Molecular and Clinical Aspects of COVID-19 Vaccines and Other Therapeutic Interventions Apropos Emerging Variants of Concern. Front Pharmacol 2021; 12:778219. [PMID: 35002711 PMCID: PMC8734653 DOI: 10.3389/fphar.2021.778219] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 11/08/2021] [Indexed: 11/13/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) has overwhelmed the healthcare and economy of the world, with emerging new variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) posing an everlasting threat to humanity. While most COVID-19 vaccines provide adequate protective immunological response against the original SARS-CoV-2 variant, there is a pressing need to understand their biological and clinical responses. Recent evidence suggests that some of the new variants of SARS-CoV-2 evade the protection conferred by the existing vaccines, which may impede the ongoing efforts to expedite the vaccination programs worldwide. These concerns have also highlighted the importance of a pan-COVID-19 vaccine, which is currently in the making. Thus, it is imperative to have a better molecular and clinical understanding of the various COVID-19 vaccines and their immunological trajectory against any emerging variant of concerns (VOCs) in particular to break this vicious cycle. Furthermore, other treatment regimens based on cellular therapies and monoclonal antibodies should be explored systematically as an alternative and readily available option considering the possibility of the emergence of more virulent SARS-CoV-2 mutants. In this review, we shed light on the various molecular mechanisms and clinical responses of COVID-19 vaccines. Importantly, we review the recent findings of their long-term immune protection and efficacy against emerging VOCs. Considering that other targeted and effective treatments will complement vaccine therapy, we provide a comprehensive understanding of the role of cell-based therapies, monoclonal antibodies, and immunomodulatory agents as alternative and readily available treatment modalities against any emerging SARS-CoV-2 variant.
Collapse
Affiliation(s)
- Khursheed Ul Islam
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | | | - Sarah Afaq
- Department of Clinical Biochemistry, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Tanveer Ahmad
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Jawed Iqbal
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
1436
|
Bushman M, Kahn R, Taylor BP, Lipsitch M, Hanage WP. Population impact of SARS-CoV-2 variants with enhanced transmissibility and/or partial immune escape. Cell 2021; 184:6229-6242.e18. [PMID: 34910927 PMCID: PMC8603072 DOI: 10.1016/j.cell.2021.11.026] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/10/2021] [Accepted: 11/15/2021] [Indexed: 12/20/2022]
Abstract
SARS-CoV-2 variants of concern exhibit varying degrees of transmissibility and, in some cases, escape from acquired immunity. Much effort has been devoted to measuring these phenotypes, but understanding their impact on the course of the pandemic-especially that of immune escape-has remained a challenge. Here, we use a mathematical model to simulate the dynamics of wild-type and variant strains of SARS-CoV-2 in the context of vaccine rollout and nonpharmaceutical interventions. We show that variants with enhanced transmissibility frequently increase epidemic severity, whereas those with partial immune escape either fail to spread widely or primarily cause reinfections and breakthrough infections. However, when these phenotypes are combined, a variant can continue spreading even as immunity builds up in the population, limiting the impact of vaccination and exacerbating the epidemic. These findings help explain the trajectories of past and present SARS-CoV-2 variants and may inform variant assessment and response in the future.
Collapse
Affiliation(s)
- Mary Bushman
- Center for Communicable Disease Dynamics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Rebecca Kahn
- Center for Communicable Disease Dynamics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Bradford P Taylor
- Center for Communicable Disease Dynamics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Marc Lipsitch
- Center for Communicable Disease Dynamics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - William P Hanage
- Center for Communicable Disease Dynamics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
1437
|
Zhang L, Dutta S, Xiong S, Chan M, Chan KK, Fan TM, Bailey KL, Lindeblad M, Cooper LM, Rong L, Gugliuzza AF, Shukla D, Procko E, Rehman J, Malik AB. Engineered High-Affinity ACE2 Peptide Mitigates ARDS and Death Induced by Multiple SARS-CoV-2 Variants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.12.21.473668. [PMID: 34981059 PMCID: PMC8722596 DOI: 10.1101/2021.12.21.473668] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Vaccine hesitancy and continuing emergence of SARS-CoV-2 variants of concern that may escape vaccine-induced immune responses highlight the urgent need for effective COVID-19 therapeutics. Monoclonal antibodies used in the clinic have varying efficacies against distinct SARS-CoV-2 variants; thus, there is considerable interest in engineered ACE2 peptides with augmented binding affinities for SARS-CoV-2 Spike protein. These could have therapeutic benefit against multiple viral variants. Using molecular dynamics simulations, we show how three amino acid substitutions in an engineered soluble ACE2 peptide (sACE2 2 .v2.4-IgG1) markedly increase affinity for the SARS-CoV-2 Spike (S) protein. We demonstrate high binding affinity to S protein of the early SARS-CoV-2 WA-1/2020 isolate and also to multiple variants of concern: B.1.1.7 (Alpha), B.1.351 (Beta), P.1 (Gamma), and B.1.617.2 (Delta) SARS-CoV-2 variants. In humanized K18-hACE2 mice, prophylactic and therapeutic administration of sACE2 2 .v2.4-IgG1 peptide prevented acute lung vascular endothelial injury and lung edema (essential features of ARDS) and significantly improved survival after infection by SARS-CoV-2 WA-1/2020 as well as P.1 variant of concern. These studies demonstrate for the first time broad efficacy in vivo of an ACE2 decoy peptide against multiple SARS-CoV-2 variants and point to its therapeutic potential.
Collapse
Affiliation(s)
- Lianghui Zhang
- Department of Pharmacology and Regenerative Medicine and the Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Soumajit Dutta
- Department of Chemical and Biomolecular Engineering, University of Illinois, Urbana, IL 61801, USA
| | - Shiqin Xiong
- Department of Pharmacology and Regenerative Medicine and the Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Matthew Chan
- Department of Chemical and Biomolecular Engineering, University of Illinois, Urbana, IL 61801, USA
| | - Kui K. Chan
- Cyrus Biotechnology, Inc., Seattle, WA 98101, USA
| | - Timothy M. Fan
- Department of Veterinary Clinical Medicine, University of Illinois College of Veterinary Medicine, Urbana, IL 61802, USA
| | - Keith L. Bailey
- Department of Veterinary Clinical Medicine, University of Illinois College of Veterinary Medicine, Urbana, IL 61802, USA
| | - Matthew Lindeblad
- Toxicology Research Laboratory, Department of Pharmacology and Regenerative Medicine, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Laura M. Cooper
- Department of Microbiology and Immunology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Lijun Rong
- Department of Microbiology and Immunology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Anthony F. Gugliuzza
- Department of Pharmacology and Regenerative Medicine and the Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Diwakar Shukla
- Department of Chemical and Biomolecular Engineering, University of Illinois, Urbana, IL 61801, USA
| | - Erik Procko
- Department of Biochemistry, University of Illinois, Urbana, IL 61801, USA
| | - Jalees Rehman
- Department of Pharmacology and Regenerative Medicine and the Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
- Division of Cardiology, Department of Medicine, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Asrar B. Malik
- Department of Pharmacology and Regenerative Medicine and the Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| |
Collapse
|
1438
|
Bolletta E, Iannetta D, Mastrofilippo V, De Simone L, Gozzi F, Croci S, Bonacini M, Belloni L, Zerbini A, Adani C, Fontana L, Salvarani C, Cimino L. Uveitis and Other Ocular Complications Following COVID-19 Vaccination. J Clin Med 2021; 10:5960. [PMID: 34945256 PMCID: PMC8704915 DOI: 10.3390/jcm10245960] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/07/2021] [Accepted: 12/17/2021] [Indexed: 12/20/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) vaccines can cause transient local and systemic post-vaccination reactions. The aim of this study was to report uveitis and other ocular complications following COVID-19 vaccination. The study included 42 eyes of 34 patients (20 females, 14 males), with a mean age of 49.8 years (range 18-83 years). The cases reported were three herpetic keratitis, two anterior scleritis, five anterior uveitis (AU), three toxoplasma retinochoroiditis, two Vogt-Koyanagi-Harada (VKH) disease reactivations, two pars planitis, two retinal vasculitis, one bilateral panuveitis in new-onset Behçet's disease, three multiple evanescent white dot syndromes (MEWDS), one acute macular neuroretinopathy (AMN), five retinal vein occlusions (RVO), one non-arteritic ischemic optic neuropathy (NAION), three activations of quiescent choroidal neovascularization (CNV) secondary to myopia or uveitis, and one central serous chorioretinopathy (CSCR). Mean time between vaccination and ocular complication onset was 9.4 days (range 1-30 days). Twenty-three cases occurred after Pfizer-BioNTech vaccination (BNT162b2 mRNA), 7 after Oxford-AstraZeneca vaccine (ChAdOx1 nCoV-19), 3 after ModernaTX vaccination (mRNA-1273), and 1 after Janssen Johnson & Johnson vaccine (Ad26.COV2). Uveitis and other ocular complications may develop after the administration of COVID-19 vaccine.
Collapse
Affiliation(s)
- Elena Bolletta
- Ocular Immunology Unit, Azienda USL-IRCCS, 42123 Reggio Emilia, Italy; (E.B.); (V.M.); (L.D.S.); (F.G.); (C.A.)
| | - Danilo Iannetta
- Ophthalmology Unit, Azienda USL-IRCCS, 42123 Reggio Emilia, Italy;
| | - Valentina Mastrofilippo
- Ocular Immunology Unit, Azienda USL-IRCCS, 42123 Reggio Emilia, Italy; (E.B.); (V.M.); (L.D.S.); (F.G.); (C.A.)
| | - Luca De Simone
- Ocular Immunology Unit, Azienda USL-IRCCS, 42123 Reggio Emilia, Italy; (E.B.); (V.M.); (L.D.S.); (F.G.); (C.A.)
| | - Fabrizio Gozzi
- Ocular Immunology Unit, Azienda USL-IRCCS, 42123 Reggio Emilia, Italy; (E.B.); (V.M.); (L.D.S.); (F.G.); (C.A.)
| | - Stefania Croci
- Clinical Immunology, Allergy and Advanced Biotechnologies Unit, Azienda USL-IRCCS, 42123 Reggio Emilia, Italy; (S.C.); (M.B.); (L.B.); (A.Z.)
| | - Martina Bonacini
- Clinical Immunology, Allergy and Advanced Biotechnologies Unit, Azienda USL-IRCCS, 42123 Reggio Emilia, Italy; (S.C.); (M.B.); (L.B.); (A.Z.)
| | - Lucia Belloni
- Clinical Immunology, Allergy and Advanced Biotechnologies Unit, Azienda USL-IRCCS, 42123 Reggio Emilia, Italy; (S.C.); (M.B.); (L.B.); (A.Z.)
| | - Alessandro Zerbini
- Clinical Immunology, Allergy and Advanced Biotechnologies Unit, Azienda USL-IRCCS, 42123 Reggio Emilia, Italy; (S.C.); (M.B.); (L.B.); (A.Z.)
| | - Chantal Adani
- Ocular Immunology Unit, Azienda USL-IRCCS, 42123 Reggio Emilia, Italy; (E.B.); (V.M.); (L.D.S.); (F.G.); (C.A.)
| | - Luigi Fontana
- Ophthalmology Unit, DIMES, Alma Mater Studiorum, University of Bologna, S. Orsola-Malpighi Teaching Hospital, 40138 Bologna, Italy;
| | - Carlo Salvarani
- Rheumatology Unit, Azienda USL-IRCCS, 42123 Reggio Emilia, Italy;
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, with Interest in Transplants, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Luca Cimino
- Ocular Immunology Unit, Azienda USL-IRCCS, 42123 Reggio Emilia, Italy; (E.B.); (V.M.); (L.D.S.); (F.G.); (C.A.)
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, with Interest in Transplants, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, 41124 Modena, Italy
| |
Collapse
|
1439
|
Duerr R, Dimartino D, Marier C, Zappile P, Levine S, François F, Iturrate E, Wang G, Dittmann M, Lighter J, Elbel B, Troxel AB, Goldfeld KS, Heguy A. Clinical and genomic signatures of rising SARS-CoV-2 Delta breakthrough infections in New York. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.12.07.21267431. [PMID: 34909779 PMCID: PMC8669846 DOI: 10.1101/2021.12.07.21267431] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
In 2021, Delta has become the predominant SARS-CoV-2 variant worldwide. While vaccines effectively prevent COVID-19 hospitalization and death, vaccine breakthrough infections increasingly occur. The precise role of clinical and genomic determinants in Delta infections is not known, and whether they contribute to increased rates of breakthrough infections compared to unvaccinated controls. Here, we show a steep and near complete replacement of circulating variants with Delta between May and August 2021 in metropolitan New York. We observed an increase of the Delta sublineage AY.25, its spike mutation S112L, and nsp12 mutation F192V in breakthroughs. Delta infections were associated with younger age and lower hospitalization rates than Alpha. Delta breakthroughs increased significantly with time since vaccination, and, after adjusting for confounders, they rose at similar rates as in unvaccinated individuals. Our data indicate a limited impact of vaccine escape in favor of Delta's increased epidemic growth in times of waning vaccine protection.
Collapse
Affiliation(s)
- Ralf Duerr
- Department of Microbiology, NYU Grossman School of Medicine
| | - Dacia Dimartino
- Genome Technology Center, Office of Science and Research, NYU Langone Health
| | - Christian Marier
- Genome Technology Center, Office of Science and Research, NYU Langone Health
| | - Paul Zappile
- Genome Technology Center, Office of Science and Research, NYU Langone Health
| | | | | | | | - Guiqing Wang
- Department of Pathology, NYU Grossman School of Medicine
| | - Meike Dittmann
- Department of Microbiology, NYU Grossman School of Medicine
| | - Jennifer Lighter
- Department of Pediatric Infectious Diseases, NYU Grossman School of Medicine
| | - Brian Elbel
- Department of Population Health, NYU Grossman School of Medicine
- NYU Wagner Graduate School of Public Service
| | - Andrea B. Troxel
- Department of Population Health, NYU Grossman School of Medicine
| | | | - Adriana Heguy
- Genome Technology Center, Office of Science and Research, NYU Langone Health
- Department of Pathology, NYU Grossman School of Medicine
| |
Collapse
|
1440
|
Wu K, Choi A, Koch M, Elbashir S, Ma L, Lee D, Woods A, Henry C, Palandjian C, Hill A, Jani H, Quinones J, Nunna N, O'Connell S, McDermott AB, Falcone S, Narayanan E, Colpitts T, Bennett H, Corbett KS, Seder R, Graham BS, Stewart-Jones GBE, Carfi A, Edwards DK. Variant SARS-CoV-2 mRNA vaccines confer broad neutralization as primary or booster series in mice. Vaccine 2021; 39:7394-7400. [PMID: 34815117 PMCID: PMC8572694 DOI: 10.1016/j.vaccine.2021.11.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 10/04/2021] [Accepted: 11/01/2021] [Indexed: 12/12/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of a global pandemic. Safe and effective COVID-19 vaccines are now available, including mRNA-1273, which has shown 94% efficacy in prevention of symptomatic COVID-19 disease. However, the emergence of SARS-CoV-2 variants has led to concerns of viral escape from vaccine-induced immunity. Several variants have shown decreased susceptibility to neutralization by vaccine-induced immunity, most notably B.1.351 (Beta), although the overall impact on vaccine efficacy remains to be determined. Here, we present the initial evaluation in mice of 2 updated mRNA vaccines designed to target SARS-CoV-2 variants: (1) monovalent mRNA-1273.351 encodes for the spike protein found in B.1.351 and (2) mRNA-1273.211 comprising a 1:1 mix of mRNA-1273 and mRNA-1273.351. Both vaccines were evaluated as a 2-dose primary series in mice; mRNA-1273.351 was also evaluated as a booster dose in animals previously vaccinated with mRNA-1273. The results demonstrated that a primary vaccination series of mRNA-1273.351 was effective at increasing neutralizing antibody titers against B.1.351, while mRNA-1273.211 was effective at providing broad cross-variant neutralization. A third (booster) dose of mRNA-1273.351 significantly increased both wild-type and B.1.351-specific neutralization titers. Both mRNA-1273.351 and mRNA-1273.211 are being evaluated in pre-clinical challenge and clinical studies.
Collapse
Affiliation(s)
- Kai Wu
- Moderna, Inc., 200 Technology Square, Cambridge, MA 02139, USA.
| | - Angela Choi
- Moderna, Inc., 200 Technology Square, Cambridge, MA 02139, USA
| | - Matthew Koch
- Moderna, Inc., 200 Technology Square, Cambridge, MA 02139, USA
| | - Sayda Elbashir
- Moderna, Inc., 200 Technology Square, Cambridge, MA 02139, USA
| | - LingZhi Ma
- Moderna, Inc., 200 Technology Square, Cambridge, MA 02139, USA
| | - Diana Lee
- Moderna, Inc., 200 Technology Square, Cambridge, MA 02139, USA
| | - Angela Woods
- Moderna, Inc., 200 Technology Square, Cambridge, MA 02139, USA
| | - Carole Henry
- Moderna, Inc., 200 Technology Square, Cambridge, MA 02139, USA
| | | | - Anna Hill
- Moderna, Inc., 200 Technology Square, Cambridge, MA 02139, USA
| | - Hardik Jani
- Moderna, Inc., 200 Technology Square, Cambridge, MA 02139, USA
| | - Julian Quinones
- Moderna, Inc., 200 Technology Square, Cambridge, MA 02139, USA
| | - Naveen Nunna
- Moderna, Inc., 200 Technology Square, Cambridge, MA 02139, USA
| | - Sarah O'Connell
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 10 Center Dr, Bethesda, MD 20814, USA
| | - Adrian B McDermott
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 10 Center Dr, Bethesda, MD 20814, USA
| | | | | | - Tonya Colpitts
- Moderna, Inc., 200 Technology Square, Cambridge, MA 02139, USA
| | | | - Kizzmekia S Corbett
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 10 Center Dr, Bethesda, MD 20814, USA
| | - Robert Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 10 Center Dr, Bethesda, MD 20814, USA
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 10 Center Dr, Bethesda, MD 20814, USA
| | | | - Andrea Carfi
- Moderna, Inc., 200 Technology Square, Cambridge, MA 02139, USA
| | - Darin K Edwards
- Moderna, Inc., 200 Technology Square, Cambridge, MA 02139, USA.
| |
Collapse
|
1441
|
Cheng CJ, Lu CY, Chang YH, Sun Y, Chu HJ, Lee CY, Liu CH, Lin CH, Lu CJ, Li CY. Effectiveness of the WHO-Authorized COVID-19 Vaccines: A Rapid Review of Global Reports till 30 June 2021. Vaccines (Basel) 2021; 9:1489. [PMID: 34960235 PMCID: PMC8708265 DOI: 10.3390/vaccines9121489] [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: 11/13/2021] [Revised: 12/01/2021] [Accepted: 12/13/2021] [Indexed: 01/06/2023] Open
Abstract
Large clinical trials have proven the efficacy of the COVID-19 vaccine, and the number of studies about the effectiveness rapidly grew in the first half of the year after mass vaccination was administrated globally. This rapid review aims to provide evidence syntheses as a means to complement the current evidence on the vaccine effectiveness (VE) against various outcomes in real-world settings. Databases (PubMed, EMBASE, and MedRxiv) were searched up to 30 June 2021, (PROSPERO ID: 266866). A total of 39 studies were included, covering over 15 million participants from 11 nations. Among the general population being fully vaccinated, the VE against symptomatic SARS-CoV-2 infection was estimated at 89-97%, 92% (95% CI, 78-97%), and 94% (95% CI, 86-97%) for BNT162b2, ChAdOx1, and mRNA-1273, respectively. As for the protective effects against B.1.617.2-related symptomatic infection, the VE was 88% (95% CI, 85.3-90.1%) by BNT162b2 and 67.0% (95% CI, 61.3-71.8%) by ChAdOx1 after full vaccination. This review revealed a consistently high effectiveness of certain vaccines among the general population in real-world settings. However, scarce data on the major variants of SARS-CoV-2 and the shortness of the study time may limit the conclusions to the mRNA vaccines and ChAdOx1.
Collapse
Affiliation(s)
- Chang-Jie Cheng
- Department of Neurology, En Chu Kong Hospital, New Taipei City 237, Taiwan; (C.-J.C.); (H.-J.C.); (C.-Y.L.); (C.-H.L.); (C.-H.L.); (C.-J.L.)
- Department of Neurology, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Chun-Yi Lu
- Division of Pediatric Infectious Diseases, Department of Pediatrics, National Taiwan University Hospital, Taipei 100, Taiwan;
- College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Ya-Hui Chang
- Department of Public Health, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan;
| | - Yu Sun
- Department of Neurology, En Chu Kong Hospital, New Taipei City 237, Taiwan; (C.-J.C.); (H.-J.C.); (C.-Y.L.); (C.-H.L.); (C.-H.L.); (C.-J.L.)
- Department of Neurology, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Hai-Jui Chu
- Department of Neurology, En Chu Kong Hospital, New Taipei City 237, Taiwan; (C.-J.C.); (H.-J.C.); (C.-Y.L.); (C.-H.L.); (C.-H.L.); (C.-J.L.)
- Department of Neurology, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Chun-Yu Lee
- Department of Neurology, En Chu Kong Hospital, New Taipei City 237, Taiwan; (C.-J.C.); (H.-J.C.); (C.-Y.L.); (C.-H.L.); (C.-H.L.); (C.-J.L.)
- Department of Neurology, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Chang-Hsiu Liu
- Department of Neurology, En Chu Kong Hospital, New Taipei City 237, Taiwan; (C.-J.C.); (H.-J.C.); (C.-Y.L.); (C.-H.L.); (C.-H.L.); (C.-J.L.)
| | - Cheng-Huai Lin
- Department of Neurology, En Chu Kong Hospital, New Taipei City 237, Taiwan; (C.-J.C.); (H.-J.C.); (C.-Y.L.); (C.-H.L.); (C.-H.L.); (C.-J.L.)
| | - Chien-Jung Lu
- Department of Neurology, En Chu Kong Hospital, New Taipei City 237, Taiwan; (C.-J.C.); (H.-J.C.); (C.-Y.L.); (C.-H.L.); (C.-H.L.); (C.-J.L.)
- Department of Neurology, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Chung-Yi Li
- Department of Public Health, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan;
- Department of Public Health, College of Public Health, China Medical University, Taichung 404, Taiwan
- Department of Healthcare Administration, College of Medical and Health Science, Asia University, Taichung 413, Taiwan
| |
Collapse
|
1442
|
Silvestro M, Tessitore A, Orologio I, Sozio P, Napolitano G, Siciliano M, Tedeschi G, Russo A. Headache Worsening after COVID-19 Vaccination: An Online Questionnaire-Based Study on 841 Patients with Migraine. J Clin Med 2021; 10:jcm10245914. [PMID: 34945208 PMCID: PMC8708794 DOI: 10.3390/jcm10245914] [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: 11/15/2021] [Revised: 12/09/2021] [Accepted: 12/12/2021] [Indexed: 12/12/2022] Open
Abstract
Vaccines have represented the breakthrough in the fight against COVID-19. Based on reported headache attacks after vaccination in randomized controlled trials, we focused on the effects of COVID-19 vaccine administration on the migraine population, using an online questionnaire published on Italian Facebook groups oriented to headache patients. We collected data about the demographics and clinical parameters of migraine severity, COVID-19 infection, vaccination, and characteristics of headaches following vaccination. Out of 841 migraine patients filling in the questionnaire, 66.47% and 60.15% patients experienced a headache attack (from 1 hour to 7 days) after the first and the second vaccine dose, respectively. The main finding concerns headaches perceived by 57.60% of patients: attacks following vaccination were referred to as more severe (50.62% of patients), long-lasting (52.80% of patients) and hardwearing (49.69% of patients) compared to the usually experienced migraine attacks. This could be related to the production of inflammatory mediators such as type Iβ interferon. Considering the high prevalence of migraine in the general population, awareness of the possibility of headaches worsening following COVID-19 vaccination in these patients may allow both patients and clinicians to face this clinical entity with conscious serenity, and to reduce the waste of resources towards inappropriate health-care.
Collapse
Affiliation(s)
- Marcello Silvestro
- Headache Center, Department of Advanced Medical and Surgical Sciences (DAMS), University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.S.); (A.T.); (I.O.); (P.S.); (M.S.); (G.T.)
| | - Alessandro Tessitore
- Headache Center, Department of Advanced Medical and Surgical Sciences (DAMS), University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.S.); (A.T.); (I.O.); (P.S.); (M.S.); (G.T.)
| | - Ilaria Orologio
- Headache Center, Department of Advanced Medical and Surgical Sciences (DAMS), University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.S.); (A.T.); (I.O.); (P.S.); (M.S.); (G.T.)
| | - Pasquale Sozio
- Headache Center, Department of Advanced Medical and Surgical Sciences (DAMS), University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.S.); (A.T.); (I.O.); (P.S.); (M.S.); (G.T.)
| | - Giuseppe Napolitano
- Intensity Care Unit, Department of Emergency and Acceptance “Antonio Cardarelli” Hospital, 80131 Naples, Italy;
| | - Mattia Siciliano
- Headache Center, Department of Advanced Medical and Surgical Sciences (DAMS), University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.S.); (A.T.); (I.O.); (P.S.); (M.S.); (G.T.)
| | - Gioacchino Tedeschi
- Headache Center, Department of Advanced Medical and Surgical Sciences (DAMS), University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.S.); (A.T.); (I.O.); (P.S.); (M.S.); (G.T.)
| | - Antonio Russo
- Headache Center, Department of Advanced Medical and Surgical Sciences (DAMS), University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.S.); (A.T.); (I.O.); (P.S.); (M.S.); (G.T.)
- Correspondence: ; Tel.: +39-081-5665119
| |
Collapse
|
1443
|
Peng XL, Cheng JSY, Gong HL, Yuan MD, Zhao XH, Li Z, Wei DX. Advances in the design and development of SARS-CoV-2 vaccines. Mil Med Res 2021; 8:67. [PMID: 34911569 PMCID: PMC8674100 DOI: 10.1186/s40779-021-00360-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 11/15/2021] [Indexed: 01/18/2023] Open
Abstract
Since the end of 2019, coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has spread worldwide. The RNA genome of SARS-CoV-2, which is highly infectious and prone to rapid mutation, encodes both structural and nonstructural proteins. Vaccination is currently the only effective method to prevent COVID-19, and structural proteins are critical targets for vaccine development. Currently, many vaccines are in clinical trials or are already on the market. This review highlights ongoing advances in the design of prophylactic or therapeutic vaccines against COVID-19, including viral vector vaccines, DNA vaccines, RNA vaccines, live-attenuated vaccines, inactivated virus vaccines, recombinant protein vaccines and bionic nanoparticle vaccines. In addition to traditional inactivated virus vaccines, some novel vaccines based on viral vectors, nanoscience and synthetic biology also play important roles in combating COVID-19. However, many challenges persist in ongoing clinical trials.
Collapse
Affiliation(s)
- Xue-Liang Peng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine, Northwest University, Xi’an, 710069 China
| | - Ji-Si-Yu Cheng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine, Northwest University, Xi’an, 710069 China
| | - Hai-Lun Gong
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine, Northwest University, Xi’an, 710069 China
| | - Meng-Di Yuan
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine, Northwest University, Xi’an, 710069 China
| | - Xiao-Hong Zhao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine, Northwest University, Xi’an, 710069 China
| | - Zibiao Li
- Institute of Materials Research and Engineering, A*STAR (Agency for Science, Technology and Research), 2 Fusionopolis Way, Innovis, #08-03, Singapore, 138634 Singapore
| | - Dai-Xu Wei
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine, Northwest University, Xi’an, 710069 China
| |
Collapse
|
1444
|
Jain L, Vij J, Satapathy P, Chakrapani V, Patro B, Kar SS, Singh R, Pala S, Sankhe L, Modi B, Bali S, Rustagi N, Rajagopal V, Kiran T, Goel K, Aggarwal AK, Gupta M, Padhi BK. Factors Influencing COVID-19 Vaccination Intentions Among College Students: A Cross-Sectional Study in India. Front Public Health 2021; 9:735902. [PMID: 34976911 PMCID: PMC8714761 DOI: 10.3389/fpubh.2021.735902] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 11/11/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Students act as messengers in delivering effective messages for better uptake of health-promoting behavior. Understanding their knowledge about coronavirus disease 2019 (COVID-19), intentions to use the COVID-19 vaccine, and its associated factors will help develop promising strategies in vaccine promotion concerning the current COVID-19 pandemic. Methods: A cross-sectional online survey was carried out among students in the healthcare and non-healthcare sectors to assess their intentions to get vaccinated against the COVID-19. A non-probability snowball sampling technique was used to recruit study participants (N = 655) through social media platforms and emails. Study participants were recruited across the country, including six major geographical regions (Eastern, Western, Northern, Southern, North-east, and Central) in India between November 2020 and January 2021 before the introduction of the COVID-19 vaccine. Descriptive statistics were used to present the sociodemographic, and vaccine-related behaviors of the study participants. Key determinants that likely predict vaccine acceptance among students were modeled using logistic regression analysis. For each analysis, p < 0.05 was considered significant. Results: A total of 655 students were recruited, 323 from healthcare and 332 from non-healthcare sectors, to assess their intentions to receive the COVID-19 vaccine. Of the 655 students, 63.8% expressed intentions to receive the COVID-19 vaccine. The acceptance was higher among non-healthcare students (54.07 vs. 45.93%). At the time of the study, 27.8% of the students indicated that they had been exposed to a confirmed COVID-19 patient. A vast majority (93.4%) of the students knew about the COVID-19 virus, and most (89.3%) of them were aware of the development of a COVID-19 vaccine. The history of vaccine hesitancy was found to be low (17.1%). Only one-third (33.4%) of the students showed concern about contracting COVID-19. Trust in the healthcare system [adjusted odds ratio (aOR): 4.13; (95% CI: 2.83-6.04), p < 0.00] and trust in domestic vaccines [aOR: 1.46; (95% CI: 1.02-2.08), p < 0.05] emerged as the significant predictors of student's intention to get vaccinated. Higher acceptance for vaccine was observed among students in the non-healthcare [aOR: 1.982; 95% CI: 1.334-2.946, p < 0.00]. Conclusion: This study shows that the Indian college students had relatively high levels of positive intentions to receive COVID-19 vaccines, although about one-third were not sure or unwilling to receive the vaccine, highlighting possible vaccine hesitancy. Informational campaigns and other strategies to address vaccine hesitancy are needed to promote uptake of COVID-19 vaccines.
Collapse
Affiliation(s)
- Lovely Jain
- Department of Community Medicine, School of Public Health, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, India
| | - Jatina Vij
- Department of Community Medicine, School of Public Health, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, India
| | | | | | - Binod Patro
- Department of Community and Family Medicine, All India Institute of Medical Sciences (AIIMS), Bhubaneswar, India
| | - Sitanshu Sekhar Kar
- Department of Preventive and Social Medicine, Jawaharlal Institute of Postgraduate Medical Education & Research (JIPMER), Puducherry, India
| | - Ritesh Singh
- Department of Community and Family Medicine, All India Institute of Medical Sciences (AIIMS), Kalyani, India
| | - Star Pala
- Department of Community Medicine, North Eastern Indira Gandhi Regional Institute of Health & Medical Sciences (NEIGRIHMS), Shillong, India
| | - Lalit Sankhe
- Grant Medical College, JJ Hospital, Mumbai, India
| | - Bhavesh Modi
- Community Medicine Health & Family Welfare Department, Government of Gujarat, Gandhinagar, India
| | - Surya Bali
- Department of Community and Family Medicine, All India Institute of Medical Sciences (AIIMS), Bhopal, India
| | - Neeti Rustagi
- Department of Community and Family Medicine, All India Institute of Medical Sciences (AIIMS), Medical Research Public University, Jodhpur, India
| | - Vineeth Rajagopal
- Department of Community Medicine, School of Public Health, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, India
| | - Tanvi Kiran
- Department of Community Medicine, School of Public Health, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, India
| | - Kapil Goel
- Department of Community Medicine, School of Public Health, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, India
| | - Arun Kumar Aggarwal
- Department of Community Medicine, School of Public Health, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, India
| | - Madhu Gupta
- Department of Community Medicine, School of Public Health, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, India
| | - Bijaya Kumar Padhi
- Department of Community Medicine, School of Public Health, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, India
| |
Collapse
|
1445
|
Huang Y, Borisov O, Kee JJ, Carpp LN, Wrin T, Cai S, Sarzotti-Kelsoe M, McDanal C, Eaton A, Pajon R, Hural J, Posavad CM, Gill K, Karuna S, Corey L, McElrath MJ, Gilbert PB, Petropoulos CJ, Montefiori DC. Calibration of two validated SARS-CoV-2 pseudovirus neutralization assays for COVID-19 vaccine evaluation. Sci Rep 2021; 11:23921. [PMID: 34907214 PMCID: PMC8671391 DOI: 10.1038/s41598-021-03154-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/19/2021] [Indexed: 12/26/2022] Open
Abstract
Vaccine-induced neutralizing antibodies (nAbs) are key biomarkers considered to be associated with vaccine efficacy. In United States government-sponsored phase 3 efficacy trials of COVID-19 vaccines, nAbs are measured by two different validated pseudovirus-based SARS-CoV-2 neutralization assays, with each trial using one of the two assays. Here we describe and compare the nAb titers obtained in the two assays. We observe that one assay consistently yielded higher nAb titers than the other when both assays were performed on the World Health Organization's anti-SARS-CoV-2 immunoglobulin International Standard, COVID-19 convalescent sera, and mRNA-1273 vaccinee sera. To overcome the challenge this difference in readout poses in comparing/combining data from the two assays, we evaluate three calibration approaches and show that readouts from the two assays can be calibrated to a common scale. These results may aid decision-making based on data from these assays for the evaluation and licensure of new or adapted COVID-19 vaccines.
Collapse
Affiliation(s)
- Yunda Huang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
- Department of Global Health, University of Washington, Seattle, WA, USA.
| | - Oleg Borisov
- Biomedical Advanced Research and Development Authority, Washington, DC, USA
| | - Jia Jin Kee
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Lindsay N Carpp
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Terri Wrin
- LabCorp-Monogram Biosciences, South San Francisco, CA, USA
| | - Suqin Cai
- LabCorp-Monogram Biosciences, South San Francisco, CA, USA
| | - Marcella Sarzotti-Kelsoe
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
- Department of Immunology, Duke University Medical Center, Durham, NC, USA
| | - Charlene McDanal
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Amanda Eaton
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | | | - John Hural
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Christine M Posavad
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Katherine Gill
- Desmond Tutu HIV Centre, University of Cape Town, Cape Town, South Africa
| | - Shelly Karuna
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Lawrence Corey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - M Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Departments of Medicine and Laboratory Medicine, University of Washington, Seattle, WA, USA
| | - Peter B Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | | | | |
Collapse
|
1446
|
Saharia K, Anjan S, Streit J, Beekmann SE, Polgreen PM, Kuehnert M, Segev DL, Baddley JW, Miller RA. Clinical characteristics of COVID-19 in solid organ transplant recipients following COVID-19 vaccination: A multicenter case series. Transpl Infect Dis 2021; 24:e13774. [PMID: 34905269 DOI: 10.1111/tid.13774] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 11/23/2021] [Accepted: 11/29/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND Solid organ transplant recipients (SOTR) have diminished humoral immune responses to COVID-19 vaccination and higher rates of COVID-19 vaccine breakthrough infection than the general population. Little is known about COVID-19 disease severity in SOTR with COVID-19 vaccine breakthrough infections. METHODS Between 4/7/21 and 6/21/21 we requested case reports via the Emerging Infections Network (EIN) listserv of SARS-CoV-2 infection following COVID-19 vaccination in SOTR. Online data collection included patient demographics, dates of COVID-19 vaccine administration and clinical data related to COVID-19. We performed a descriptive analysis of patient factors and evaluated variables contributing to critical disease or need for hospitalization. RESULTS Sixty-six cases of SARS-CoV-2 infection after vaccination in SOTR were collected. COVID-19 occurred after the second vaccine dose in 52 (78.8%) cases of which 43 (82.7%) occurred ≥14 days post-vaccination. There were 6 deaths, 3 occurring in fully vaccinated individuals (7.0%, n = 3/43). There was no difference in the percentage of patients who recovered from COVID-19 (70.7% vs 72.2%, p = 0.90) among fully and partially vaccinated individuals. We did not identify any differences in hospitalization (60.5% vs. 55.6%, p = 0.72) or critical disease (20.9% vs. 33.3%, p = 0.30) among those who were fully vs. partially vaccinated. CONCLUSIONS SOTR vaccinated against COVID-19 can still develop severe, and even critical, COVID-19 disease. Two doses of mRNA COVID-19 vaccine may be insufficient to protect against severe disease and mortality in SOTR. Future studies to define correlates of protection in SOTR are needed. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Kapil Saharia
- Institute of Human Virology, Division of Infectious Diseases, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Shweta Anjan
- Dept of Medicine, Division of Infectious Diseases, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Judy Streit
- Dept. of Medicine, Division of Infectious Diseases, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Susan E Beekmann
- Dept. of Medicine, Division of Infectious Diseases, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Philip M Polgreen
- Dept. of Medicine, Division of Infectious Diseases, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Matthew Kuehnert
- Dept. of Medicine, Hackensack Meridian School of Medicine, Hackensack, NJ, USA
| | - Dorry L Segev
- Dept. of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - John W Baddley
- Institute of Human Virology, Division of Infectious Diseases, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Rachel A Miller
- Dept. of Medicine, Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
1447
|
Garcia-Beltran WF, St. Denis KJ, Hoelzemer A, Lam EC, Nitido AD, Sheehan ML, Berrios C, Ofoman O, Chang CC, Hauser BM, Feldman J, Gregory DJ, Poznansky MC, Schmidt AG, Iafrate AJ, Naranbhai V, Balazs AB. mRNA-based COVID-19 vaccine boosters induce neutralizing immunity against SARS-CoV-2 Omicron variant. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.12.14.21267755. [PMID: 34931201 PMCID: PMC8687472 DOI: 10.1101/2021.12.14.21267755] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Recent surveillance has revealed the emergence of the SARS-CoV-2 Omicron variant (BA.1/B.1.1.529) harboring up to 36 mutations in spike protein, the target of vaccine-induced neutralizing antibodies. Given its potential to escape vaccine-induced humoral immunity, we measured neutralization potency of sera from 88 mRNA-1273, 111 BNT162b, and 40 Ad26.COV2.S vaccine recipients against wild type, Delta, and Omicron SARS-CoV-2 pseudoviruses. We included individuals that were vaccinated recently (<3 months), distantly (6-12 months), or recently boosted, and accounted for prior SARS-CoV-2 infection. Remarkably, neutralization of Omicron was undetectable in most vaccinated individuals. However, individuals boosted with mRNA vaccines exhibited potent neutralization of Omicron only 4-6-fold lower than wild type, suggesting that boosters enhance the cross-reactivity of neutralizing antibody responses. In addition, we find Omicron pseudovirus is more infectious than any other variant tested. Overall, this study highlights the importance of boosters to broaden neutralizing antibody responses against highly divergent SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Wilfredo F. Garcia-Beltran
- These authors contributed equally
- Department of Pathology, Massachusetts General Hospital, Boston, MA, 02114, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | - Kerri J. St. Denis
- These authors contributed equally
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | - Angelique Hoelzemer
- First Department of Internal Medicine, Division of Infectious Diseases, University Medical Centre Eppendorf, Hamburg, Germany
- German Center for Infection Research (DZIF), Site Hamburg-Lübeck-Borstel-Riems, Germany
| | - Evan C. Lam
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | - Adam D. Nitido
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | | | - Cristhian Berrios
- Department of Pathology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Onosereme Ofoman
- Department of Pathology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Christina C. Chang
- Center for the AIDS Programme of Research in South Africa, Durban, 4001, South Africa
| | - Blake M. Hauser
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | - Jared Feldman
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | - David J. Gregory
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Boston, MA, 02129, USA
- Pediatric Infectious Disease, Massachusetts General Hospital for Children, Boston, MA 02114, USA
| | - Mark C. Poznansky
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Boston, MA, 02129, USA
- Massachusetts General Hospital Cancer Center, Boston, MA, 02114, USA
| | - Aaron G. Schmidt
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | - A. John Iafrate
- Department of Pathology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Vivek Naranbhai
- Center for the AIDS Programme of Research in South Africa, Durban, 4001, South Africa
- Massachusetts General Hospital Cancer Center, Boston, MA, 02114, USA
- Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | | |
Collapse
|
1448
|
Chen RE, Gorman MJ, Zhu DY, Carreño JM, Yuan D, VanBlargan LA, Burdess S, Lauffenburger DA, Kim W, Turner JS, Droit L, Handley SA, Chahin S, Deepak P, O'Halloran JA, Paley MA, Presti RM, Wu GF, Krammer F, Alter G, Ellebedy AH, Kim AHJ, Diamond MS. Reduced antibody activity against SARS-CoV-2 B.1.617.2 delta virus in serum of mRNA-vaccinated individuals receiving tumor necrosis factor-α inhibitors. MED 2021; 2:1327-1341.e4. [PMID: 34812429 PMCID: PMC8599018 DOI: 10.1016/j.medj.2021.11.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/27/2021] [Accepted: 11/11/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Although vaccines effectively prevent coronavirus disease 2019 (COVID-19) in healthy individuals, they appear to be less immunogenic in individuals with chronic inflammatory disease (CID) or receiving chronic immunosuppression therapy. METHODS Here we assessed a cohort of 77 individuals with CID treated as monotherapy with chronic immunosuppressive drugs for antibody responses in serum against historical and variant severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) viruses after immunization with the BNT162b2 mRNA vaccine. FINDINGS Longitudinal analysis showed the greatest reductions in neutralizing antibodies and Fc effector function capacity in individuals treated with tumor necrosis factor alpha (TNF-α) inhibitors (TNFi), and this pattern appeared to be worse against the B.1.617.2 delta virus. Within 5 months of vaccination, serum neutralizing titers of all TNFi-treated individuals tested fell below the presumed threshold correlate for antibody-mediated protection. However, TNFi-treated individuals receiving a third mRNA vaccine dose boosted their serum neutralizing antibody titers by more than 16-fold. CONCLUSIONS Vaccine boosting or administration of long-acting prophylaxis (e.g., monoclonal antibodies) will likely be required to prevent SARS-CoV-2 infection in this susceptible population. FUNDING This study was supported by grants and contracts from the NIH (R01 AI157155, R01AI151178, and HHSN75N93019C00074; NIAID Centers of Excellence for Influenza Research and Response (CEIRR) contracts HHSN272201400008C and 75N93021C00014; and Collaborative Influenza Vaccine Innovation Centers [CIVIC] contract 75N93019C00051).
Collapse
Affiliation(s)
- Rita E Chen
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Daniel Y Zhu
- Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Juan Manuel Carreño
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dansu Yuan
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Laura A VanBlargan
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Samantha Burdess
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Wooseob Kim
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jackson S Turner
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Lindsay Droit
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Scott A Handley
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Salim Chahin
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Parakkal Deepak
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Jane A O'Halloran
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Michael A Paley
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Rachel M Presti
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO, USA
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, Saint Louis, MO, USA
| | - Gregory F Wu
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Ali H Ellebedy
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
- Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO, USA
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, Saint Louis, MO, USA
| | - Alfred H J Kim
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO, USA
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
- Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO, USA
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
1449
|
Classen JM, Muzalyova A, Nagl S, Fleischmann C, Ebigbo A, Römmele C, Messmann H, Schnoy E. Antibody Response to SARS-CoV-2 Vaccination in Patients with Inflammatory Bowel Disease: Results of a Single-Center Cohort Study in a Tertiary Hospital in Germany. Dig Dis 2021; 40:719-727. [PMID: 34915480 PMCID: PMC8805066 DOI: 10.1159/000521343] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 12/01/2021] [Indexed: 02/02/2023]
Abstract
BACKGROUND COVID-19 is a viral disease caused by severe acute respiratory syndrome corona virus 2 (SARS-CoV-2), first described in 2019, with a significant impact on everyday life since then. In December 2020, the first vaccine against COVID-19 from BioNTech/Pfizer was approved for the first time. However, little is known about the immune response to vaccination in patients with inflammatory bowel disease (IBD) and immunomodulators or biologics. The aim of our study was to investigate antibody response to SARS-CoV-2 vaccination in patients with IBD receiving immunomodulators or biologics compared to healthy controls. METHODS This was a single-center study with a retrospective observational design. Seventy-two patients with ulcerative colitis or Crohn's disease were included. Matching data from 72 healthy employees of our hospital were used as the control group. Data were matched by propensity score to patients with IBD. Blood samples were taken from both groups for antibody response, and both groups received an accompanying questionnaire. RESULTS Sixty-five (90.3%) patients of the IBD group reported taking immunomodulatory therapy. The mean antibody level for all IBD patients was 1,257.1 U/mL (standard deviation [SD] 1,109.626) in males and 1,500.1 U/mL (SD 1142.760) in female IBD patients after full vaccination. Compared to the healthy group, reduced antibody response could be detected (IBD group 1,383.76 U/mL SD 1,125.617; control group 1,885.65 U/mL SD 727.572, p < 0.05). In this group, blood samples were taken with an average of 61.9 days after the first vaccination. There was no vaccination failure in the IBD group after 2 vaccinations. After the first vaccination, side effects, including muscle pain, pain at the injection site, and fatigue, were reported more often in IBD patients than in the control group (total symptoms IBD group 58.3%, control group 34.5%, p < 0.007). The opposite occurred after the second vaccination when side effects were higher in the control group (total symptoms IBD group 55.4%, control group 76%, p = 0.077). There was a trend to a reduced immune response in elderly patients. Disease duration and concomitant immunomodulatory therapy (TNF-alpha blockers, interleukin inhibitors, integrin inhibitors, methotrexate, or azathioprine) had no impact on the immune response. However, longer time to last medication given and time passed to vaccination in patients with IBD seems to have a positive impact on antibody levels. CONCLUSION Overall, we could show a high antibody response to vaccination with COVID-19 in all patients with IBD after 2 vaccinations. Vaccination was well tolerated, and no other adverse events were detected. Concomitant immunomodulatory therapy (TNF-alpha blockers, interleukin inhibitors, integrin inhibitors, methotrexate, or azathioprine) had no impact on seroconversion. Further evaluation of antibody titers over time is mandatory to detect early the need for re-vaccination in these patients.
Collapse
|
1450
|
Al-Emran HM, Hasan MS, Ahasan Setu MA, Rahman MS, Alam ARU, Sarkar SL, Islam MT, Islam MR, Rahman MM, Islam OK, Jahid IK, Hossain MA. Genomic analysis of SARS-CoV-2 variants of concern identified from the ChAdOx1 nCoV-19 immunized patients from Southwest part of Bangladesh. J Infect Public Health 2021; 15:156-163. [PMID: 34952247 PMCID: PMC8688836 DOI: 10.1016/j.jiph.2021.12.002] [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: 08/05/2021] [Revised: 11/25/2021] [Accepted: 12/01/2021] [Indexed: 11/21/2022] Open
Abstract
Background Bangladesh introduced ChAdOx1 nCoV-19 since February, 2021 and in six months, only a small population (12.8%) received either one or two dose of vaccination like other low-income countries. The COVID-19 infections were continued to roll all over the places although the information on genomic variations of SARS-CoV-2 between both immunized and unimmunized group was unavailable. The objective of this study was to compare the proportion of immune escaping variants between those groups. Methods A total of 4718 nasopharygeal samples were collected from March 1 until April 15, 2021, of which, 834 (18%) were SARS-CoV-2 positive. The minimum sample size was calculated as 108 who were randomly selected for telephone interview and provided consent. The prevalence of SARS-CoV-2 variants and disease severity among both immunized and unimmunized groups was measured. A total of 63 spike protein sequences and 14 whole-genome sequences were performed from both groups and phylogenetic reconstruction and mutation analysis were compared. Results A total of 40 respondents (37%, N = 108) received single-dose and 2 (2%) received both doses of ChAdOx1 nCoV-19 vaccine, which significantly reduce dry cough, loss of appetite and difficulties in breathing compared to none. There was no significant difference in hospitalization, duration of hospitalization or reduction of other symptoms like running nose, muscle pain, shortness of breathing or generalized weakness between immunized and unimmunized groups. Spike protein sequence assumed 21 (87.5%) B.1.351, one B.1.526 and two 20B variants in immunized group compared to 27 (69%) B.1.351, 5 (13%) B.1.1.7, 4 (10%) 20B, 2 B.1.526 and one B.1.427 variant in unimmunized group. Whole genome sequence analysis of 14 cases identified seven B.1.351 Beta V2, three B.1.1.7 Alpha V1, one B.1.526 Eta and the rest three 20B variants. Conclusion Our study observed that ChAdOx1 could not prevent the new infection or severe COVID-19 disease outcome with single dose while the infections were mostly caused by B.1.351 variants in Bangladesh.
Collapse
Affiliation(s)
- Hassan M Al-Emran
- Department of Biomedical Engineering, Jashore University of Science and Technology, Jashore 7408, Bangladesh; Genome Centre, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - Md Shazid Hasan
- Genome Centre, Jashore University of Science and Technology, Jashore 7408, Bangladesh; Department of Microbiology, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - Md Ali Ahasan Setu
- Genome Centre, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - M Shaminur Rahman
- Department of Microbiology, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - Asm Rubayet Ul Alam
- Genome Centre, Jashore University of Science and Technology, Jashore 7408, Bangladesh; Department of Microbiology, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - Shovon Lal Sarkar
- Genome Centre, Jashore University of Science and Technology, Jashore 7408, Bangladesh; Department of Microbiology, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - Md Tanvir Islam
- Genome Centre, Jashore University of Science and Technology, Jashore 7408, Bangladesh; Department of Microbiology, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - Mir Raihanul Islam
- Poverty, Health, and Nutrition Division, International Food Policy Research Institute, Dhaka, Bangladesh
| | - Mohammad Mahfuzur Rahman
- Department of Environmental Science and Technology, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - Ovinu Kibria Islam
- Genome Centre, Jashore University of Science and Technology, Jashore 7408, Bangladesh; Department of Microbiology, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - Iqbal Kabir Jahid
- Genome Centre, Jashore University of Science and Technology, Jashore 7408, Bangladesh; Department of Microbiology, Jashore University of Science and Technology, Jashore 7408, Bangladesh.
| | - M Anwar Hossain
- Genome Centre, Jashore University of Science and Technology, Jashore 7408, Bangladesh; Department of Microbiology, University of Dhaka, Dhaka 1000, Bangladesh; Vice-Chancellor, Jashore University of Science and Technology, Jashore 7408, Bangladesh.
| |
Collapse
|