1
|
Ahmad MI, Amorim CG, Abu Qatouseh LF, Montenegro MCBSM. Nanobody-based immunosensor for the detection of H. pylori in saliva. Biosens Bioelectron 2024; 260:116423. [PMID: 38810413 DOI: 10.1016/j.bios.2024.116423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 05/20/2024] [Accepted: 05/22/2024] [Indexed: 05/31/2024]
Abstract
Helicobacter pylori (H. pylori) infection is highly prevalent worldwide, affecting more than 43% of world population. The infection can be transmitted through different routes, like oral-oral, fecal-oral, and gastric-oral. Electrochemical sensors play a crucial role in the early detection of various substances, including biomolecules. In this study, the development of nanobody (Nb)-based immunosensor for the detection of H. pylori antigens in saliva samples was investigated. The D2_Nb was isolated and characterized using Western blot and ELISA and employed in the fabrication of the immunosensor. The sensor was prepared using gold screen-printed electrodes, with the immobilization of Nb achieved through chemical linkage using cysteamine-glutaraldehyde. The surface of the electrode was characterized using EIS, FTIR and SEM. Initially, the Nb-based immunosensor's performance was evaluated through cyclic voltammetry (CV), differential pulse voltammetry (DPV), and square wave voltammetry (SWV). The sensor exhibited excellent linearity with an R2 value of 0.96. However, further assessment with the DPV technique revealed both a low limit of detection (5.9 ng/mL, <1 cfu/mL) and high selectivity when exposed to a mixture of similar antigens. Moreover, the immunosensor demonstrated robust recovery rates (96.2%-103.4%) when spiked into artificial saliva and maintained its functionality when stored at room temperature for 24 days.
Collapse
Affiliation(s)
- Mohammad Ia Ahmad
- LAQV-REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Célia G Amorim
- LAQV-REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal.
| | - Luay F Abu Qatouseh
- Department of Pharmacology and Biomedical Sciences, Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, Jordan.
| | - Maria C B S M Montenegro
- LAQV-REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| |
Collapse
|
2
|
Holloway PM, Gibson MD, Holloway TT, van Doremalen N, Munster VJ, Al-Omari B, Letko MC, Nash S, Cardwell JM, Abu-Basha EA, Hayajneh W, Mangtani P, Guitian J. MERS-CoV exposure and risk factors for MERS-CoV ELISA seropositivity among members of livestock-owning households in southern Jordan: a population based cross-sectional study. THE LANCET. MICROBE 2024; 5:100866. [PMID: 39053480 DOI: 10.1016/s2666-5247(24)00082-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 03/05/2024] [Accepted: 03/19/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND Although dromedary camels (Camelus dromedarius) are known to be the host reservoir for MERS-CoV, the virus causing Middle East respiratory syndrome (MERS), zoonotic transmission pathways and camel subpopulations posing highest transmission risk are poorly understood. Extensively managed herds, ubiquitous across the Arabian Peninsula, present a major potential source of primary infection. In this study we aimed to address key knowledge gaps regarding MERS epidemiology among high-risk communities associated with such herds, which is essential information for effective control strategies. METHODS We did a cross-sectional study between Sept 27, 2017, and Oct 11, 2018, among members of livestock-owning households in southern Jordan (Aqaba East, Aqaba West, Ma'an East, and Ma'an West regions), with random selection of households (house and tent dwellings) from Ministry of Agriculture lists via computer-generated randomisation lists. Household visits were done, with questionnaires administered to household members regarding potential risk factors for MERS-CoV exposure in the past 6 months and blood samples and nasal and oral swabs collected, alongside physical examination data including blood pressure and blood glucose. Children younger than 5 years and individuals without capacity to provide informed consent were excluded. Serum was tested for IgG antibodies to MERS-CoV spike protein (S1 subunit) and nucleocapsid (N) protein with in-house indirect ELISAs, and viral RNA was detected in nasal and oral samples by RT-PCR. The primary outcome was evidence of MERS-CoV exposure (ascertained by seropositive status on S1 or N ELISAs, or a positive swab sample on RT-PCR); secondary outcomes were potential associations between possible risk factors and seropositive status. RT-PCR data were to be presented descriptively. Seroprevalence estimates were obtained at the individual and household levels, and associations between hypothetical risk factors and seropositive status were assessed with use of mixed-effects logistic regression. FINDINGS We sampled 879 household members (median age 27 years [IQR 16-44]; 471 [54%] males and 408 [46%] females) from 204 households. 72 (8%) household members were seropositive on S1 ELISA (n=25, 3%) or N ELISA (n=52, 6%). No positive nasal or oral swab samples were identified on RT-PCR. Within-household clustering was identified for seropositivity on S1 ELISA (intraclass correlation coefficient 0·88 [0·35-0·96]) but not N ELISA (0·00 [0·00-0·27]). On multivariable analysis, S1 ELISA seropositivity was associated with frequently (≥weekly) interacting with young (age <1 year) camels (adjusted odds ratio [ORadj] 3·85 [95% CI 1·41-11·61], p=0·011), with frequent kissing and petting (ORadj 4·56 [1·55-15·42], p=0·0074), and frequent feeding and watering (ORadj 4·97 [1·80-15·29], p=0·0027) of young camels identified as risk activities. Attending camel races (ORadj 3·73 [1·11-12·47], p=0·029), frequently feeding and watering camels of any age (ORadj 3·18 [1·12-10·84], p=0·040), and elevated blood glucose (>150 mg/dL; ORadj 4·59 [1·23-18·36], p=0·021) were also associated with S1 ELISA seropositivity. Among individuals without history of camel contact, S1 ELISA seropositivity was associated with sharing a household with an S1 ELISA-positive household member (ORadj 8·92 [1·06-92·99], p=0·044), and with sharing a household with an S1 ELISA-positive household member with history of camel contact (ORadj 24·74 [2·72-306·14], p=0·0050). N ELISA seropositivity was associated with age (categorical, p=0·0069), a household owning a young camel (age <18 months; ORadj 1·98 [1·02-4·09], p=0·043), and frequently feeding and watering camels of any age (ORadj 1·98 [1·09-3·69]; p=0·025). INTERPRETATION The study findings highlight the importance of effective MERS-CoV surveillance and control strategies among camel-owning communities in Jordan and the Arabian Peninsula. Juvenile dromedaries pose increased risk for zoonotic MERS-CoV transmission and should be prioritised for vaccination once such vaccines become available. Among high-risk communities, vaccination strategies should prioritise camel-owning households, particularly individuals engaged in camel husbandry or racing, and household members who are older or diabetic, with evidence to suggest secondary within-household transmission. FUNDING UK Medical Research Council and US National Institute of Allergy and Infectious Diseases.
Collapse
Affiliation(s)
- Peter M Holloway
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, London, UK.
| | - Matthew D Gibson
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, London, UK
| | - Tanja T Holloway
- Department of Infectious Disease Epidemiology and International Health, London School of Hygiene & Tropical Medicine, London, UK
| | - Neeltje van Doremalen
- Virus Ecology Section, Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Vincent J Munster
- Virus Ecology Section, Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Bilal Al-Omari
- Faculty of Veterinary Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Michael C Letko
- Laboratory of Functional Viromics, Paul Allen School for Global Health, Washington State University, Pullman, WA, USA
| | - Stephen Nash
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Jacqueline M Cardwell
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, London, UK
| | - Ehab A Abu-Basha
- Faculty of Veterinary Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Wail Hayajneh
- Department of Paediatrics, School of Medicine, Saint Louis University, St Louis, MO, USA
| | - Punam Mangtani
- Department of Infectious Disease Epidemiology and International Health, London School of Hygiene & Tropical Medicine, London, UK
| | - Javier Guitian
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, London, UK
| |
Collapse
|
3
|
Chandran D, Chakraborty S, Chandran D, Subedi D, Jisha AI, Chopra H, Rabaan AA, Al-Tawfiq JA, Islam MR, Dhama K. Middle East Respiratory Syndrome Coronavirus Could be a Priority Pathogen to Cause Public Health Emergency: Noticeable Features and Counteractive Measures. ENVIRONMENTAL HEALTH INSIGHTS 2024; 18:11786302241271545. [PMID: 39156879 PMCID: PMC11329908 DOI: 10.1177/11786302241271545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 07/02/2024] [Indexed: 08/20/2024]
Abstract
Middle East respiratory syndrome (MERS) is caused by a specific strain of the 6 types of human coronaviruses (HCoV). MERS-CoV has spread unchecked since it was first discovered in Saudi Arabia in 2012. The virus most likely spreads through nosocomial and zoonotic channels. Genetic analyses suggest that bats were the initial hosts and that the disease spread to camels. Person-to-person transmission occurs with varying frequency, being most prevalent in clinical settings and the least common among the general population and among close relatives. Due to the severity of the illness, high fatality rate, potential for epidemic spread, and lack of adequate medical countermeasures, the World Health Organization (WHO) continues to list MERS-CoV as a priority pathogen. While no specific antiviral medicines exist, a combination of antivirals has shown promise in recent clinical trials. Vaccines against MERS-CoV are critically needed and are currently being developed. Early diagnosis and implementing appropriate infection control measures are keys to preventing hospital-associated outbreaks. Preventive measures include avoiding raw or undercooked meats and other animal products, ensuring proper hand hygiene in healthcare settings and around dromedaries, educating the public and healthcare personnel about the disease, and adhering to other recommended practices. Countries with a high prevalence of MERS should adhere to regulations designed to limit the transmission of the virus. The recent spread of MERS-CoV highlights the importance of public awareness regarding the significance of reporting symptoms so that appropriate control measures can be adopted. The narrative review discusses the incidence of MERS, its clinical presentation, potential transmission routes, recent reports, preventative and control measures, and current therapeutic options.
Collapse
Affiliation(s)
- Deepak Chandran
- Department of Animal Husbandry, Government of Kerala, Kerala, India
| | - Sandip Chakraborty
- State Disease Investigation Laboratory, ARDD, Abhoynagar, Agartala, West Tripura, India
| | - Diljith Chandran
- KVG Dental College & Hospital, Kurunjibhag, Sullia, Karnataka, India
| | - Deepak Subedi
- Department of Poultry Science, University of Georgia, Athens, Georgia, USA
| | - Ankitha Indu Jisha
- Amrita School of Agricultural Sciences, Amrita Vishwa Vidyapeetham University, Coimbatore, Tamil Nadu, India
| | - Hitesh Chopra
- Department of Biosciences, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
| | - Ali A Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
- Department of Public Health and Nutrition, The University of Haripur, Haripur, Pakistan
| | - Jaffar A Al-Tawfiq
- Specialty Internal Medicine and Quality Patient Safety Department, Johns Hopkins Aramco Healthcare, Dhahran, Saudi Arabia
- Infectious Diseases Division, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Infectious Diseases Division, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Md. Rabiul Islam
- School of Pharmacy, BRAC University, Merul Badda, Dhaka, Bangladesh
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Izatnagar, Uttar Pradesh, India
| |
Collapse
|
4
|
Abdelazim M, Abdelkader R, Ali A, Shahein MA, Tadesse Z, Saad A, Mansour A, Ali SF, Atea M, Gardner E, VonDobschuetz S, Morzaria S, Makonnen Y, Lubroth J, Sumption K, ElMasry I, Zakaria T, Eid S, Hatab EA, Hagag NM, Yousef HMY, Emara M, Abdelwahed DA, Abdelmegeed HK, Hamdy ME, Mansour ONO, Guitian J. A longitudinal study of Middle East respiratory syndrome coronavirus (MERS-CoV) in dromedary camels. BMC Vet Res 2023; 19:228. [PMID: 37919680 PMCID: PMC10621169 DOI: 10.1186/s12917-023-03769-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 10/03/2023] [Indexed: 11/04/2023] Open
Abstract
BACKGROUND Middle East respiratory syndrome coronavirus (MERS-CoV) was identified in humans in 2012. Since then, 2605 cases and 937 associated deaths have been reported globally. Camels are the natural host for MERS-CoV and camel to human transmission has been documented. The relationship between MERS-CoV shedding and presence of neutralizing antibodies in camels is critical to inform surveillance and control, including future deployment of camel vaccines. However, it remains poorly understood. The longitudinal study conducted in a closed camel herd in Egypt between December 2019 and March 2020 helped to characterize the kinetics of MERS-CoV neutralizing antibodies and its relation with viral shedding. RESULTS During the 100-day longitudinal study, 27 out of 54 camels (50%) consistently tested negative for presence of antibodies against MERS-CoV, 19 (35.2%) tested positive and 8 (14.8%) had both, positive and negative test results. Fourteen events that could be interpreted as serological indication of probable infection (two seroconversions and twelve instances of positive camels more than doubling their optical density ratio (OD ratio) in consecutive samples) were identified. Observed times between the identified events provided strong evidence (p = 0.002) against the null hypothesis that they occurred with constant rate during the study, as opposed to clustering at certain points in time. A generalized additive model showed that optical density ratio (OD ratio) is positively associated with being an adult and varies across individual camels and days, peaking at around days 20 and 90 of the study. Despite serological indication of probable virus circulation and intense repeated sampling, none of the tested nasal swab samples were positive for MERS-CoV RNA, suggesting that, if the identified serological responses are the result of virus circulation, the virus may be present in nasal tissue of infected camels during a very narrow time window. CONCLUSIONS Longitudinal testing of a closed camel herd with past history of MERS-CoV infection is compatible with the virus continuing to circulate in the herd despite lack of contact with other camels. It is likely that episodes of MERS-CoV infection in camels can take place with minimal presence of the virus in their nasal tissues, which has important implications for future surveillance and control of MERS-CoV in camel herds and prevention of its zoonotic transmission.
Collapse
Affiliation(s)
| | | | - Abdelhakim Ali
- General Organization for Veterinary Service, Cairo, Egypt
| | - Momtaz A Shahein
- Agriculture Research Center, Animal Health Research Institute, Cairo, Egypt
| | - Zelalem Tadesse
- Food and Agriculture Organizations of the United Nations (FAO), Cairo, Egypt
| | - Ahmed Saad
- Food and Agriculture Organizations of the United Nations (FAO), Cairo, Egypt
| | - Amal Mansour
- Food and Agriculture Organizations of the United Nations (FAO), Regional Office for the Middle East and North Africa, Cairo, Egypt
| | - Samah F Ali
- Agriculture Research Center, Animal Health Research Institute, Cairo, Egypt
| | - Mohamed Atea
- General Organization for Veterinary Service, Cairo, Egypt
| | - Emma Gardner
- Food and Agriculture Organizations of the United Nations (FAO), Rome, Italy
| | | | - Subhash Morzaria
- Institute for Infectious Animal Diseases, Texas A & M University, College Station, USA
| | - Yilma Makonnen
- Food and Agriculture Organizations of the United Nations (FAO), Sub-regional Office for Eastern Africa, Addis Ababa, Ethiopia
| | | | - Keith Sumption
- Food and Agriculture Organizations of the United Nations (FAO), Rome, Italy
| | - Ihab ElMasry
- Food and Agriculture Organizations of the United Nations (FAO), Rome, Italy
| | - Tarek Zakaria
- General Organization for Veterinary Service, Cairo, Egypt
| | - Samah Eid
- Agriculture Research Center, Animal Health Research Institute, Cairo, Egypt
| | - Eman Abo Hatab
- Agriculture Research Center, Animal Health Research Institute, Cairo, Egypt
| | - Naglaa M Hagag
- Agriculture Research Center, Animal Health Research Institute, Cairo, Egypt
| | | | - Mervate Emara
- General Organization for Veterinary Service, Cairo, Egypt
| | - Dina A Abdelwahed
- Agriculture Research Center, Animal Health Research Institute, Cairo, Egypt
| | - Hala K Abdelmegeed
- Agriculture Research Center, Animal Health Research Institute, Cairo, Egypt
| | - Mervat E Hamdy
- Agriculture Research Center, Animal Health Research Institute, Cairo, Egypt
| | - Othman N O Mansour
- Agriculture Research Center, Animal Health Research Institute, Cairo, Egypt
| | | |
Collapse
|
5
|
Cantoni D, Wilkie C, Bentley EM, Mayora-Neto M, Wright E, Scott S, Ray S, Castillo-Olivares J, Heeney JL, Mattiuzzo G, Temperton NJ. Correlation between pseudotyped virus and authentic virus neutralisation assays, a systematic review and meta-analysis of the literature. Front Immunol 2023; 14:1184362. [PMID: 37790941 PMCID: PMC10544934 DOI: 10.3389/fimmu.2023.1184362] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 08/28/2023] [Indexed: 10/05/2023] Open
Abstract
Background The virus neutralization assay is a principal method to assess the efficacy of antibodies in blocking viral entry. Due to biosafety handling requirements of viruses classified as hazard group 3 or 4, pseudotyped viruses can be used as a safer alternative. However, it is often queried how well the results derived from pseudotyped viruses correlate with authentic virus. This systematic review and meta-analysis was designed to comprehensively evaluate the correlation between the two assays. Methods Using PubMed and Google Scholar, reports that incorporated neutralisation assays with both pseudotyped virus, authentic virus, and the application of a mathematical formula to assess the relationship between the results, were selected for review. Our searches identified 67 reports, of which 22 underwent a three-level meta-analysis. Results The three-level meta-analysis revealed a high level of correlation between pseudotyped viruses and authentic viruses when used in an neutralisation assay. Reports that were not included in the meta-analysis also showed a high degree of correlation, with the exception of lentiviral-based pseudotyped Ebola viruses. Conclusion Pseudotyped viruses identified in this report can be used as a surrogate for authentic virus, though care must be taken in considering which pseudotype core to use when generating new uncharacterised pseudotyped viruses.
Collapse
Affiliation(s)
- Diego Cantoni
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow, United Kingdom
| | - Craig Wilkie
- School of Mathematics & Statistics, University of Glasgow, Glasgow, United Kingdom
| | - Emma M. Bentley
- Medicines and Healthcare Products Regulatory Agency, South Mimms, United Kingdom
| | - Martin Mayora-Neto
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Greenwich and Kent at Medway, Chatham, United Kingdom
| | - Edward Wright
- Viral Pseudotype Unit, School of Life Sciences, University of Sussex, Brighton, United Kingdom
| | - Simon Scott
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Greenwich and Kent at Medway, Chatham, United Kingdom
| | - Surajit Ray
- School of Mathematics & Statistics, University of Glasgow, Glasgow, United Kingdom
| | - Javier Castillo-Olivares
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge University, Cambridge, United Kingdom
| | - Jonathan Luke Heeney
- Laboratory of Viral Zoonotics, Department of Veterinary Medicine, University of Cambridge, Cambridge University, Cambridge, United Kingdom
- DIOSynVax, University of Cambridge, Cambridge, United Kingdom
| | - Giada Mattiuzzo
- Medicines and Healthcare Products Regulatory Agency, South Mimms, United Kingdom
| | - Nigel James Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Greenwich and Kent at Medway, Chatham, United Kingdom
| |
Collapse
|
6
|
Rodon J, Te N, Segalés J, Vergara-Alert J, Bensaid A. Enhanced antiviral immunity and dampened inflammation in llama lymph nodes upon MERS-CoV sensing: bridging innate and adaptive cellular immune responses in camelid reservoirs. Front Immunol 2023; 14:1205080. [PMID: 37388723 PMCID: PMC10300347 DOI: 10.3389/fimmu.2023.1205080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 05/30/2023] [Indexed: 07/01/2023] Open
Abstract
Middle East respiratory syndrome coronavirus (MERS-CoV) infection can cause fatal pulmonary inflammatory disease in humans. Contrarily, camelids and bats are the main reservoir hosts, tolerant for MERS-CoV replication without suffering clinical disease. Here, we isolated cervical lymph node (LN) cells from MERS-CoV convalescent llamas and pulsed them with two different viral strains (clades B and C). Viral replication was not supported in LN, but a cellular immune response was mounted. Reminiscent Th1 responses (IFN-γ, IL-2, IL-12) were elicited upon MERS-CoV sensing, accompanied by a marked and transient peak of antiviral responses (type I IFNs, IFN-λ3, ISGs, PRRs and TFs). Importantly, expression of inflammatory cytokines (TNF-α, IL-1β, IL-6, IL-8) or inflammasome components (NLRP3, CASP1, PYCARD) was dampened. The role of IFN-λ3 to counterbalance inflammatory processes and bridge innate and adaptive immune responses in camelid species is discussed. Our findings shed light into key mechanisms on how reservoir species control MERS-CoV in the absence of clinical disease.
Collapse
Affiliation(s)
- Jordi Rodon
- Unitat Mixta d’Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Catalonia, Spain
- IRTA, Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Catalonia, Spain
| | - Nigeer Te
- Unitat Mixta d’Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Catalonia, Spain
- IRTA, Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Catalonia, Spain
| | - Joaquim Segalés
- Unitat Mixta d’Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Catalonia, Spain
- Departament de Sanitat i Anatomia Animals, Facultat de Veterinaria, Universitat Autònoma de Barcelona (UAB), Catalonia, Spain
| | - Júlia Vergara-Alert
- Unitat Mixta d’Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Catalonia, Spain
- IRTA, Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Catalonia, Spain
| | - Albert Bensaid
- Unitat Mixta d’Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Catalonia, Spain
- IRTA, Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Catalonia, Spain
| |
Collapse
|
7
|
Meyer Zu Natrup C, Schünemann LM, Saletti G, Clever S, Herder V, Joseph S, Rodriguez M, Wernery U, Sutter G, Volz A. MERS-CoV‒Specific T-Cell Responses in Camels after Single MVA-MERS-S Vaccination. Emerg Infect Dis 2023; 29:1236-1239. [PMID: 37209676 PMCID: PMC10202854 DOI: 10.3201/eid2906.230128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2023] Open
Abstract
We developed an ELISPOT assay for evaluating Middle East respiratory syndrome coronavirus (MERS-CoV)‒specific T-cell responses in dromedary camels. After single modified vaccinia virus Ankara-MERS-S vaccination, seropositive camels showed increased levels of MERS-CoV‒specific T cells and antibodies, indicating suitability of camel vaccinations in disease-endemic areas as a promising approach to control infection.
Collapse
|
8
|
Laydon DJ, Cauchemez S, Hinsley WR, Bhatt S, Ferguson NM. Impact of proactive and reactive vaccination strategies for health-care workers against MERS-CoV: a mathematical modelling study. Lancet Glob Health 2023; 11:e759-e769. [PMID: 37061313 PMCID: PMC10101755 DOI: 10.1016/s2214-109x(23)00117-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 04/17/2023]
Abstract
BACKGROUND Several vaccine candidates are in development against MERS-CoV, which remains a major public health concern. In anticipation of available MERS-CoV vaccines, we examine strategies for their optimal deployment among health-care workers. METHODS Using data from the 2013-14 Saudi Arabia epidemic, we use a counterfactual analysis on inferred transmission trees (who-infected-whom analysis) to assess the potential impact of vaccination campaigns targeting health-care workers, as quantified by the proportion of cases or deaths averted. We investigate the conditions under which proactive campaigns (ie vaccinating in anticipation of the next outbreak) would outperform reactive campaigns (ie vaccinating in response to an unfolding outbreak), considering vaccine efficacy, duration of vaccine protection, effectiveness of animal reservoir control measures, wait (time between vaccination and next outbreak, for proactive campaigns), reaction time (for reactive campaigns), and spatial level (hospital, regional, or national, for reactive campaigns). We also examine the relative efficiency (cases averted per thousand doses) of different strategies. FINDINGS The spatial scale of reactive campaigns is crucial. Proactive campaigns outperform campaigns that vaccinate health-care workers in response to outbreaks at their hospital, unless vaccine efficacy has waned significantly. However, reactive campaigns at the regional or national levels consistently outperform proactive campaigns, regardless of vaccine efficacy. When considering the number of cases averted per vaccine dose administered, the rank order is reversed: hospital-level reactive campaigns are most efficient, followed by regional-level reactive campaigns, with national-level and proactive campaigns being least efficient. If the number of cases required to trigger reactive vaccination increases, the performance of hospital-level campaigns is greatly reduced; the impact of regional-level campaigns is variable, but that of national-level campaigns is preserved unless triggers have high thresholds. INTERPRETATION Substantial reduction of MERS-CoV morbidity and mortality is possible when vaccinating only health-care workers, underlining the need for countries at risk of outbreaks to stockpile vaccines when available. FUNDING UK Medical Research Council, UK National Institute for Health Research, UK Research and Innovation, UK Academy of Medical Sciences, The Novo Nordisk Foundation, The Schmidt Foundation, and Investissement d'Avenir France.
Collapse
Affiliation(s)
- Daniel J Laydon
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, School of Public Health, Faculty of Medicine, Imperial College London, London, UK.
| | - Simon Cauchemez
- Mathematical Modelling of Infectious Diseases Unit, Institut Pasteur, Université Paris Cité, CNRS UMR2000, Paris, France
| | - Wes R Hinsley
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, School of Public Health, Faculty of Medicine, Imperial College London, London, UK
| | - Samir Bhatt
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, School of Public Health, Faculty of Medicine, Imperial College London, London, UK; Section of Epidemiology, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Neil M Ferguson
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, School of Public Health, Faculty of Medicine, Imperial College London, London, UK
| |
Collapse
|
9
|
Peletta A, Lemoine C, Courant T, Collin N, Borchard G. Meeting vaccine formulation challenges in an emergency setting: Towards the development of accessible vaccines. Pharmacol Res 2023; 189:106699. [PMID: 36796463 DOI: 10.1016/j.phrs.2023.106699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/16/2023]
Abstract
Vaccination is considered one of the most successful strategies to prevent infectious diseases. In the event of a pandemic or epidemic, the rapid development and distribution of the vaccine to the population is essential to reduce mortality, morbidity and transmission. As seen during the COVID-19 pandemic, the production and distribution of vaccines has been challenging, in particular for resource-constrained settings, essentially slowing down the process of achieving global coverage. Pricing, storage, transportation and delivery requirements of several vaccines developed in high-income countries resulted in limited access for low-and-middle income countries (LMICs). The capacity to manufacture vaccines locally would greatly improve global vaccine access. In particular, for the development of classical subunit vaccines, the access to vaccine adjuvants is a pre-requisite for more equitable access to vaccines. Vaccine adjuvants are agents required to augment or potentiate, and possibly target the specific immune response to such type of vaccine antigens. Openly accessible or locally produced vaccine adjuvants may allow for faster immunization of the global population. For local research and development of adjuvanted vaccines to expand, knowledge on vaccine formulation is of paramount importance. In this review, we aim to discuss the optimal characteristics of a vaccine developed in an emergency setting by focusing on the importance of vaccine formulation, appropriate use of adjuvants and how this may help overcome barriers for vaccine development and production in LMICs, achieve improved vaccine regimens, delivery and storage requirements.
Collapse
Affiliation(s)
- Allegra Peletta
- Section of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland (ISPSO), University of Geneva, Rue Michel-Servet 1, 1221 Geneva, Switzerland.
| | - Céline Lemoine
- Vaccine Formulation Institute, Rue du Champ-Blanchod 4, 1228 Plan-les-Ouates, Switzerland.
| | - Thomas Courant
- Vaccine Formulation Institute, Rue du Champ-Blanchod 4, 1228 Plan-les-Ouates, Switzerland.
| | - Nicolas Collin
- Vaccine Formulation Institute, Rue du Champ-Blanchod 4, 1228 Plan-les-Ouates, Switzerland.
| | - Gerrit Borchard
- Section of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland (ISPSO), University of Geneva, Rue Michel-Servet 1, 1221 Geneva, Switzerland.
| |
Collapse
|
10
|
Rabaan AA, Mutair AA, Aljeldah M, Shammari BRA, Sulaiman T, Alshukairi AN, Alfaresi M, Al-Jishi JM, Al Bati NA, Al-Mozaini MA, Bshabshe AA, Almatouq JA, Abuzaid AA, Alfaraj AH, Al-Adsani W, Alabdullah M, Alwarthan S, Alsalman F, Alwashmi ASS, Alhumaid S. Genetic Variants and Protective Immunity against SARS-CoV-2. Genes (Basel) 2022; 13:2355. [PMID: 36553622 PMCID: PMC9778397 DOI: 10.3390/genes13122355] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/09/2022] [Accepted: 12/10/2022] [Indexed: 12/16/2022] Open
Abstract
The novel coronavirus-19 (SARS-CoV-2), has infected numerous individuals worldwide, resulting in millions of fatalities. The pandemic spread with high mortality rates in multiple waves, leaving others with moderate to severe symptoms. Co-morbidity variables, including hypertension, diabetes, and immunosuppression, have exacerbated the severity of COVID-19. In addition, numerous efforts have been made to comprehend the pathogenic and host variables that contribute to COVID-19 susceptibility and pathogenesis. One of these endeavours is understanding the host genetic factors predisposing an individual to COVID-19. Genome-Wide Association Studies (GWAS) have demonstrated the host predisposition factors in different populations. These factors are involved in the appropriate immune response, their imbalance influences susceptibility or resistance to viral infection. This review investigated the host genetic components implicated at the various stages of viral pathogenesis, including viral entry, pathophysiological alterations, and immunological responses. In addition, the recent and most updated genetic variations associated with multiple host factors affecting COVID-19 pathogenesis are described in the study.
Collapse
Affiliation(s)
- Ali A. Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran 31311, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
- Department of Public Health and Nutrition, The University of Haripur, Haripur 22610, Pakistan
| | - Abbas Al Mutair
- Research Center, Almoosa Specialist Hospital, Al-Ahsa 36342, Saudi Arabia
- College of Nursing, Princess Norah Bint Abdulrahman University, Riyadh 11564, Saudi Arabia
- School of Nursing, Wollongong University, Wollongong, NSW 2522, Australia
- Nursing Department, Prince Sultan Military College of Health Sciences, Dhahran 33048, Saudi Arabia
| | - Mohammed Aljeldah
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Hafr Al Batin, Hafr Al Batin 39831, Saudi Arabia
| | - Basim R. Al Shammari
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Hafr Al Batin, Hafr Al Batin 39831, Saudi Arabia
| | - Tarek Sulaiman
- Infectious Diseases Section, Medical Specialties Department, King Fahad Medical City, Riyadh 12231, Saudi Arabia
| | - Abeer N. Alshukairi
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
- Department of Medicine, King Faisal Specialist Hospital and Research Center, Jeddah 21499, Saudi Arabia
| | - Mubarak Alfaresi
- Department of Pathology and Laboratory Medicine, Sheikh Khalifa General Hospital, Umm Al Quwain 499, United Arab Emirates
- Department of Pathology, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai 505055, United Arab Emirates
| | - Jumana M. Al-Jishi
- Internal Medicine Department, Qatif Central Hospital, Qatif 35342, Saudi Arabia
| | - Neda A. Al Bati
- Medical and Clinical Affairs, Rural Health Network, Eastern Health Cluster, Dammam 31444, Saudi Arabia
| | - Maha A. Al-Mozaini
- Immunocompromised Host Research Section, Department of Infection and Immunity, King Faisal, Specialist Hospital and Research Centre, Riyadh 11564, Saudi Arabia
| | - Ali Al Bshabshe
- Adult Critical Care Department of Medicine, Division of Adult Critical Care, College of Medicine, King Khalid University, Abha 62561, Saudi Arabia
| | - Jenan A. Almatouq
- Department of Clinical Laboratory Sciences, Mohammed Al-Mana College of Health Sciences, Dammam 34222, Saudi Arabia
| | - Abdulmonem A. Abuzaid
- Medical Microbiology Department, Security Forces Hospital Programme, Dammam 32314, Saudi Arabia
| | - Amal H. Alfaraj
- Pediatric Department, Abqaiq General Hospital, First Eastern Health Cluster, Abqaiq 33261, Saudi Arabia
| | - Wasl Al-Adsani
- Department of Medicine, Infectious Diseases Hospital, Kuwait City 63537, Kuwait
- Department of Infectious Diseases, Hampton Veterans Administration Medical Center, Hampton, VA 23667, USA
| | - Mohammed Alabdullah
- Department of Infectious Diseases, Almoosa Specialist Hospital, Al Mubarraz 36342, Saudi Arabia
| | - Sara Alwarthan
- Department of Internal Medicine, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam 34212, Saudi Arabia
| | - Fatimah Alsalman
- Department of Emergency Medicine, Oyun City Hospital, Al-Ahsa 36312, Saudi Arabia
| | - Ameen S. S. Alwashmi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Saad Alhumaid
- Administration of Pharmaceutical Care, Al-Ahsa Health Cluster, Ministry of Health, Al-Ahsa 31982, Saudi Arabia
| |
Collapse
|
11
|
Sunagar R, Prasad SD, Ella R, Vadrevu KM. Preclinical evaluation of safety and immunogenicity of a primary series intranasal COVID-19 vaccine candidate (BBV154) and humoral immunogenicity evaluation of a heterologous prime-boost strategy with COVAXIN (BBV152). Front Immunol 2022; 13:1063679. [PMID: 36569867 PMCID: PMC9773076 DOI: 10.3389/fimmu.2022.1063679] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 11/24/2022] [Indexed: 12/13/2022] Open
Abstract
Most if not all vaccine candidates developed to combat COVID-19 due to SARS-CoV-2 infection are administered parenterally. As SARS-CoV-2 is transmitted through infectious respiratory fluids, vaccine-induced mucosal immunity could provide an important contribution to control this pandemic. ChAd-SARS-CoV-2-S (BBV154), a replication-defective chimpanzee adenovirus (ChAd)-vectored intranasal (IN) COVID-19 vaccine candidate, encodes a prefusion-stabilized version of the SARS-CoV-2 spike protein containing two proline substitutions in the S2 subunit. We performed preclinical evaluations of BBV154 in mice, rats, hamsters and rabbits. Repeated dose toxicity studies presented excellent safety profiles in terms of pathology and biochemical analysis. IN administration of BBV154 elicited robust mucosal and systemic humoral immune responses coupled with Th1 cell-mediated immune responses. BBV154 IN vaccination also elicited potent variant (omicron) cross neutralization antibodies. Assessment of anti-vector (ChAd36) neutralizing antibodies following repeated doses of BBV154 IN administration showed insignificant titers of ChAd36 neutralizing antibodies. However, the immune sera derived from the same animals displayed significantly higher levels of SARS-CoV-2 virus neutralization (p<0.003). We also evaluated the safety and immunogenicity of heterologous prime-boost vaccination with intramuscular (IM) COVAXIN-prime followed by BBV154 IN administration. COVAXIN priming followed by BBV154 IN-booster showed an acceptable reactogenicity profile comparable to the homologous COVAXIN/COVAXIN or BBV154/BBV154 vaccination model. Heterologous vaccination of COVAXIN-prime and BBV154 booster also elicited superior (p<0.005) and cross variant (omicron) protective immune responses (p<0.013) compared with the homologous COVAXIN/COVAXIN schedule. BBV154 has successfully completed both homologous and heterologous combination schedules of human phase 3 clinical trials and received the restricted emergency use approval (in those aged above 18 years) from the Drugs Controller General of India (DCGI).
Collapse
|
12
|
Abdolmaleki G, Taheri MA, Paridehpour S, Mohammadi NM, Tabatabaei YA, Mousavi T, Amin M. A comparison between SARS-CoV-1 and SARS-CoV2: an update on current COVID-19 vaccines. Daru 2022; 30:379-406. [PMID: 36050585 PMCID: PMC9436716 DOI: 10.1007/s40199-022-00446-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 08/05/2022] [Indexed: 10/31/2022] Open
Abstract
Since the outbreak of the novel coronavirus disease 2019 (COVID-19) in Wuhan, China, many health care systems have been heavily engaged in treating and preventing the disease, and the year 2020 may be called as "historic COVID-19 vaccine breakthrough". Due to the COVID-19 pandemic, many companies have initiated investigations on developing an efficient and safe vaccine against the virus. From Moderna and Pfizer in the United States to PastocoVac in Pasteur Institute of Iran and the University of Oxford in the United Kingdom, different candidates have been introduced to the market. COVID-19 vaccine research has been facilitated based on genome and structural information, bioinformatics predictions, epitope mapping, and data obtained from the previous developments of severe acute respiratory syndrome coronavirus (SARS-CoV or SARS-CoV-1) and middle east respiratory syndrome coronavirus (MERS-CoV) vaccine candidates. SARS-CoV genome sequence is highly homologous to the one in COVID-19 and both viruses use the same receptor, angiotensin-converting enzyme 2 (ACE2). Moreover, the immune system responds to these viruses, partially in the same way. Considering the on-going COVID-19 pandemic and previous attempts to manufacture SARS-CoV vaccines, this paper is going to discuss clinical cases as well as vaccine challenges, including those related to infrastructures, transportation, possible adverse reactions, utilized delivery systems (e.g., nanotechnology and electroporation) and probable vaccine-induced mutations.
Collapse
Affiliation(s)
- Gelareh Abdolmaleki
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Room No. 1-221, 16th Azar Street, Tehran, Iran
| | - Mina Azam Taheri
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Room No. 1-221, 16th Azar Street, Tehran, Iran
| | - Sarina Paridehpour
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Room No. 1-221, 16th Azar Street, Tehran, Iran
| | - Neshaut Mashreghi Mohammadi
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Room No. 1-221, 16th Azar Street, Tehran, Iran
- Pharmaceutical Microbiology Group, Pharmaceutical Quality Assurance Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Yasaman Ahmadi Tabatabaei
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Room No. 1-221, 16th Azar Street, Tehran, Iran
| | - Taraneh Mousavi
- Toxicology and Diseases Group (TDG), Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences, Tehran, Iran
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Amin
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Room No. 1-221, 16th Azar Street, Tehran, Iran.
- Pharmaceutical Microbiology Group, Pharmaceutical Quality Assurance Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
13
|
Babaeimarzangou SS, Zaker H, Soleimannezhadbari E, Gamchi NS, Kazeminia M, Tarighi S, Seyedian H, Tsatsakis A, Spandidos DA, Margina D. Vaccine development for zoonotic viral diseases caused by positive‑sense single‑stranded RNA viruses belonging to the Coronaviridae and Togaviridae families (Review). Exp Ther Med 2022; 25:42. [PMID: 36569444 PMCID: PMC9768462 DOI: 10.3892/etm.2022.11741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/10/2022] [Indexed: 12/02/2022] Open
Abstract
Outbreaks of zoonotic viral diseases pose a severe threat to public health and economies worldwide, with this currently being more prominent than it previously was human history. These emergency zoonotic diseases that originated and transmitted from vertebrates to humans have been estimated to account for approximately one billion cases of illness and have caused millions of deaths worldwide annually. The recent emergence of severe acute respiratory syndrome coronavirus-2 (coronavirus disease 2019) is an excellent example of the unpredictable public health threat causing a pandemic. The present review summarizes the literature data regarding the main vaccine developments in human clinical phase I, II and III trials against the zoonotic positive-sense single-stranded RNA viruses belonging to the Coronavirus and Alphavirus genera, including severe acute respiratory syndrome, Middle east respiratory syndrome, Venezuelan equine encephalitis virus, Semliki Forest virus, Ross River virus, Chikungunya virus and O'nyong-nyong virus. That there are neither vaccines nor effective antiviral drugs available against most of these viruses is undeniable. Therefore, new explosive outbreaks of these zoonotic viruses may surely be expected. The present comprehensive review provides an update on the status of vaccine development in different clinical trials against these viruses, as well as an overview of the present results of these trials.
Collapse
Affiliation(s)
- Seyed Sajjad Babaeimarzangou
- Division of Poultry Health and Diseases, Department of Clinical Sciences, Faculty of Veterinary Medicine, Urmia University, Urmia 5756151818, Iran
| | - Himasadat Zaker
- Histology and Microscopic Analysis Division, RASTA Specialized Research Institute (RSRI), West Azerbaijan Science and Technology Park (WASTP), Urmia 5756115322, Iran
| | | | - Naeimeh Shamsi Gamchi
- Histology and Microscopic Analysis Division, RASTA Specialized Research Institute (RSRI), West Azerbaijan Science and Technology Park (WASTP), Urmia 5756115322, Iran
| | - Masoud Kazeminia
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran 1417935840, Iran
| | - Shima Tarighi
- Veterinary Office of West Azerbaijan Province, Urmia 5717617695, Iran
| | - Homayon Seyedian
- Faculty of Veterinary Medicine, Urmia University, Urmia 5756151818, Iran
| | - Aristidis Tsatsakis
- Laboratory of Toxicology, Department of Medicine, University of Crete, 71307 Heraklion, Greece,Correspondence to: Professor Denisa Margina, Department of Biochemistry, Faculty of Pharmacy, ‘Carol Davila’ University of Medicine and Pharmacy, 6 Traian Vuia Street, 020956 Bucharest, Romania
| | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Denisa Margina
- Department of Biochemistry, Faculty of Pharmacy, ‘Carol Davila’ University of Medicine and Pharmacy, 020956 Bucharest, Romania,Correspondence to: Professor Denisa Margina, Department of Biochemistry, Faculty of Pharmacy, ‘Carol Davila’ University of Medicine and Pharmacy, 6 Traian Vuia Street, 020956 Bucharest, Romania
| |
Collapse
|
14
|
Araújo NM, Rubio IGS, Toneto NPA, Morale MG, Tamura RE. The use of adenoviral vectors in gene therapy and vaccine approaches. Genet Mol Biol 2022; 45:e20220079. [PMID: 36206378 PMCID: PMC9543183 DOI: 10.1590/1678-4685-gmb-2022-0079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 07/12/2022] [Indexed: 11/04/2022] Open
Abstract
Adenovirus was first identified in the 1950s and since then this pathogenic group
of viruses has been explored and transformed into a genetic transfer vehicle.
Modification or deletion of few genes are necessary to transform it into a
conditionally or non-replicative vector, creating a versatile tool capable of
transducing different tissues and inducing high levels of transgene expression.
In the early years of vector development, the application in monogenic diseases
faced several hurdles, including short-term gene expression and even a fatality.
On the other hand, an adenoviral delivery strategy for treatment of cancer was
the first approved gene therapy product. There is an increasing interest in
expressing transgenes with therapeutic potential targeting the cancer hallmarks,
inhibiting metastasis, inducing cancer cell death or modulating the immune
system to attack the tumor cells. Replicative adenovirus as vaccines may be even
older and date to a few years of its discovery, application of non-replicative
adenovirus for vaccination against different microorganisms has been
investigated, but only recently, it demonstrated its full potential being one of
the leading vaccination tools for COVID-19. This is not a new vector nor a new
technology, but the result of decades of careful and intense work in this
field.
Collapse
Affiliation(s)
- Natália Meneses Araújo
- Universidade Federal de São Paulo, Laboratório de Biologia Molecular
do Câncer, São Paulo, SP, Brazil.
| | - Ileana Gabriela Sanchez Rubio
- Universidade Federal de São Paulo, Laboratório de Biologia Molecular
do Câncer, São Paulo, SP, Brazil. ,Universidade Federal de São Paulo, Departamento de Ciências
Biológicas, Diadema, SP, Brazil. ,Universidade Federal de São Paulo, Laboratório de Ciências
Moleculares da Tireóide, Diadema, SP, Brazil.
| | | | - Mirian Galliote Morale
- Universidade Federal de São Paulo, Laboratório de Biologia Molecular
do Câncer, São Paulo, SP, Brazil. ,Universidade Federal de São Paulo, Departamento de Ciências
Biológicas, Diadema, SP, Brazil. ,Universidade Federal de São Paulo, Laboratório de Ciências
Moleculares da Tireóide, Diadema, SP, Brazil.
| | - Rodrigo Esaki Tamura
- Universidade Federal de São Paulo, Laboratório de Biologia Molecular
do Câncer, São Paulo, SP, Brazil. ,Universidade Federal de São Paulo, Departamento de Ciências
Biológicas, Diadema, SP, Brazil.
| |
Collapse
|
15
|
Lipid Raft Integrity and Cellular Cholesterol Homeostasis Are Critical for SARS-CoV-2 Entry into Cells. Nutrients 2022; 14:nu14163417. [PMID: 36014919 PMCID: PMC9415163 DOI: 10.3390/nu14163417] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/22/2022] [Accepted: 07/26/2022] [Indexed: 12/20/2022] Open
Abstract
Lipid rafts in cell plasma membranes play a critical role in the life cycle of many viruses. However, the involvement of membrane cholesterol-rich lipid rafts in the entry of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) into target cells is not well known. In this study, we investigated whether the presence of cholesterol-rich microdomains is required for the entry of SARS-CoV-2 into host cells. Our results show that depletion of cholesterol in the rafts by methyl-beta-cyclodextrin (MβCD) treatment impaired the expression of the cell surface receptor angiotensin-converting enzyme 2 (ACE2), resulting in a significant increase in SARS-CoV-2 entry into cells. The effects exerted by MβCD could be substantially reversed by exogenous cholesterol replenishment. In contrast, disturbance of intracellular cholesterol homeostasis by statins or siRNA knockdown of key genes involved in the cholesterol biosynthesis and transport pathways reduced SARS-CoV-2 entry into cells. Our study also reveals that SREBP2-mediated cholesterol biosynthesis is involved in the process of SARS-CoV-2 entry in target cells. These results suggest that the host membrane cholesterol-enriched lipid rafts and cellular cholesterol homeostasis are essential for SARS-CoV-2 entry into cells. Pharmacological manipulation of intracellular cholesterol might provide new therapeutic strategies to alleviate SARS-CoV-2 entry into cells.
Collapse
|
16
|
Folegatti PM, Jenkin D, Morris S, Gilbert S, Kim D, Robertson JS, Smith ER, Martin E, Gurwith M, Chen RT. Vaccines based on the replication-deficient simian adenoviral vector ChAdOx1: Standardized template with key considerations for a risk/benefit assessment. Vaccine 2022; 40:5248-5262. [PMID: 35715352 PMCID: PMC9194875 DOI: 10.1016/j.vaccine.2022.06.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 05/10/2022] [Accepted: 06/02/2022] [Indexed: 02/07/2023]
Abstract
Replication-deficient adenoviral vectors have been under investigation as a platform technology for vaccine development for several years and have recently been successfully deployed as an effective COVID-19 counter measure. A replication-deficient adenoviral vector based on the simian adenovirus type Y25 and named ChAdOx1 has been evaluated in several clinical trials since 2012. The Brighton Collaboration Benefit-Risk Assessment of VAccines by TechnolOgy (BRAVATO) was formed to evaluate the safety and other key features of new platform technology vaccines. This manuscript reviews key features of the ChAdOx1-vectored vaccines. The simian adenovirus Y25 was chosen as a strategy to circumvent pre-existing immunity to common human adenovirus serotypes which could impair immune responses induced by adenoviral vectored vaccines. Deletion of the E1 gene renders the ChAdOx1 vector replication incompetent and further genetic engineering of the E3 and E4 genes allows for increased insertional capability and optimizes vaccine manufacturing processes. ChAdOx1 vectored vaccines can be manufactured in E1 complementing cell lines at scale and are thermostable. The first ChAdOx1 vectored vaccines approved for human use, against SARS-CoV-2, received emergency use authorization in the UK on 30th December 2020, and is now approved in more than 180 countries. Safety data were compiled from phase I-III clinical trials of ChAdOx1 vectored vaccines expressing different antigens (influenza, tuberculosis, malaria, meningococcal B, prostate cancer, MERS-CoV, Chikungunya, Zika and SARS-CoV-2), conducted by the University of Oxford, as well as post marketing surveillance data for the COVID-19 Oxford-AstraZeneca vaccine. Overall, ChAdOx1 vectored vaccines have been well tolerated. Very rarely, thrombosis with thrombocytopenia syndrome (TTS), capillary leak syndrome (CLS), immune thrombocytopenia (ITP), and Guillain-Barre syndrome (GBS) have been reported following mass administration of the COVID-19 Oxford-AstraZeneca vaccine. The benefits of this COVID-19 vaccination have outweighed the risks of serious adverse events in most settings, especially with mitigation of risks when possible. Extensive immunogenicity clinical evaluation of ChAdOx1 vectored vaccines reveal strong, durable humoral and cellular immune responses to date; studies to refine the COVID-19 protection (e.g., via homologous/heterologous booster, fractional dose) are also underway. New prophylactic and therapeutic vaccines based on the ChAdOx1 vector are currently undergoing pre-clinical and clinical assessment, including vaccines against viral hemorrhagic fevers, Nipah virus, HIV, Hepatitis B, amongst others.
Collapse
Affiliation(s)
| | | | | | | | - Denny Kim
- Brighton Collaboration, a program of the Task Force for Global Health, Decatur, GA, USA
| | - James S. Robertson
- Brighton Collaboration, a program of the Task Force for Global Health, Decatur, GA, USA
| | - Emily R. Smith
- Brighton Collaboration, a program of the Task Force for Global Health, Decatur, GA, USA,Corresponding author
| | - Emalee Martin
- Brighton Collaboration, a program of the Task Force for Global Health, Decatur, GA, USA
| | - Marc Gurwith
- Brighton Collaboration, a program of the Task Force for Global Health, Decatur, GA, USA
| | - Robert T. Chen
- Brighton Collaboration, a program of the Task Force for Global Health, Decatur, GA, USA
| | | |
Collapse
|
17
|
Alharbi NK, Aljamaan F, Aljami HA, Alenazi MW, Albalawi H, Almasoud A, Alharthi FJ, Azhar EI, Barhoumi T, Bosaeed M, Gilbert SC, Hashem AM. Immunogenicity of High-Dose MVA-Based MERS Vaccine Candidate in Mice and Camels. Vaccines (Basel) 2022; 10:vaccines10081330. [PMID: 36016218 PMCID: PMC9413082 DOI: 10.3390/vaccines10081330] [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: 06/14/2022] [Revised: 08/12/2022] [Accepted: 08/15/2022] [Indexed: 11/16/2022] Open
Abstract
The Middle East respiratory syndrome coronavirus (MERS-CoV) is a zoonotic pathogen that can transmit from dromedary camels to humans, causing severe pneumonia, with a 35% mortality rate. Vaccine candidates have been developed and tested in mice, camels, and humans. Previously, we developed a vaccine based on the modified vaccinia virus Ankara (MVA) viral vector, encoding a full-length spike protein of MERS-CoV, MVA-MERS. Here, we report the immunogenicity of high-dose MVA-MERS in prime–boost vaccinations in mice and camels. Methods: Three groups of mice were immunised with MVA wild-type (MVA-wt) and MVA-MERS (MVA-wt/MVA-MERS), MVA-MERS/MVA-wt, or MVA-MERS/MVA-MERS. Camels were immunised with two doses of PBS, MVA-wt, or MVA-MERS. Antibody (Ab) responses were evaluated using ELISA and MERS pseudovirus neutralisation assays. Results: Two high doses of MVA-MERS induced strong Ab responses in both mice and camels, including neutralising antibodies. Anti-MVA Ab responses did not affect the immune responses to the vaccine antigen (MERS-CoV spike). Conclusions: MVA-MERS vaccine, administered in a homologous prime–boost regimen, induced high levels of neutralising anti-MERS-CoV antibodies in mice and camels. This could be considered for further development and evaluation as a dromedary vaccine to reduce MERS-CoV transmission to humans.
Collapse
Affiliation(s)
- Naif Khalaf Alharbi
- Vaccine Development Unit, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh 14611, Saudi Arabia
- Correspondence:
| | - Fahad Aljamaan
- Animal Facilities, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
| | - Haya A. Aljami
- Vaccine Development Unit, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
| | - Mohammed W. Alenazi
- Vaccine Development Unit, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
| | - Hind Albalawi
- Vaccine Development Unit, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
| | - Abdulrahman Almasoud
- Vaccine Development Unit, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
| | - Fatima J. Alharthi
- Vaccine Development Unit, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
| | - Esam I. Azhar
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 22254, Saudi Arabia
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 22254, Saudi Arabia
| | - Tlili Barhoumi
- Vaccine Development Unit, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh 14611, Saudi Arabia
| | - Mohammad Bosaeed
- Vaccine Development Unit, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh 14611, Saudi Arabia
- Department of Medicine, King Abdulaziz Medical City, Riyadh 12746, Saudi Arabia
| | | | - Anwar M. Hashem
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah 22254, Saudi Arabia
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 22254, Saudi Arabia
| |
Collapse
|
18
|
Rodon J, Mykytyn AZ, Cantero G, Albulescu IC, Bosch BJ, Brix A, Audonnet JC, Bensaid A, Vergara-Alert J, Haagmans BL, Segalés J. Protective efficacy of an RBD-based Middle East respiratory syndrome coronavirus (MERS-CoV) particle vaccine in llamas. ONE HEALTH OUTLOOK 2022; 4:12. [PMID: 35739576 PMCID: PMC9225808 DOI: 10.1186/s42522-022-00068-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 06/07/2022] [Indexed: 06/15/2023]
Abstract
Ongoing outbreaks of Middle East respiratory syndrome coronavirus (MERS-CoV) continue posing a global health threat. Vaccination of livestock reservoir species is a recommended strategy to prevent spread of MERS-CoV among animals and potential spillover to humans. Using a direct-contact llama challenge model that mimics naturally occurring viral transmission, we tested the efficacy of a multimeric receptor binding domain (RBD) particle-display based vaccine candidate. While MERS-CoV was transmitted to naïve animals exposed to virus-inoculated llamas, immunization induced robust virus-neutralizing antibody responses and prevented transmission in 1/3 vaccinated, in-contact animals. Our exploratory study supports further improvement of the RBD-based vaccine to prevent zoonotic spillover of MERS-CoV.
Collapse
Affiliation(s)
- Jordi Rodon
- Unitat Mixta d'Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Barcelona, Catalonia, 08193, Spain
- IRTA Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB),Bellaterra, Barcelona, Catalonia, 08193, Spain
| | - Anna Z Mykytyn
- Department of Viroscience, Erasmus Medical Centre, Rotterdam, CA, 3000, The Netherlands
| | - Guillermo Cantero
- Unitat Mixta d'Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Barcelona, Catalonia, 08193, Spain
- IRTA Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB),Bellaterra, Barcelona, Catalonia, 08193, Spain
| | - Irina C Albulescu
- Virology Division, Department of Infectious Diseases & Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, CL, 3584, The Netherlands
| | - Berend-Jan Bosch
- Virology Division, Department of Infectious Diseases & Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, CL, 3584, The Netherlands
| | - Alexander Brix
- Boehringer Ingelheim Veterinary Research Center GmbH & Co. KG, Hanover, Germany
| | - Jean-Christophe Audonnet
- Boehringer Ingelheim Animal Health, Global Innovation, 813 Cours du 3ème millénaire, Saint-Priest, 69380, France
| | - Albert Bensaid
- Unitat Mixta d'Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Barcelona, Catalonia, 08193, Spain
- IRTA Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB),Bellaterra, Barcelona, Catalonia, 08193, Spain
| | - Júlia Vergara-Alert
- Unitat Mixta d'Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Barcelona, Catalonia, 08193, Spain
- IRTA Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB),Bellaterra, Barcelona, Catalonia, 08193, Spain
| | - Bart L Haagmans
- Department of Viroscience, Erasmus Medical Centre, Rotterdam, CA, 3000, The Netherlands.
| | - Joaquim Segalés
- Unitat Mixta d'Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Barcelona, Catalonia, 08193, Spain.
- Departament de Sanitat i Anatomia Animals, Facultat de Veterinària, UAB, Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Barcelona, Catalonia, 08193, Spain.
| |
Collapse
|
19
|
Carpenter A, Waltenburg MA, Hall A, Kile J, Killerby M, Knust B, Negron M, Nichols M, Wallace RM, Behravesh CB, McQuiston JH. Vaccine Preventable Zoonotic Diseases: Challenges and Opportunities for Public Health Progress. Vaccines (Basel) 2022; 10:vaccines10070993. [PMID: 35891157 PMCID: PMC9319643 DOI: 10.3390/vaccines10070993] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/08/2022] [Accepted: 06/14/2022] [Indexed: 01/18/2023] Open
Abstract
Zoonotic diseases represent a heavy global burden, causing important economic losses, impacting animal health and production, and costing millions of human lives. The vaccination of animals and humans to prevent inter-species zoonotic disease transmission is an important intervention. However, efforts to develop and implement vaccine interventions to reduce zoonotic disease impacts are often limited to the veterinary and agricultural sectors and do not reflect the shared burden of disease. Multisectoral collaboration, including co-development opportunities for human and animal vaccines, expanding vaccine use to include animal reservoirs such as wildlife, and strategically using vaccines to interrupt complex transmission cycles is needed. Addressing zoonoses requires a multi-faceted One Health approach, wherein vaccinating people and animals plays a critical role.
Collapse
|
20
|
Coughlan L, Kremer EJ, Shayakhmetov DM. Adenovirus-based vaccines-a platform for pandemic preparedness against emerging viral pathogens. Mol Ther 2022; 30:1822-1849. [PMID: 35092844 PMCID: PMC8801892 DOI: 10.1016/j.ymthe.2022.01.034] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/17/2022] [Accepted: 01/21/2022] [Indexed: 11/24/2022] Open
Abstract
Zoonotic viruses continually pose a pandemic threat. Infection of humans with viruses for which we typically have little or no prior immunity can result in epidemics with high morbidity and mortality. These epidemics can have public health and economic impact and can exacerbate civil unrest or political instability. Changes in human behavior in the past few decades-increased global travel, farming intensification, the exotic animal trade, and the impact of global warming on animal migratory patterns, habitats, and ecosystems-contribute to the increased frequency of cross-species transmission events. Investing in the pre-clinical advancement of vaccine candidates against diverse emerging viral threats is crucial for pandemic preparedness. Replication-defective adenoviral (Ad) vectors have demonstrated their utility as an outbreak-responsive vaccine platform during the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic. Ad vectors are easy to engineer; are amenable to rapid, inexpensive manufacturing; are relatively safe and immunogenic in humans; and, importantly, do not require specialized cold-chain storage, making them an ideal platform for equitable global distribution or stockpiling. In this review, we discuss the progress in applying Ad-based vaccines against emerging viruses and summarize their global safety profile, as reflected by their widespread geographic use during the SARS-CoV-2 pandemic.
Collapse
Affiliation(s)
- Lynda Coughlan
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Center for Vaccine Development and Global Health (CVD), University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Eric J Kremer
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, CNRS 5535, Montpellier, France.
| | - Dmitry M Shayakhmetov
- Lowance Center for Human Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA; Emory Vaccine Center, Departments of Pediatrics and Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; Discovery and Developmental Therapeutics Program, Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
21
|
Tai W, Zhang X, Yang Y, Zhu J, Du L. Advances in mRNA and other vaccines against MERS-CoV. Transl Res 2022; 242:20-37. [PMID: 34801748 PMCID: PMC8603276 DOI: 10.1016/j.trsl.2021.11.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 11/16/2021] [Accepted: 11/17/2021] [Indexed: 11/03/2022]
Abstract
Middle East respiratory syndrome coronavirus (MERS-CoV) is a highly pathogenic human coronavirus (CoV). Belonging to the same beta-CoV genus as severe acute respiratory syndrome coronavirus-1 (SARS-CoV-1) and SARS-CoV-2, MERS-CoV has a significantly higher fatality rate with limited human-to-human transmissibility. MERS-CoV causes sporadic outbreaks, but no vaccines have yet been approved for use in humans, thus calling for continued efforts to develop effective vaccines against this important CoV. Similar to SARS-CoV-1 and SARS-CoV-2, MERS-CoV contains 4 structural proteins, among which the surface spike (S) protein has been used as a core component in the majority of currently developed MERS-CoV vaccines. Here, we illustrate the importance of the MERS-CoV S protein as a key vaccine target and provide an update on the currently developed MERS-CoV vaccines, including those based on DNAs, proteins, virus-like particles or nanoparticles, and viral vectors. Additionally, we describe approaches for designing MERS-CoV mRNA vaccines and explore the role and importance of naturally occurring pseudo-nucleosides in the design of effective MERS-CoV mRNA vaccines. This review also provides useful insights into designing and evaluating mRNA vaccines against other viral pathogens.
Collapse
Affiliation(s)
- Wanbo Tai
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York
| | - Xiujuan Zhang
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York
| | - Yang Yang
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, Iowa
| | - Jiang Zhu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, Califonia; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California
| | - Lanying Du
- Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia.
| |
Collapse
|
22
|
Chi H, Wang Y, Li E, Wang X, Wang H, Jin H, Han Q, Wang Z, Wang X, Zhu A, Sun J, Zhuang Z, Zhang L, Ye J, Wang H, Feng N, Hu M, Gao Y, Zhao J, Zhao Y, Yang S, Xia X. Inactivated Rabies Virus Vectored MERS-Coronavirus Vaccine Induces Protective Immunity in Mice, Camels, and Alpacas. Front Immunol 2022; 13:823949. [PMID: 35173733 PMCID: PMC8842186 DOI: 10.3389/fimmu.2022.823949] [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: 11/28/2021] [Accepted: 01/10/2022] [Indexed: 12/26/2022] Open
Abstract
Middle East respiratory syndrome coronavirus (MERS-CoV) is an emergent coronavirus that has caused frequent zoonotic events through camel-to-human spillover. An effective camelid vaccination strategy is probably the best way to reduce human exposure risk. Here, we constructed and evaluated an inactivated rabies virus-vectored MERS-CoV vaccine in mice, camels, and alpacas. Potent antigen-specific antibody and CD8+ T-cell responses were generated in mice; moreover, the vaccination reduced viral replication and accelerated virus clearance in MERS-CoV-infected mice. Besides, protective antibody responses against both MERS-CoV and rabies virus were induced in camels and alpacas. Satisfyingly, the immune sera showed broad cross-neutralizing activity against the three main MERS-CoV clades. For further characterization of the antibody response induced in camelids, MERS-CoV-specific variable domains of heavy-chain-only antibody (VHHs) were isolated from immunized alpacas and showed potent prophylactic and therapeutic efficacies in the Ad5-hDPP4-transduced mouse model. These results highlight the inactivated rabies virus-vectored MERS-CoV vaccine as a promising camelid candidate vaccine.
Collapse
Affiliation(s)
- Hang Chi
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Yanqun Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Entao Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Xiwen Wang
- Food and Drug Inspection Laboratory, Administration for Drug and Instrument Supervision and Inspection, Beijing, China
| | - Hualei Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Hongli Jin
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Qiuxue Han
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Zhenshan Wang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Xinyue Wang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Airu Zhu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jing Sun
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhen Zhuang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lu Zhang
- Health and Quarantine Laboratory, Guangzhou Customs Technology Center, Guangzhou, China
| | - Jingmeiqi Ye
- Institute of Education, Tsinghua University, Beijing, China
| | - Haijun Wang
- Jilin Wild Animal Rescue Breeding Center Committee, Jilin Province Northeast Tiger Garden, Changchun, China
| | - Na Feng
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Mingda Hu
- Sate Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, China
| | - Yuwei Gao
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Jincun Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Institute of Infectious Disease, Guangzhou Eighth People's Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Basic Research, Guangzhou Laboratory, Guangzhou, China
| | - Yongkun Zhao
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Songtao Yang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Xianzhu Xia
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| |
Collapse
|
23
|
Nanoparticle and virus-like particle vaccine approaches against SARS-CoV-2. J Microbiol 2022; 60:335-346. [PMID: 35089583 PMCID: PMC8795728 DOI: 10.1007/s12275-022-1608-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/16/2021] [Accepted: 12/16/2021] [Indexed: 02/06/2023]
Abstract
The global spread of coronavirus disease 2019 caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has provoked an urgent need for prophylactic measures. Several innovative vaccine platforms have been introduced and billions of vaccine doses have been administered worldwide. To enable the creation of safer and more effective vaccines, additional platforms are under development. These include the use of nanoparticle (NP) and virus-like particle (VLP) technology. NP vaccines utilize self-assembling scaffold structures designed to load the entire spike protein or receptor-binding domain of SARS-CoV-2 in a trimeric configuration. In contrast, VLP vaccines are genetically modified recombinant viruses that are considered safe, as they are generally replication-defective. Furthermore, VLPs have indigenous immunogenic potential due to their microbial origin. Importantly, NP and VLP vaccines have shown stronger immunogenicity with greater protection by mimicking the physicochemical characteristics of SARS-CoV-2. The study of NP- and VLP-based coronavirus vaccines will help ensure the development of rapid-response technology against SARS-CoV-2 variants and future coronavirus pandemics.
Collapse
|
24
|
Middle East Respiratory Syndrome coronavirus vaccine development: updating clinical studies using platform technologies. J Microbiol 2022; 60:238-246. [PMID: 35089585 PMCID: PMC8795722 DOI: 10.1007/s12275-022-1547-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/12/2021] [Accepted: 12/15/2021] [Indexed: 12/25/2022]
Abstract
Middle East Respiratory Syndrome coronavirus (MERS-CoV), a contagious zoonotic virus, causes severe respiratory infection with a case fatality rate of approximately 35% in humans. Intermittent sporadic cases in communities and healthcare facility outbreaks have continued to occur since its first identification in 2012. The World Health Organization has declared MERS-CoV a priority pathogen for worldwide research and vaccine development due to its epidemic potential and the insufficient countermeasures available. The Coalition for Epidemic Preparedness Innovations is supporting vaccine development against emerging diseases, including MERS-CoV, based on platform technologies using DNA, mRNA, viral vector, and protein subunit vaccines. In this paper, we review the usefulness and structure of a spike glycoprotein as a MERS-CoV vaccine candidate molecule, and provide an update on the status of MERS-CoV vaccine development. Vaccine candidates based on both DNA and viral vectors coding MERS-CoV spike gene have completed early phase clinical trials. A harmonized approach is required to assess the immunogenicity of various candidate vaccine platforms. Platform technologies accelerated COVID-19 vaccine development and can also be applied to developing vaccines against other emerging viral diseases.
Collapse
|
25
|
Te N, Ciurkiewicz M, van den Brand JMA, Rodon J, Haverkamp AK, Vergara-Alert J, Bensaid A, Haagmans BL, Baumgartner W, Segalés J. Middle East respiratory syndrome coronavirus infection in camelids. Vet Pathol 2022; 59:546-555. [PMID: 35001773 DOI: 10.1177/03009858211069120] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Middle East respiratory syndrome coronavirus (MERS-CoV) is the cause of a severe respiratory disease with a high case fatality rate in humans. Since its emergence in mid-2012, 2578 laboratory-confirmed cases in 27 countries have been reported by the World Health Organization, leading to 888 known deaths due to the disease and related complications. Dromedary camels are considered the major reservoir host for this virus leading to zoonotic infection in humans. Dromedary camels, llamas, and alpacas are susceptible to MERS-CoV, developing a mild-to-moderate upper respiratory tract infection characterized by epithelial hyperplasia as well as infiltration of neutrophils, lymphocytes, and some macrophages within epithelium, lamina propria, in association with abundant viral antigen. The very mild lesions in the lower respiratory tract of these camelids correlate with absence of overt illness following MERS-CoV infection. Unfortunately, there is no approved antiviral treatment or vaccine for MERS-CoV infection in humans. Thus, there is an urgent need to develop intervention strategies in camelids, such as vaccination, to minimize virus spillover to humans. Therefore, the development of camelid models of MERS-CoV infection is key not only to assess vaccine prototypes but also to understand the biologic mechanisms by which the infection can be naturally controlled in these reservoir species. This review summarizes information on virus-induced pathological changes, pathogenesis, viral epidemiology, and control strategies in camelids, as the intermediate hosts and primary source of MERS-CoV infection in humans.
Collapse
Affiliation(s)
- Nigeer Te
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Spain
| | | | | | - Jordi Rodon
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Spain
| | | | - Júlia Vergara-Alert
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Albert Bensaid
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Bart L Haagmans
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Joaquim Segalés
- UAB, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Spain.,Departament de Sanitat i Anatomia Animals, Facultat de Veterinària, UAB, Bellaterra, Barcelona, Spain
| |
Collapse
|
26
|
Bosaeed M, Balkhy HH, Almaziad S, Aljami HA, Alhatmi H, Alanazi H, Alahmadi M, Jawhary A, Alenazi MW, Almasoud A, Alanazi R, Bittaye M, Aboagye J, Albaalharith N, Batawi S, Folegatti P, Ramos Lopez F, Ewer K, Almoaikel K, Aljeraisy M, Alothman A, Gilbert SC, Khalaf Alharbi N. Safety and immunogenicity of ChAdOx1 MERS vaccine candidate in healthy Middle Eastern adults (MERS002): an open-label, non-randomised, dose-escalation, phase 1b trial. THE LANCET. MICROBE 2022; 3:e11-e20. [PMID: 34751259 PMCID: PMC8565931 DOI: 10.1016/s2666-5247(21)00193-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
BACKGROUND ChAdOx1-vectored vaccine candidates against several pathogens have been developed and tested in clinical trials and ChAdOx1 nCoV-19 has now been licensed for emergency use for COVID-19. We assessed the safety and immunogenicity of the ChAdOx1 MERS vaccine in a phase 1b trial in healthy Middle Eastern adults. METHOD MERS002 is an open-label, non-randomised, dose-escalation, phase 1b trial. Healthy Middle Eastern adults aged 18-50 years were included in the study. ChAdOx1 MERS was administered as a single intramuscular injection into the deltoid muscle of the non-dominant arm at three different dose groups: 5·0 × 109 viral particles in a low-dose group, 2·5 × 1010 viral particles in an intermediate-dose group, and 5·0 × 1010 viral particles in a high-dose group. The primary objective was to assess the safety and tolerability of ChAdOx1 MERS, measured by the occurrence of solicited and unsolicited adverse events after vaccination for up to 28 days and occurrence of serious adverse events up to 6 months. The study is registered with ClinicalTrials.gov, NCT04170829. FINDINGS Between Dec 17, 2019, and June 1, 2020, 24 participants were enrolled (six to the low-dose, nine to the intermediate-dose, and nine to the high-dose group) and received a dose; 23 were available for follow-up at 6 months. The one dose of ChAdOx1 MERS vaccine was well tolerated with no serious adverse event reported during the 6 months of follow-up. Most adverse events were mild (67, 74%) and moderate (17, 19%). Six (7%) severe adverse events were reported by two participants in the intermediate-dose group (two feverish, two headache, one joint pain, and one muscle pain). Pain at the injection site was the most common local and overall adverse event, reported by 15 (63%) of the 24 participants. The most common systemic adverse event was headache, reported by 14 (58%), followed by muscle pain reported by 13 (54%). The vaccine induced both antibody and T cell immune responses in all volunteers; antibodies peaked at day 28 and T cell responses peaked at day 14; and continued until the end of follow-up at 6 months. INTERPRETATION The acceptable safety and immunogenicity data from this phase 1b trial of ChAdOx1 MERS vaccine candidate in Healthy Middle Eastern adults, combined with previous safety and immunogenicity data from a trial in the UK, support selecting the ChAdOx1 MERS vaccine for advancement into phase 2 clinical evaluation. FUNDING UK Department of Health and Social Care, using UK Aid funding, managed by the UK National Institute for Health Research; and King Abdullah International Medical Research Center.
Collapse
Affiliation(s)
- Mohammad Bosaeed
- Clinical Trial Services, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- Department of Medicine, King Abdulaziz Medical City in Riyadh, Riyadh, Saudi Arabia
| | | | - Sultan Almaziad
- Department of Medicine, King Abdulaziz Medical City in Riyadh, Riyadh, Saudi Arabia
| | - Haya A Aljami
- Vaccine Development Unit, Infectious Disease Research Department, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Hind Alhatmi
- Department of Medicine, King Abdulaziz Medical City in Riyadh, Riyadh, Saudi Arabia
| | - Hala Alanazi
- Clinical Trial Services, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Mashael Alahmadi
- Vaccine Development Unit, Infectious Disease Research Department, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Ayah Jawhary
- Clinical Trial Services, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Mohammed W Alenazi
- Vaccine Development Unit, Infectious Disease Research Department, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Abdulrahman Almasoud
- Vaccine Development Unit, Infectious Disease Research Department, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Rawan Alanazi
- Clinical Trial Services, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Mustapha Bittaye
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Jeremy Aboagye
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Nahla Albaalharith
- Department of Nursing, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Sarah Batawi
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Pedro Folegatti
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Fernando Ramos Lopez
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Katie Ewer
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Khalid Almoaikel
- Clinical Trial Services, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Majed Aljeraisy
- Clinical Trial Services, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Adel Alothman
- Clinical Trial Services, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- Department of Medicine, King Abdulaziz Medical City in Riyadh, Riyadh, Saudi Arabia
| | - Sarah C Gilbert
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Naif Khalaf Alharbi
- Vaccine Development Unit, Infectious Disease Research Department, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| |
Collapse
|
27
|
Barry M, Temsah MH, Alhuzaimi A, Alamro N, Al-Eyadhy A, Aljamaan F, Saddik B, Alhaboob A, Alsohime F, Alhasan K, Alrabiaah A, Alaraj A, Halwani R, Jamal A, Alsubaie S, Al-Shahrani FS, Memish ZA, Al-Tawfiq JA. COVID-19 vaccine confidence and hesitancy among health care workers: A cross-sectional survey from a MERS-CoV experienced nation. PLoS One 2021; 16:e0244415. [PMID: 34843462 PMCID: PMC8629228 DOI: 10.1371/journal.pone.0244415] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 10/13/2021] [Indexed: 12/21/2022] Open
Abstract
Objectives This study aimed to identify coronavirus disease 2019 (COVID-19) vaccine perception, acceptance, confidence, hesitancy, and barriers among health care workers (HCWs). Methods An online national cross-sectional pilot-validated questionnaire was self-administered by HCWs in Saudi Arabia, which is a nation with MERS-CoV experience. The main outcome variable was HCWs’ acceptance of COVID-19 vaccine candidates. The factors associated with vaccination acceptance were identified through a logistic regression analysis, and the level of anxiety was measured using a validated instrument to measure general anxiety levels. Results Out of the 1512 HCWs who completed the study questionnaire—of which 62.4% were women—70% were willing to receive COVID-19 vaccines. A logistic regression analysis revealed that male HCWs (ORa = 1.551, 95% CI: 1.122–2.144), HCWs who believe in vaccine safety (ORa = 2.151; 95% CI: 1.708–2.708), HCWs who believe that COVID vaccines are the most likely way to stop the pandemic (ORa = 1.539; 95% CI: 1.259–1.881), and HCWs who rely on the Centers for Disease Control and Prevention website for COVID 19 updates (ORa = 1.505, 95% CI: 1.125–2.013) were significantly associated with reporting a willingness to be vaccinated. However, HCWs who believed that the vaccines were rushed without evidence-informed testing were found to be 60% less inclined to accept COVID-19 vaccines (ORa = 0.394, 95% CI: 0.298–0.522). Conclusion Most HCWs are willing to receive COVID-19 vaccines once they are available; the satisfactoriness of COVID-19 vaccination among HCWs is crucial because health professionals’ knowledge and confidence toward vaccines are important determining factors for not only their own vaccine acceptance but also recommendation for such vaccines to their patients.
Collapse
Affiliation(s)
- Mazin Barry
- Division of Infectious Diseases, Department of Internal Medicine, College of Medicine, King Saud University and King Saud University Medical City, Riyadh, Saudi Arabia
- * E-mail:
| | - Mohamad-Hani Temsah
- Pediatric Department, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Abdullah Alhuzaimi
- College of Medicine, King Saud University, Riyadh, Saudi Arabia
- Division of Pediatric Cardiology, Cardiac Science Department, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Nurah Alamro
- College of Medicine, King Saud University, Riyadh, Saudi Arabia
- Department of Family and Community Medicine, King Saud University Medical City, Riyadh, Saudi Arabia
| | - Ayman Al-Eyadhy
- Pediatric Department, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Fadi Aljamaan
- College of Medicine, King Saud University, Riyadh, Saudi Arabia
- Critical Care Dept, College of Medicine, King Saud University, Riyadh, Saudi Arabia
- Dr. Sulaiman Al Habib Medical Group, Riyadh, Saudi Arabia
| | - Basema Saddik
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Ali Alhaboob
- Pediatric Department, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Fahad Alsohime
- Pediatric Department, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Khalid Alhasan
- Pediatric Department, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Abdulkarim Alrabiaah
- Pediatric Department, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Ali Alaraj
- Dr. Sulaiman Al Habib Medical Group, Riyadh, Saudi Arabia
- Department of Medicine, College of Medicine, Qassim University, Qassim, Saudi Arabia
| | - Rabih Halwani
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Amr Jamal
- College of Medicine, King Saud University, Riyadh, Saudi Arabia
- Department of Family and Community Medicine, King Saud University Medical City, Riyadh, Saudi Arabia
| | - Sarah Alsubaie
- Pediatric Department, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Fatimah S. Al-Shahrani
- Division of Infectious Diseases, Department of Internal Medicine, College of Medicine, King Saud University and King Saud University Medical City, Riyadh, Saudi Arabia
| | - Ziad A. Memish
- Director Research and Innovation Centre, King Saud Medical City, Ministry of Health & College of Medicine, Alfaisal University, Riyadh, Kingdom of Saudi Arabia
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States of America
| | - Jaffar A. Al-Tawfiq
- Specialty Internal Medicine and Quality Department, Johns Hopkins Aramco Health Care, Dhahran, Saudi Arabia
- Infectious disease division, Department of Medicine, Indiana University School of Medicine, Indiana, United States of America
- Infectious Disease Division, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| |
Collapse
|
28
|
The Combined Expression of the Non-structural Protein NS1 and the N-Terminal Half of NS2 (NS2 1-180) by ChAdOx1 and MVA Confers Protection against Clinical Disease in Sheep upon Bluetongue Virus Challenge. J Virol 2021; 96:e0161421. [PMID: 34787454 PMCID: PMC8826911 DOI: 10.1128/jvi.01614-21] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Bluetongue, caused by bluetongue virus (BTV), is a widespread arthropod-borne disease of ruminants that entails a recurrent threat to the primary sector of developed and developing countries. In this work, we report modified vaccinia virus Ankara (MVA) and ChAdOx1-vectored vaccines designed to simultaneously express the immunogenic NS1 protein and/or NS2-Nt, the N-terminal half of protein NS2 (NS21-180). A single dose of MVA or ChAdOx1 expressing NS1-NS2-Nt improved the protection conferred by NS1 alone in IFNAR(-/-) mice. Moreover, mice immunized with ChAdOx1/MVA-NS1, ChAdOx1/MVA-NS2-Nt, or ChAdOx1/MVA-NS1-NS2-Nt developed strong cytotoxic CD8+ T-cell responses against NS1, NS2-Nt, or both proteins and were fully protected against a lethal infection with BTV serotypes 1, 4, and 8. Furthermore, although a single immunization with ChAdOx1-NS1-NS2-Nt partially protected sheep against BTV-4, the administration of a booster dose of MVA-NS1-NS2-Nt promoted a faster viral clearance, reduction of the period and level of viremia and also protected from the pathology produced by BTV infection. IMPORTANCE Current BTV vaccines are effective but they do not allow to distinguish between vaccinated and infected animals (DIVA strategy) and are serotype specific. In this work we have develop a DIVA multiserotype vaccination strategy based on adenoviral (ChAdOx1) and MVA vaccine vectors, the most widely used in current phase I and II clinical trials, and the conserved nonstructural BTV proteins NS1 and NS2. This immunization strategy solves the major drawbacks of the current marketed vaccines.
Collapse
|
29
|
Elkashif A, Alhashimi M, Sayedahmed EE, Sambhara S, Mittal SK. Adenoviral vector-based platforms for developing effective vaccines to combat respiratory viral infections. Clin Transl Immunology 2021; 10:e1345. [PMID: 34667600 PMCID: PMC8510854 DOI: 10.1002/cti2.1345] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 09/10/2021] [Accepted: 09/15/2021] [Indexed: 02/06/2023] Open
Abstract
Since the development of the first vaccine against smallpox over two centuries ago, vaccination strategies have been at the forefront of significantly impacting the incidences of infectious diseases globally. However, the increase in the human population, deforestation and climate change, and the rise in worldwide travel have favored the emergence of new viruses with the potential to cause pandemics. The ongoing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic is a cruel reminder of the impact of novel pathogens and the suboptimal capabilities of conventional vaccines. Therefore, there is an urgent need to develop new vaccine strategies that allow the production of billions of doses in a short duration and are broadly protective against emerging and re-emerging infectious diseases. Extensive knowledge of the molecular biology and immunology of adenoviruses (Ad) has favored Ad vectors as platforms for vaccine design. The Ad-based vaccine platform represents an attractive strategy as it induces robust humoral and cell-mediated immune responses and can meet the global demand in a pandemic situation. This review describes the status of Ad vector-based vaccines in preclinical and clinical studies for current and emerging respiratory viruses, particularly coronaviruses, influenza viruses and respiratory syncytial viruses.
Collapse
Affiliation(s)
- Ahmed Elkashif
- Department of Comparative PathobiologyPurdue Institute for Inflammation, Immunology and Infectious Disease, and Purdue University Center for Cancer ResearchCollege of Veterinary MedicinePurdue UniversityWest LafayetteINUSA
| | - Marwa Alhashimi
- Department of Comparative PathobiologyPurdue Institute for Inflammation, Immunology and Infectious Disease, and Purdue University Center for Cancer ResearchCollege of Veterinary MedicinePurdue UniversityWest LafayetteINUSA
| | - Ekramy E Sayedahmed
- Department of Comparative PathobiologyPurdue Institute for Inflammation, Immunology and Infectious Disease, and Purdue University Center for Cancer ResearchCollege of Veterinary MedicinePurdue UniversityWest LafayetteINUSA
| | | | - Suresh K Mittal
- Department of Comparative PathobiologyPurdue Institute for Inflammation, Immunology and Infectious Disease, and Purdue University Center for Cancer ResearchCollege of Veterinary MedicinePurdue UniversityWest LafayetteINUSA
| |
Collapse
|
30
|
Günl F, Mecate-Zambrano A, Rehländer S, Hinse S, Ludwig S, Brunotte L. Shooting at a Moving Target-Effectiveness and Emerging Challenges for SARS-CoV-2 Vaccine Development. Vaccines (Basel) 2021; 9:1052. [PMID: 34696160 PMCID: PMC8540924 DOI: 10.3390/vaccines9101052] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/17/2021] [Accepted: 09/18/2021] [Indexed: 02/06/2023] Open
Abstract
Since late 2019 the newly emerged pandemic SARS-CoV-2, the causative agent of COVID-19, has hit the world with recurring waves of infections necessitating the global implementation of non-pharmaceutical interventions, including strict social distancing rules, the wearing of masks and the isolation of infected individuals in order to restrict virus transmissions and prevent the breakdown of our healthcare systems. These measures are not only challenging on an economic level but also have a strong impact on social lifestyles. Using traditional and novel technologies, highly efficient vaccines against SARS-CoV-2 were developed and underwent rapid clinical evaluation and approval to accelerate the immunization of the world population, aiming to end the pandemic and return to normality. However, the emergence of virus variants with improved transmission, enhanced fitness and partial immune escape from the first generation of vaccines poses new challenges, which are currently being addressed by scientists and pharmaceutical companies all over the world. In this ongoing pandemic, the evaluation of SARS-CoV-2 vaccines underlies diverse unpredictable dynamics, posed by the first broad application of the mRNA vaccine technology and their compliance, the occurrence of unexpected side effects and the rapid emergence of variations in the viral antigen. However, despite these hurdles, we conclude that the available SARS-CoV-2 vaccines are very safe and efficiently protect from severe COVID-19 and are thereby the most powerful tools to prevent further harm to our healthcare systems, economics and individual lives. This review summarizes the unprecedented pathways of vaccine development and approval during the ongoing SARS-CoV-2 pandemic. We focus on the real-world effectiveness and unexpected positive and negative side effects of the available vaccines and summarize the timeline of the applied adaptations to the recommended vaccination strategies in the light of emerging virus variants. Finally, we highlight upcoming strategies to improve the next generations of SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
- Franziska Günl
- Institute of Virology (IVM), University of Münster, 48149 Münster, Germany; (F.G.); (A.M.-Z.); (S.R.); (S.H.); (S.L.)
| | - Angeles Mecate-Zambrano
- Institute of Virology (IVM), University of Münster, 48149 Münster, Germany; (F.G.); (A.M.-Z.); (S.R.); (S.H.); (S.L.)
- Interdisciplinary Centre for Clinical Research (IZKF), Medical Faculty, University of Münster, 48149 Münster, Germany
| | - Selina Rehländer
- Institute of Virology (IVM), University of Münster, 48149 Münster, Germany; (F.G.); (A.M.-Z.); (S.R.); (S.H.); (S.L.)
| | - Saskia Hinse
- Institute of Virology (IVM), University of Münster, 48149 Münster, Germany; (F.G.); (A.M.-Z.); (S.R.); (S.H.); (S.L.)
| | - Stephan Ludwig
- Institute of Virology (IVM), University of Münster, 48149 Münster, Germany; (F.G.); (A.M.-Z.); (S.R.); (S.H.); (S.L.)
- Interdisciplinary Centre for Clinical Research (IZKF), Medical Faculty, University of Münster, 48149 Münster, Germany
| | - Linda Brunotte
- Institute of Virology (IVM), University of Münster, 48149 Münster, Germany; (F.G.); (A.M.-Z.); (S.R.); (S.H.); (S.L.)
- Interdisciplinary Centre for Clinical Research (IZKF), Medical Faculty, University of Münster, 48149 Münster, Germany
| |
Collapse
|
31
|
Holloway P, Gibson M, van Doremalen N, Nash S, Holloway T, Letko M, Cardwell JM, Al Omari B, Al-Majali A, Abu-Basha E, Mangtani P, Munster VJ, Guitian J. Risk Factors for Middle East Respiratory Syndrome Coronavirus Infection among Camel Populations, Southern Jordan, 2014-2018. Emerg Infect Dis 2021; 27:2301-2311. [PMID: 34423762 PMCID: PMC8386791 DOI: 10.3201/eid2709.203508] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
After the first detection of Middle East respiratory syndrome coronavirus (MERS-CoV) in camels in Jordan in 2013, we conducted 2 consecutive surveys in 2014–2015 and 2017–2018 investigating risk factors for MERS-CoV infection among camel populations in southern Jordan. Multivariate analysis to control for confounding demonstrated that borrowing of camels, particularly males, for breeding purposes was associated with increased MERS-CoV seroprevalence among receiving herds, suggesting a potential route of viral transmission between herds. Increasing age, herd size, and use of water troughs within herds were also associated with increased seroprevalence. Closed herd management practices were found to be protective. Future vaccination strategies among camel populations in Jordan could potentially prioritize breeding males, which are likely to be shared between herds. In addition, targeted management interventions with the potential to reduce transmission between herds should be considered; voluntary closed herd schemes offer a possible route to achieving disease-free herds.
Collapse
|
32
|
Transcriptomic Profiling of Dromedary Camels Immunised with a MERS Vaccine Candidate. Vet Sci 2021; 8:vetsci8080156. [PMID: 34437478 PMCID: PMC8402689 DOI: 10.3390/vetsci8080156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 11/17/2022] Open
Abstract
Middle East Respiratory Syndrome coronavirus (MERS-CoV) infects dromedary camels and zoonotically infects humans, causing a respiratory disease with severe pneumonia and death. With no approved antiviral or vaccine interventions for MERS, vaccines are being developed for camels to prevent virus transmission into humans. We have previously developed a chimpanzee adenoviral vector-based vaccine for MERS-CoV (ChAdOx1 MERS) and reported its strong humoral immunogenicity in dromedary camels. Here, we looked back at total RNA isolated from whole blood of three immunised dromedaries pre and post-vaccination during the first day; and performed RNA sequencing and bioinformatic analysis in order to shed light on the molecular immune responses following a ChAdOx1 MERS vaccination. Our finding shows that a number of transcripts were differentially regulated as an effect of the vaccination, including genes that are involved in innate and adaptive immunity, such as type I and II interferon responses. The camel Bcl-3 and Bcl-6 transcripts were significantly upregulated, indicating a strong activation of Tfh cell, B cell, and NF-κB pathways. In conclusion, this study gives an overall view of the first changes in the immune transcriptome of dromedaries after vaccination; it supports the potency of ChAdOx1 MERS as a potential camel vaccine to block transmission and prevent new human cases and outbreaks.
Collapse
|
33
|
Amplicon and Metagenomic Analysis of Middle East Respiratory Syndrome (MERS) Coronavirus and the Microbiome in Patients with Severe MERS. mSphere 2021; 6:e0021921. [PMID: 34287009 PMCID: PMC8386452 DOI: 10.1128/msphere.00219-21] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Middle East respiratory syndrome coronavirus (MERS-CoV) is a zoonotic infection that emerged in the Middle East in 2012. Symptoms range from mild to severe and include both respiratory and gastrointestinal illnesses. The virus is mainly present in camel populations with occasional zoonotic spill over into humans. The severity of infection in humans is influenced by numerous factors, and similar to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), underlying health complications can play a major role. Currently, MERS-CoV and SARS-CoV-2 are coincident in the Middle East and thus a rapid way of sequencing MERS-CoV to derive genotype information for molecular epidemiology is needed. Additionally, complicating factors in MERS-CoV infections are coinfections that require clinical management. The ability to rapidly characterize these infections would be advantageous. To rapidly sequence MERS-CoV, an amplicon-based approach was developed and coupled to Oxford Nanopore long read length sequencing. This and a metagenomic approach were evaluated with clinical samples from patients with MERS. The data illustrated that whole-genome or near-whole-genome information on MERS-CoV could be rapidly obtained. This approach provided data on both consensus genomes and the presence of minor variants, including deletion mutants. The metagenomic analysis provided information of the background microbiome. The advantage of this approach is that insertions and deletions can be identified, which are the major drivers of genotype change in coronaviruses. IMPORTANCE Middle East respiratory syndrome coronavirus (MERS-CoV) emerged in late 2012 in Saudi Arabia. The virus is a serious threat to people not only in the Middle East but also in the world and has been detected in over 27 countries. MERS-CoV is spreading in the Middle East and neighboring countries, and approximately 35% of reported patients with this virus have died. This is the most severe coronavirus infection so far described. Saudi Arabia is a destination for many millions of people in the world who visit for religious purposes (Umrah and Hajj), and so it is a very vulnerable area, which imposes unique challenges for effective control of this epidemic. The significance of our study is that clinical samples from patients with MERS were used for rapid in-depth sequencing and metagenomic analysis using long read length sequencing.
Collapse
|
34
|
Jo WK, de Oliveira‐Filho EF, Rasche A, Greenwood AD, Osterrieder K, Drexler JF. Potential zoonotic sources of SARS-CoV-2 infections. Transbound Emerg Dis 2021; 68:1824-1834. [PMID: 33034151 PMCID: PMC7675418 DOI: 10.1111/tbed.13872] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 10/02/2020] [Accepted: 10/03/2020] [Indexed: 12/18/2022]
Abstract
The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) causing coronavirus disease-2019 (COVID-19) likely has evolutionary origins in other animals than humans based on genetically related viruses existing in rhinolophid bats and pangolins. Similar to other animal coronaviruses, SARS-CoV-2 contains a functional furin cleavage site in its spike protein, which may broaden the SARS-CoV-2 host range and affect pathogenesis. Whether ongoing zoonotic infections are possible in addition to efficient human-to-human transmission remains unclear. In contrast, human-to-animal transmission can occur based on evidence provided from natural and experimental settings. Carnivores, including domestic cats, ferrets and minks, appear to be particularly susceptible to SARS-CoV-2 in contrast to poultry and other animals reared as livestock such as cattle and swine. Epidemiologic evidence supported by genomic sequencing corroborated mink-to-human transmission events in farm settings. Airborne transmission of SARS-CoV-2 between experimentally infected cats additionally substantiates the possibility of cat-to-human transmission. To evaluate the COVID-19 risk represented by domestic and farmed carnivores, experimental assessments should include surveillance and health assessment of domestic and farmed carnivores, characterization of the immune interplay between SARS-CoV-2 and carnivore coronaviruses, determination of the SARS-CoV-2 host range beyond carnivores and identification of human risk groups such as veterinarians and farm workers. Strategies to mitigate the risk of zoonotic SARS-CoV-2 infections may have to be developed in a One Health framework and non-pharmaceutical interventions may have to consider free-roaming animals and the animal farming industry.
Collapse
Affiliation(s)
- Wendy K. Jo
- Institute of Virology, Charité‐Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
| | - Edmilson Ferreira de Oliveira‐Filho
- Institute of Virology, Charité‐Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
| | - Andrea Rasche
- Institute of Virology, Charité‐Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
- German Centre for Infection Research (DZIF)Associated Partner Charité‐Universitätsmedizin BerlinBerlinGermany
| | - Alex D. Greenwood
- Leibniz Institute for Zoo and Wildlife ResearchBerlinGermany
- Institut für VirologieFreie Universität BerlinBerlinGermany
| | | | - Jan Felix Drexler
- Institute of Virology, Charité‐Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
- German Centre for Infection Research (DZIF)Associated Partner Charité‐Universitätsmedizin BerlinBerlinGermany
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector‐Borne DiseasesSechenov UniversityMoscowRussia
| |
Collapse
|
35
|
Immunotherapeutic Efficacy of IgY Antibodies Targeting the Full-Length Spike Protein in an Animal Model of Middle East Respiratory Syndrome Coronavirus Infection. PHARMACEUTICALS (BASEL, SWITZERLAND) 2021. [PMID: 34073502 DOI: 10.3390/ph14060511.] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Identified in 2012, the Middle East respiratory syndrome coronavirus (MERS-CoV) causes severe and often fatal acute respiratory illness in humans. No approved prophylactic or therapeutic interventions are currently available. In this study, we developed chicken egg yolk antibodies (IgY Abs) specific to the MERS-CoV spike (S) protein and evaluated their neutralizing efficiency against MERS-CoV infection. S-specific IgY Abs were produced by injecting chickens with the purified recombinant S protein of MERS-CoV at a high titer (4.4 mg/mL per egg yolk) at week 7 post immunization. Western blotting and immune-dot blot assays demonstrated specific binding to the MERS-CoV S protein. In vitro neutralization of the generated IgY Abs against MERS-CoV was evaluated and showed a 50% neutralizing concentration of 51.42 μg/mL. In vivo testing using a human-transgenic mouse model showed a reduction of viral antigen positive cells in treated mice, compared to the adjuvant-only controls. Moreover, the lung cells of the treated mice showed significantly reduced inflammation, compared to the controls. Our results show efficient neutralization of MERS-CoV infection both in vitro and in vivo using S-specific IgY Abs. Clinical trials are needed to evaluate the efficiency of the IgY Abs in camels and humans.
Collapse
|
36
|
El-Kafrawy SA, Abbas AT, Sohrab SS, Tabll AA, Hassan AM, Iwata-Yoshikawa N, Nagata N, Azhar EI. Immunotherapeutic Efficacy of IgY Antibodies Targeting the Full-Length Spike Protein in an Animal Model of Middle East Respiratory Syndrome Coronavirus Infection. Pharmaceuticals (Basel) 2021; 14:ph14060511. [PMID: 34073502 PMCID: PMC8229159 DOI: 10.3390/ph14060511] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 05/19/2021] [Accepted: 05/21/2021] [Indexed: 02/07/2023] Open
Abstract
Identified in 2012, the Middle East respiratory syndrome coronavirus (MERS-CoV) causes severe and often fatal acute respiratory illness in humans. No approved prophylactic or therapeutic interventions are currently available. In this study, we developed chicken egg yolk antibodies (IgY Abs) specific to the MERS-CoV spike (S) protein and evaluated their neutralizing efficiency against MERS-CoV infection. S-specific IgY Abs were produced by injecting chickens with the purified recombinant S protein of MERS-CoV at a high titer (4.4 mg/mL per egg yolk) at week 7 post immunization. Western blotting and immune-dot blot assays demonstrated specific binding to the MERS-CoV S protein. In vitro neutralization of the generated IgY Abs against MERS-CoV was evaluated and showed a 50% neutralizing concentration of 51.42 μg/mL. In vivo testing using a human-transgenic mouse model showed a reduction of viral antigen positive cells in treated mice, compared to the adjuvant-only controls. Moreover, the lung cells of the treated mice showed significantly reduced inflammation, compared to the controls. Our results show efficient neutralization of MERS-CoV infection both in vitro and in vivo using S-specific IgY Abs. Clinical trials are needed to evaluate the efficiency of the IgY Abs in camels and humans.
Collapse
Affiliation(s)
- Sherif A. El-Kafrawy
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (S.A.E.-K.); (S.S.S.); (A.M.H.)
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Aymn T. Abbas
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (S.A.E.-K.); (S.S.S.); (A.M.H.)
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Biotechnology Research Laboratories, Gastroenterology, Surgery Centre, Mansoura University, Mansoura 35516, Egypt
- Correspondence: (A.T.A.); (E.I.A.); Tel.: +966-546-315-514 (A.T.A.); +966-566-615-222 (E.I.A.)
| | - Sayed S. Sohrab
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (S.A.E.-K.); (S.S.S.); (A.M.H.)
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ashraf A. Tabll
- Microbial Biotechnology Department, Genetic Engineering and Biotechnology Division, National Research Centre, Dokki 12622, Egypt;
- Department of Immunology, Egypt Center for Research and Regenerative Medicine (ECRRM), Cairo 11517, Egypt
| | - Ahmed M. Hassan
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (S.A.E.-K.); (S.S.S.); (A.M.H.)
| | - Naoko Iwata-Yoshikawa
- Department of Pathology, National Institute of Infectious Diseases, Tokyo 208-0011, Japan; (N.I.-Y.); (N.N.)
| | - Noriyo Nagata
- Department of Pathology, National Institute of Infectious Diseases, Tokyo 208-0011, Japan; (N.I.-Y.); (N.N.)
| | - Esam I. Azhar
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (S.A.E.-K.); (S.S.S.); (A.M.H.)
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Correspondence: (A.T.A.); (E.I.A.); Tel.: +966-546-315-514 (A.T.A.); +966-566-615-222 (E.I.A.)
| |
Collapse
|
37
|
Costanzo M, De Giglio MAR, Roviello GN. Anti-Coronavirus Vaccines: Past Investigations on SARS-CoV-1 and MERS-CoV, the Approved Vaccines from BioNTech/Pfizer, Moderna, Oxford/AstraZeneca and others under Development Against SARS-CoV-2 Infection. Curr Med Chem 2021; 29:4-18. [PMID: 34355678 DOI: 10.2174/0929867328666210521164809] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/07/2021] [Accepted: 04/10/2021] [Indexed: 11/22/2022]
Abstract
The aim of this review article is to summarize the knowledge available to date on prophylaxis achievements to fight against Coronavirus. This work will give an overview of what is reported in the most recent literature on vaccines (under investigation or already developed like BNT162b2, mRNA-1273, and ChAdOx1-S) effective against the most pathogenic Coronaviruses (SARS-CoV-1, MERS-CoV-1, and SARS-CoV-2), with of course particular attention paid to those under development or already in use to combat the current COVID-19 (COronaVIrus Disease 19) pandemic. Our main objective is to make a contribution to the comprehension, additionally at a molecular level, of what is currently ready for anti-SARS-CoV-2 prophylactic intervention, as well as to provide the reader with an overall picture of the most innovative approaches for the development of vaccines that could be of general utility in the fight against the most pathogenic Coronaviruses.
Collapse
Affiliation(s)
- Michele Costanzo
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples 'Federico II', Via S. Pansini 5, I-80131 Naples, Italy
| | | | - Giovanni N Roviello
- Istituto di Biostrutture e Bioimmagini IBB - CNR, Via Mezzocannone 16; I-80134 Naples, Italy
| |
Collapse
|
38
|
Marsh GA, McAuley AJ, Au GG, Riddell S, Layton D, Singanallur NB, Layton R, Payne J, Durr PA, Bender H, Barr JA, Bingham J, Boyd V, Brown S, Bruce MP, Burkett K, Eastwood T, Edwards S, Gough T, Halpin K, Harper J, Holmes C, Horman WSJ, van Vuren PJ, Lowther S, Maynard K, McAuley KD, Neave MJ, Poole T, Rootes C, Rowe B, Soldani E, Stevens V, Stewart CR, Suen WW, Tachedjian M, Todd S, Trinidad L, Walter D, Watson N, Drew TW, Gilbert SC, Lambe T, Vasan SS. ChAdOx1 nCoV-19 (AZD1222) vaccine candidate significantly reduces SARS-CoV-2 shedding in ferrets. NPJ Vaccines 2021; 6:67. [PMID: 33972565 PMCID: PMC8110954 DOI: 10.1038/s41541-021-00315-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/12/2021] [Indexed: 01/05/2023] Open
Abstract
Vaccines against SARS-CoV-2 are likely to be critical in the management of the ongoing pandemic. A number of candidates are in Phase III human clinical trials, including ChAdOx1 nCoV-19 (AZD1222), a replication-deficient chimpanzee adenovirus-vectored vaccine candidate. In preclinical trials, the efficacy of ChAdOx1 nCoV-19 against SARS-CoV-2 challenge was evaluated in a ferret model of infection. Groups of ferrets received either prime-only or prime-boost administration of ChAdOx1 nCoV-19 via the intramuscular or intranasal route. All ChAdOx1 nCoV-19 administration combinations resulted in significant reductions in viral loads in nasal-wash and oral swab samples. No vaccine-associated adverse events were observed associated with the ChAdOx1 nCoV-19 candidate, with the data from this study suggesting it could be an effective and safe vaccine against COVID-19. Our study also indicates the potential for intranasal administration as a way to further improve the efficacy of this leading vaccine candidate.
Collapse
Affiliation(s)
- Glenn A Marsh
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | | | - Gough G Au
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Sarah Riddell
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Daniel Layton
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | | | - Rachel Layton
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Jean Payne
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Peter A Durr
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Hannah Bender
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Jennifer A Barr
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - John Bingham
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Victoria Boyd
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Sheree Brown
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Matthew P Bruce
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Kathie Burkett
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Teresa Eastwood
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Sarah Edwards
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Tamara Gough
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Kim Halpin
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Jenni Harper
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Clare Holmes
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - William S J Horman
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | | | - Suzanne Lowther
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Kate Maynard
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Kristen D McAuley
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Matthew J Neave
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Timothy Poole
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Christina Rootes
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Brenton Rowe
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Elisha Soldani
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Vittoria Stevens
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Cameron R Stewart
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Willy W Suen
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Mary Tachedjian
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Shawn Todd
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Lee Trinidad
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Duane Walter
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Naomi Watson
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Trevor W Drew
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Sarah C Gilbert
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Teresa Lambe
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - S S Vasan
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC, Australia.
- Department of Health Sciences, University of York, York, UK.
| |
Collapse
|
39
|
Toon K, Bentley EM, Mattiuzzo G. More Than Just Gene Therapy Vectors: Lentiviral Vector Pseudotypes for Serological Investigation. Viruses 2021; 13:217. [PMID: 33572589 PMCID: PMC7911487 DOI: 10.3390/v13020217] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/22/2021] [Accepted: 01/26/2021] [Indexed: 12/13/2022] Open
Abstract
Serological assays detecting neutralising antibodies are important for determining the immune responses following infection or vaccination and are also often considered a correlate of protection. The target of neutralising antibodies is usually located in the Envelope protein on the viral surface, which mediates cell entry. As such, presentation of the Envelope protein on a lentiviral particle represents a convenient alternative to handling of a potentially high containment virus or for those viruses with no established cell culture system. The flexibility, relative safety and, in most cases, ease of production of lentiviral pseudotypes, have led to their use in serological assays for many applications such as the evaluation of candidate vaccines, screening and characterization of anti-viral therapeutics, and sero-surveillance. Above all, the speed of production of the lentiviral pseudotypes, once the envelope sequence is published, makes them important tools in the response to viral outbreaks, as shown during the COVID-19 pandemic in 2020. In this review, we provide an overview of the landscape of the serological applications of pseudotyped lentiviral vectors, with a brief discussion on their production and batch quality analysis. Finally, we evaluate their role as surrogates for the real virus and possible alternatives.
Collapse
Affiliation(s)
- Kamilla Toon
- Division of Virology, National Institute for Biological Standards and Control-MHRA, Blanche Lane, South Mimms EN6 3QG, UK;
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK
| | - Emma M. Bentley
- Division of Virology, National Institute for Biological Standards and Control-MHRA, Blanche Lane, South Mimms EN6 3QG, UK;
| | - Giada Mattiuzzo
- Division of Virology, National Institute for Biological Standards and Control-MHRA, Blanche Lane, South Mimms EN6 3QG, UK;
| |
Collapse
|
40
|
Liu CH, Huang HY, Tu YF, Lai WY, Wang CL, Sun JR, Chien Y, Lin TW, Lin YY, Chien CS, Huang CH, Chen YM, Huang PI, Wang FD, Yang YP. Highlight of severe acute respiratory syndrome coronavirus-2 vaccine development against COVID-19 pandemic. J Chin Med Assoc 2021; 84:9-13. [PMID: 33186212 DOI: 10.1097/jcma.0000000000000461] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The pandemic caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has brought an unprecedented impact upon the global economy and public health. Although the SARS-CoV-2 virology has been gradually investigated, measures to combat this new threat in public health are still absent. To date, no certificated drug or vaccine has been developed for the treatment or prevention of coronavirus disease Extensive researches and international coordination has been conducted to rapidly develop novel vaccines against SARS-CoV-2 pandemic. Several major breakthroughs have been made through the identification of the genetic sequence and structural/non-structural proteins of SARS-CoV-2, which enabled the development of RNA-, DNA-based vaccines, subunit vaccines, and attenuated viral vaccines. In this review article, we present an overview of the recent advances of SARS-CoV-2 vaccines and the challenges that may be encountered in the development process, highlighting the advantages and disadvantages of these approaches that may help in effectively countering COVID-19.
Collapse
Affiliation(s)
- Cheng-Hsuan Liu
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- School of Medicine, National Yang-Ming Medical University, Taipei, Taiwan, ROC
| | - Hsuan-Yang Huang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Yung-Fang Tu
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- School of Medicine, National Yang-Ming Medical University, Taipei, Taiwan, ROC
| | - Wei-Yi Lai
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Chia-Lin Wang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Jun-Ren Sun
- Institute of Preventive Medicine, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Yueh Chien
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Tzu-Wei Lin
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Yi-Ying Lin
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Chian-Shiu Chien
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Chih-Heng Huang
- Institute of Preventive Medicine, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Yuh-Min Chen
- School of Medicine, National Yang-Ming Medical University, Taipei, Taiwan, ROC
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Cancer Center, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Pin-I Huang
- School of Medicine, National Yang-Ming Medical University, Taipei, Taiwan, ROC
- Division of Radiation Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Fu-Der Wang
- School of Medicine, National Yang-Ming Medical University, Taipei, Taiwan, ROC
- Division of Infectious Diseases, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Yi-Ping Yang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- School of Medicine, National Yang-Ming Medical University, Taipei, Taiwan, ROC
| |
Collapse
|
41
|
Viral Vector Vaccines against Bluetongue Virus. Microorganisms 2020; 9:microorganisms9010042. [PMID: 33375723 PMCID: PMC7823852 DOI: 10.3390/microorganisms9010042] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/18/2020] [Accepted: 12/22/2020] [Indexed: 12/20/2022] Open
Abstract
Bluetongue virus (BTV), the prototype member of the genus Orbivirus (family Reoviridae), is the causative agent of an important livestock disease, bluetongue (BT), which is transmitted via biting midges of the genus Culicoides. To date, up to 29 serotypes of BTV have been described, which are classified as classical (BTV 1–24) or atypical (serotypes 25–27), and its distribution has been expanding since 1998, with important outbreaks in the Mediterranean Basin and devastating incursions in Northern and Western Europe. Classical vaccine approaches, such as live-attenuated and inactivated vaccines, have been used as prophylactic measures to control BT through the years. However, these vaccine approaches fail to address important matters like vaccine safety profile, effectiveness, induction of a cross-protective immune response among serotypes, and implementation of a DIVA (differentiation of infected from vaccinated animals) strategy. In this context, a wide range of recombinant vaccine prototypes against BTV, ranging from subunit vaccines to recombinant viral vector vaccines, have been investigated. This article offers a comprehensive outline of the live viral vectors used against BTV.
Collapse
|
42
|
Zhao J, Zhao S, Ou J, Zhang J, Lan W, Guan W, Wu X, Yan Y, Zhao W, Wu J, Chodosh J, Zhang Q. COVID-19: Coronavirus Vaccine Development Updates. Front Immunol 2020; 11:602256. [PMID: 33424848 PMCID: PMC7785583 DOI: 10.3389/fimmu.2020.602256] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 11/26/2020] [Indexed: 12/27/2022] Open
Abstract
Coronavirus Disease 2019 (COVID-19) is caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), a newly emerged coronavirus, and has been pandemic since March 2020 and led to many fatalities. Vaccines represent the most efficient means to control and stop the pandemic of COVID-19. However, currently there is no effective COVID-19 vaccine approved to use worldwide except for two human adenovirus vector vaccines, three inactivated vaccines, and one peptide vaccine for early or limited use in China and Russia. Safe and effective vaccines against COVID-19 are in urgent need. Researchers around the world are developing 213 COVID-19 candidate vaccines, among which 44 are in human trials. In this review, we summarize and analyze vaccine progress against SARS-CoV, Middle-East respiratory syndrome Coronavirus (MERS-CoV), and SARS-CoV-2, including inactivated vaccines, live attenuated vaccines, subunit vaccines, virus like particles, nucleic acid vaccines, and viral vector vaccines. As SARS-CoV-2, SARS-CoV, and MERS-CoV share the common genus, Betacoronavirus, this review of the major research progress will provide a reference and new insights into the COVID-19 vaccine design and development.
Collapse
Affiliation(s)
- Jing Zhao
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Shan Zhao
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Junxian Ou
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jing Zhang
- Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Wendong Lan
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Wenyi Guan
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xiaowei Wu
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yuqian Yan
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Wei Zhao
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jianguo Wu
- Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - James Chodosh
- Department of Ophthalmology, Howe Laboratory, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, United States
| | - Qiwei Zhang
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| |
Collapse
|
43
|
Li YD, Chi WY, Su JH, Ferrall L, Hung CF, Wu TC. Coronavirus vaccine development: from SARS and MERS to COVID-19. J Biomed Sci 2020; 27:104. [PMID: 33341119 PMCID: PMC7749790 DOI: 10.1186/s12929-020-00695-2] [Citation(s) in RCA: 225] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 11/20/2020] [Indexed: 02/08/2023] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is a new type of coronavirus that causes the Coronavirus Disease 2019 (COVID-19), which has been the most challenging pandemic in this century. Considering its high mortality and rapid spread, an effective vaccine is urgently needed to control this pandemic. As a result, the academia, industry, and government sectors are working tightly together to develop and test a variety of vaccines at an unprecedented pace. In this review, we outline the essential coronavirus biological characteristics that are important for vaccine design. In addition, we summarize key takeaways from previous vaccination studies of Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV) and Middle East Respiratory Syndrome Coronavirus (MERS-CoV), highlighting the pros and cons of each immunization strategy. Finally, based on these prior vaccination experiences, we discuss recent progress and potential challenges of COVID-19 vaccine development.
Collapse
Affiliation(s)
- Yen-Der Li
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Wei-Yu Chi
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Jun-Han Su
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Louise Ferrall
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Chien-Fu Hung
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - T-C Wu
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA.
- Johns Hopkins School of Medicine, 1550 Orleans St, CRB II - Room 309, Baltimore, MD, 21287, USA.
| |
Collapse
|
44
|
Adney DR, Clancy CS, Bowen RA, Munster VJ. Camelid Inoculation With Middle East Respiratory Syndrome Coronavirus: Experimental Models of Reservoir Host Infection. Viruses 2020; 12:v12121370. [PMID: 33266124 PMCID: PMC7759921 DOI: 10.3390/v12121370] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/24/2020] [Accepted: 11/25/2020] [Indexed: 12/13/2022] Open
Abstract
Within the past two decades, three zoonotic betacoronaviruses have been associated with outbreaks causing severe respiratory disease in humans. Of these, Middle East respiratory s yndrome coronavirus (MERS-CoV) is the only zoonotic coronavirus that is known to consistently result in frequent zoonotic spillover events from the proximate reservoir host—the dromedary camel. A comprehensive understanding of infection in dromedaries is critical to informing public health recommendations and implementing intervention strategies to mitigate spillover events. Experimental models of reservoir disease are absolutely critical in understanding the pathogenesis and transmission, and are key to testing potential dromedary vaccines against MERS-CoV. In this review, we describe experimental infections of dromedary camels as well as additional camelid models used to further understand the camel’s role in MERS-CoV spillover to humans.
Collapse
Affiliation(s)
- Danielle R. Adney
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA;
| | - Chad S. Clancy
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, Hamilton, MT 59840, USA;
| | - Richard A. Bowen
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80521, USA;
| | - Vincent J. Munster
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA;
- Correspondence: ; Tel.: +406-375-7489
| |
Collapse
|
45
|
Sasso E, D'Alise AM, Zambrano N, Scarselli E, Folgori A, Nicosia A. New viral vectors for infectious diseases and cancer. Semin Immunol 2020; 50:101430. [PMID: 33262065 DOI: 10.1016/j.smim.2020.101430] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 10/23/2020] [Accepted: 11/16/2020] [Indexed: 12/12/2022]
Abstract
Since the discovery in 1796 by Edward Jenner of vaccinia virus as a way to prevent and finally eradicate smallpox, the concept of using a virus to fight another virus has evolved into the current approaches of viral vectored genetic vaccines. In recent years, key improvements to the vaccinia virus leading to a safer version (Modified Vaccinia Ankara, MVA) and the discovery that some viruses can be used as carriers of heterologous genes encoding for pathological antigens of other infectious agents (the concept of 'viral vectors') has spurred a new wave of clinical research potentially providing for a solution for the long sought after vaccines against major diseases such as HIV, TB, RSV and Malaria, or emerging infectious diseases including those caused by filoviruses and coronaviruses. The unique ability of some of these viral vectors to stimulate the cellular arm of the immune response and, most importantly, T lymphocytes with cell killing activity, has also reawakened the interest toward developing therapeutic vaccines against chronic infectious diseases and cancer. To this end, existing vectors such as those based on Adenoviruses have been improved in immunogenicity and efficacy. Along the same line, new vectors that exploit viruses such as Vesicular Stomatitis Virus (VSV), Measles Virus (MV), Lymphocytic choriomeningitis virus (LCMV), cytomegalovirus (CMV), and Herpes Simplex Virus (HSV), have emerged. Furthermore, technological progress toward modifying their genome to render some of these vectors incompetent for replication has increased confidence toward their use in infant and elderly populations. Lastly, their production process being the same for every product has made viral vectored vaccines the technology of choice for rapid development of vaccines against emerging diseases and for 'personalised' cancer vaccines where there is an absolute need to reduce time to the patient from months to weeks or days. Here we review the recent developments in viral vector technologies, focusing on novel vectors based on primate derived Adenoviruses and Poxviruses, Rhabdoviruses, Paramixoviruses, Arenaviruses and Herpesviruses. We describe the rationale for, immunologic mechanisms involved in, and design of viral vectored gene vaccines under development and discuss the potential utility of these novel genetic vaccine approaches in eliciting protection against infectious diseases and cancer.
Collapse
Affiliation(s)
- Emanuele Sasso
- Nouscom srl, Via di Castel Romano 100, 00128 Rome, Italy; Ceinge-Biotecnologie Avanzate S.C. A.R.L., via Gaetano Salvatore 486, 80145 Naples, Italy.
| | | | - Nicola Zambrano
- Ceinge-Biotecnologie Avanzate S.C. A.R.L., via Gaetano Salvatore 486, 80145 Naples, Italy; Department of Molecular Medicine and Medical Biotechnology, University Federico II, Via Pansini 5, 80131 Naples, Italy.
| | | | | | - Alfredo Nicosia
- Ceinge-Biotecnologie Avanzate S.C. A.R.L., via Gaetano Salvatore 486, 80145 Naples, Italy; Department of Molecular Medicine and Medical Biotechnology, University Federico II, Via Pansini 5, 80131 Naples, Italy.
| |
Collapse
|
46
|
Goodnow CC. COVID-19, varying genetic resistance to viral disease and immune tolerance checkpoints. Immunol Cell Biol 2020; 99:177-191. [PMID: 33113212 PMCID: PMC7894315 DOI: 10.1111/imcb.12419] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/25/2020] [Accepted: 10/26/2020] [Indexed: 12/14/2022]
Abstract
Coronavirus disease 2019 (COVID‐19) is a zoonosis like most of the great plagues sculpting human history, from smallpox to pandemic influenza and human immunodeficiency virus. When viruses jump into a new species the outcome of infection ranges from asymptomatic to lethal, historically ascribed to “genetic resistance to viral disease.” People have exploited these differences for good and bad, for developing vaccines from cowpox and horsepox virus, controlling rabbit plagues with myxoma virus and introducing smallpox during colonization of America and Australia. Differences in resistance to viral disease are at the core of the severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) crisis, yet our understanding of the mechanisms in any interspecies leap falls short of the mark. Here I review how the two key parameters of viral disease are countered by fundamentally different genetic mechanisms for resistance: (1) virus transmission, countered primarily by activation of innate and adaptive immune responses; and (2) pathology, countered primarily by tolerance checkpoints to limit innate and adaptive immune responses. I discuss tolerance thresholds and the role of CD8 T cells to limit pathological immune responses, the problems posed by tolerant superspreaders and the signature coronavirus evasion strategy of eliciting only short‐lived neutralizing antibody responses. Pinpointing and targeting the mechanisms responsible for varying pathology and short‐lived antibody were beyond reach in previous zoonoses, but this time we are armed with genomic technologies and more knowledge of immune checkpoint genes. These known unknowns must now be tackled to solve the current COVID‐19 crisis and the inevitable zoonoses to follow.
Collapse
Affiliation(s)
- Christopher C Goodnow
- Garvan Institute of Medical Research, Sydney, NSW, Australia.,Cellular Genomics Futures Institute, UNSW Sydney, Sydney, NSW, Australia
| |
Collapse
|
47
|
Alserehi HA, Alqunaibet AM, Al-Tawfiq JA, Alharbi NK, Alshukairi AN, Alanazi KH, Bin Saleh GM, Alshehri AM, Almasoud A, Hashem AM, Alruwaily AR, Alaswad RH, Al-Mutlaq HM, Almudaiheem AA, Othman FM, Aldakeel SA, Abu Ghararah MR, Jokhdar HA, Algwizani AR, Almudarra SS, Albarrag AM. Seroprevalence of SARS-CoV-2 (COVID-19) among healthcare workers in Saudi Arabia: comparing case and control hospitals. Diagn Microbiol Infect Dis 2020; 99:115273. [PMID: 33296851 PMCID: PMC7677039 DOI: 10.1016/j.diagmicrobio.2020.115273] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 11/08/2020] [Accepted: 11/15/2020] [Indexed: 01/27/2023]
Abstract
Healthcare workers (HCWs) stand at the frontline for fighting coronavirus disease 2019 (COVID-19) pandemic. This puts them at higher risk of acquiring the infection than other individuals in the community. Defining immunity status among health care workers is therefore of interest since it helps to mitigate the exposure risk. This study was conducted between May 20th and 30th, 2020. Eighty-five hospitals across Kingdom of Saudi Arabia were divided into 2 groups: COVID-19 referral hospitals are those to which RT-PCR-confirmed COVID-19 patients were admitted or referred for management (Case-hospitals). COVID-19 nonaffected hospitals where no COVID-19 patients had been admitted or managed and no HCW outbreak (Control hospitals). Next, seroprevalence of severe acute respiratory syndrome coronavirus 2 among HCWs was evaluated; there were 12,621 HCWs from the 85 hospitals. There were 61 case-hospitals with 9379 (74.3%) observations, and 24 control-hospitals with 3242 (25.7%) observations. The overall positivity rate by the immunoassay was 299 (2.36%) with a significant difference between the case-hospital (2.9%) and the control-group (0.8%) (P value <0.001). There was a wide variation in the positivity rate between regions and/or cities in Saudi Arabia, ranging from 0% to 6.31%. Of the serology positive samples, 100 samples were further tested using the SAS2pp neutralization assay; 92 (92%) samples showed neutralization activity. The seropositivity rate in Kingdom of Saudi Arabia is low and varies across different regions with higher positivity in case-hospitals than control-hospitals. The lack of neutralizing antibodies (NAb) in 8% of the tested samples could mean that assay is a more sensitive assay or that neutralization assay has a lower detection limits; or possibly that some samples had cross-reaction to spike protein of other coronaviruses in the assay, but these were not specific to neutralize severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2).
Collapse
Affiliation(s)
- Haleema Ali Alserehi
- Executive Department of Global Health, Epidemiology, surveillance and preparedness affairs, Executive Department of Research, Saudi Center for Disease Prevention and Control, Riyadh, Saudi Arabia.
| | - Ada Mohammed Alqunaibet
- Executive Department of Global Health, Epidemiology, surveillance and preparedness affairs, Executive Department of Research, Saudi Center for Disease Prevention and Control, Riyadh, Saudi Arabia
| | - Jaffar A Al-Tawfiq
- Infectious Disease Unit, Specialty Internal Medicine, and Quality and Patient Safety Departement, Johns Hopkins Aramco Healthcare, Dhahran, Saudi Arabia; Infectious Disease Division, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA; Infectious Disease Division, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Naif Khalaf Alharbi
- Vaccine Development Unit, Department of Infectious Disease Research, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Abeer Nizar Alshukairi
- Department of medicine, King Faisal Specialist Hospital and Research Center, Jeddah, Saudi Arabia
| | | | | | - Amer Mohammed Alshehri
- Public Health Lab, Saudi Center for Disease Prevention and Control, Riyadh, Saudi Arabia
| | - Abdulrahman Almasoud
- Vaccine Development Unit, Department of Infectious Disease Research, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Anwar M Hashem
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia; Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Amaal Rabie Alruwaily
- Executive Department of Global Health, Epidemiology, surveillance and preparedness affairs, Executive Department of Research, Saudi Center for Disease Prevention and Control, Riyadh, Saudi Arabia
| | - Rehab Habeeb Alaswad
- Executive Department of Global Health, Epidemiology, surveillance and preparedness affairs, Executive Department of Research, Saudi Center for Disease Prevention and Control, Riyadh, Saudi Arabia
| | - Hind Mohammed Al-Mutlaq
- Executive Department of Global Health, Epidemiology, surveillance and preparedness affairs, Executive Department of Research, Saudi Center for Disease Prevention and Control, Riyadh, Saudi Arabia
| | - Abdulllah Ali Almudaiheem
- Executive Department of Global Health, Epidemiology, surveillance and preparedness affairs, Executive Department of Research, Saudi Center for Disease Prevention and Control, Riyadh, Saudi Arabia
| | - Fatmah Mahmoud Othman
- Research department, College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | | | | | | | | | - Sami Saeed Almudarra
- Executive Department of Global Health, Epidemiology, surveillance and preparedness affairs, Executive Department of Research, Saudi Center for Disease Prevention and Control, Riyadh, Saudi Arabia
| | - Ahmed Mohammed Albarrag
- Public Health Lab, Saudi Center for Disease Prevention and Control, Riyadh, Saudi Arabia; Department of Pathology, School of Medicine, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
48
|
Mohapatra RK, Pintilie L, Kandi V, Sarangi AK, Das D, Sahu R, Perekhoda L. The recent challenges of highly contagious COVID-19, causing respiratory infections: Symptoms, diagnosis, transmission, possible vaccines, animal models, and immunotherapy. Chem Biol Drug Des 2020. [PMID: 32654267 DOI: 10.1111/cbdd.v96.510.1111/cbdd.13761] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
Abstract
COVID-19 is highly contagious pathogenic viral infection initiated from Wuhan seafood wholesale market of China on December 2019 and spread rapidly around the whole world due to onward transmission. This recent outbreak of novel coronavirus (CoV) was believed to be originated from bats and causing respiratory infections such as common cold, dry cough, fever, headache, dyspnea, pneumonia, and finally Severe Acute Respiratory Syndrome (SARS) in humans. For this widespread zoonotic virus, human-to-human transmission has resulted in nearly 83 lakh cases in 213 countries and territories with 4,50,686 deaths as on 19 June 2020. This review presents a report on the origin, transmission, symptoms, diagnosis, possible vaccines, animal models, and immunotherapy for this novel virus and will provide ample references for the researchers toward the ongoing development of therapeutic agents and vaccines and also preventing the spread of this disease.
Collapse
Affiliation(s)
- Ranjan K Mohapatra
- Department of Chemistry, Government College of Engineering, Keonjhar, Odisha, India
| | - Lucia Pintilie
- Department of Synthesis of Bioactive Substances and Pharmaceutical Technologies, National Institute for Chemical and Pharmaceutical Research and Development, Bucharest, Romania
| | - Venkataramana Kandi
- Department of Microbiology, Pratima Institute of Medical Sciences, Karimnagar, Hyderabad, India
| | - Ashish K Sarangi
- Department of Chemistry, School of Applied Sciences, Centurion University of Technology and Management, Odisha, India
| | - Debadutta Das
- Department of Chemistry, Sukanti Degree College, Subarnapur, Odisha, India
| | - Raghaba Sahu
- College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Lina Perekhoda
- Department of medicinal chemistry, National University of Pharmacy, Kharkiv, Ukraine
| |
Collapse
|
49
|
Chang WH. A review of vaccine effects on women in light of the COVID-19 pandemic. Taiwan J Obstet Gynecol 2020; 59:812-820. [PMID: 33218394 PMCID: PMC7486065 DOI: 10.1016/j.tjog.2020.09.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2020] [Indexed: 11/13/2022] Open
Abstract
The pandemic situation triggered by the spread of COVID-19 has caused great harm worldwide. More than six million people have been infected, and more than 360,000 of them have died. This is the worst catastrophe suffered by mankind in recent history. In the face of this severe disaster, people all over the world are frightened of the prospect of facing an outbreak or an annual recurrence. However, the development of a vaccine will help control the impact of COVID-19. Women in particular have been more seriously affected by the pandemic. Since the pressure and physical load they suffer are often greater than what men endure, women are more threatened by COVID-19. Though women have a poorer quality of life and work and face worse economic conditions, they also tend to have better physiological immunity than men, which can ease the effect of COVID-19. The early development of a vaccine against COVID-19 is an important issue that must take into consideration women's better immune response to the virus along with the technique of hormone regulation. Relevant research has been conducted on female-specific vaccines in the past, and women's issues were considered during those clinical trials to ensure that complications and antibody responses were positive and effective in women. National policies should also propose good strategies for women to be vaccinated. This could improve consciousness, give women a better vaccination experience, enhance their willingness to vaccinate, and protect them from COVID-19 infection.
Collapse
Affiliation(s)
- Wen-Han Chang
- Department of Medicine, Mackay Medical College, New Taipei, Taiwan; Department of Emergency Medicine, Mackay Memorial Hospital, Taipei, Taiwan; Mackay Medicine, Nursing and Management College, Taipei, Taiwan; Institute of Mechatronic Engineering, National Taipei University of Technology, Taipei, Taiwan; Graduate Institute of Injury Prevention and Control, College of Public Health and Nutrition, Taipei Medical University, Taipei, Taiwan; Department of Emergency, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
50
|
Mohapatra RK, Pintilie L, Kandi V, Sarangi AK, Das D, Sahu R, Perekhoda L. The recent challenges of highly contagious COVID-19, causing respiratory infections: Symptoms, diagnosis, transmission, possible vaccines, animal models, and immunotherapy. Chem Biol Drug Des 2020; 96:1187-1208. [PMID: 32654267 PMCID: PMC7405220 DOI: 10.1111/cbdd.13761] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/20/2020] [Accepted: 06/28/2020] [Indexed: 01/07/2023]
Abstract
COVID-19 is highly contagious pathogenic viral infection initiated from Wuhan seafood wholesale market of China on December 2019 and spread rapidly around the whole world due to onward transmission. This recent outbreak of novel coronavirus (CoV) was believed to be originated from bats and causing respiratory infections such as common cold, dry cough, fever, headache, dyspnea, pneumonia, and finally Severe Acute Respiratory Syndrome (SARS) in humans. For this widespread zoonotic virus, human-to-human transmission has resulted in nearly 83 lakh cases in 213 countries and territories with 4,50,686 deaths as on 19 June 2020. This review presents a report on the origin, transmission, symptoms, diagnosis, possible vaccines, animal models, and immunotherapy for this novel virus and will provide ample references for the researchers toward the ongoing development of therapeutic agents and vaccines and also preventing the spread of this disease.
Collapse
Affiliation(s)
| | - Lucia Pintilie
- Department of Synthesis of Bioactive Substances and Pharmaceutical TechnologiesNational Institute for Chemical and Pharmaceutical Research and DevelopmentBucharestRomania
| | - Venkataramana Kandi
- Department of MicrobiologyPratima Institute of Medical SciencesKarimnagarHyderabadIndia
| | - Ashish K. Sarangi
- Department of ChemistrySchool of Applied Sciences, Centurion University of Technology and ManagementOdishaIndia
| | - Debadutta Das
- Department of ChemistrySukanti Degree CollegeSubarnapurOdishaIndia
| | - Raghaba Sahu
- College of PharmacySeoul National UniversitySeoulSouth Korea
| | - Lina Perekhoda
- Department of medicinal chemistryNational University of PharmacyKharkivUkraine
| |
Collapse
|