1
|
Fehlner-Gardiner C, Gongal G, Tenzin T, Sabeta C, De Benedictis P, Rocha SM, Vargas A, Cediel-Becerra N, Gomez LC, Maki J, Rupprecht CE. Rabies in Cats-An Emerging Public Health Issue. Viruses 2024; 16:1635. [PMID: 39459967 PMCID: PMC11512395 DOI: 10.3390/v16101635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/04/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Human rabies cases today are predominantly associated with infection from rabid domestic dogs. Unlike dogs, a common global reservoir species that perpetuates rabies viruses (RABV) within their populations, domestic cats are much less frequently reported or vaccinated. Epidemiologically, cats are important vectors of lyssaviruses but are not viral reservoirs. Typically, cats are incidental hosts only, infected with the predominant lyssavirus in their geographic locale. Human cases associated with rabid cats have occurred in Africa, Asia, Europe and throughout the Americas. As adept, solitary hunters, wild and domestic felids are at risk of lyssavirus infection based upon interactions with infected prey, such as bats, or from transmission by other mesocarnivores, such as rabid dogs, foxes, jackals, raccoons, and skunks. Current veterinary vaccines provide safe and effective immunity in cats against phylogroup I lyssaviruses, such as RABV, but not against divergent lyssaviruses in phylogroups II-IV. With the focus upon the global elimination of canine rabies, the emergence of rabies in cats represents a concerning trend. Clearly, education about the occurrence of rabies in cats needs to be improved, as well as the routine vaccination of cats to reduce the associated risks to public health, agriculture, and conservation biology from a One Health perspective.
Collapse
Affiliation(s)
| | - Gyanendra Gongal
- World Health Organization Regional Office for South-East Asia, New Delhi 110 002, India;
| | - Tenzin Tenzin
- World Organisation for Animal Health, Sub-Regional Representation for Southern Africa, Gaborone P.O. Box 25662, Botswana;
| | - Claude Sabeta
- Department of Veterinary Tropical Diseases, University of Pretoria, Pretoria 0110, South Africa;
| | | | - Silene Manrique Rocha
- Department of Health and Environment Surveillance, Ministry of Health of Brazil, Brasilia 70.719-040, Brazil; (S.M.R.); (A.V.)
| | - Alexander Vargas
- Department of Health and Environment Surveillance, Ministry of Health of Brazil, Brasilia 70.719-040, Brazil; (S.M.R.); (A.V.)
| | | | | | - Joanne Maki
- Boehringer Ingelheim Animal Health USA Inc., Athens, GA 30601, USA;
| | - Charles E. Rupprecht
- College of Forestry, Wildlife and Environment, Auburn University, Auburn, AL 36849, USA;
- College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| |
Collapse
|
2
|
Jogi HR, Smaraki N, Rajak KK, Yadav AK, Bhatt M, Einstien C, Revathi A, Thakur R, Kamothi DJ, Dedeepya PVSS, Savsani HH. Revolutionizing Veterinary Health with Viral Vector-Based Vaccines. Indian J Microbiol 2024; 64:867-878. [PMID: 39282171 PMCID: PMC11399537 DOI: 10.1007/s12088-024-01341-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/13/2024] [Indexed: 09/18/2024] Open
Abstract
Vaccines signify one of the economical and reasonable means to prevent and eradicate the important infectious diseases. Conventional vaccines like live attenuated and inactivated vaccines comprise of whole pathogen either in attenuated or killed form. While, new generation vaccines have been designed to elicit immune response by genetically modifying only the nucleic acid portion of that pathogen. These new generation therapeutics include mRNA vaccines, DNA plasmid vaccines, chimeric vaccines and recombinant viral vector-based vaccines. Nucleic acid based vaccines use genetic material itself thus, they are highly stable and potent in nature to induce long-lasting immune response. Amongst these novel vaccine platforms, viral vector-based vaccines is one such emerging field which has proven to be extremely effective and potent. Nowadays, veterinary medicine has also accepted this innovative vectored vaccine platform to develop an effective control strategy against certain important viral diseases of animals. Viral vector-based vaccine uses various DNA and RNA viruses of human or animal origin to carry an immunogenic transgene of target pathogen. These vaccines enhance both humoral and cell mediated immune response without use of any accessory immune-stimulants. Till today, several viruses have been modified to be characterized as vaccine vectors. Currently, large number of research programs are going on to develop vectored vaccines and novel viral vector for veterinary use. In the present review, different kinds of viral vectored vaccines having veterinary importance have been discussed.
Collapse
Affiliation(s)
- Harsh Rajeshbhai Jogi
- Division of Biological Products, ICAR-Indian Veterinary Research Institute (ICAR-IVRI), Izatnagar, Bareilly, Uttar Pradesh 243122 India
| | - Nabaneeta Smaraki
- CADRAD, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh 243122 India
| | - Kaushal Kishor Rajak
- Division of Biological Products, ICAR-Indian Veterinary Research Institute (ICAR-IVRI), Izatnagar, Bareilly, Uttar Pradesh 243122 India
| | - Ajay Kumar Yadav
- Division of Biological Products, ICAR-Indian Veterinary Research Institute (ICAR-IVRI), Izatnagar, Bareilly, Uttar Pradesh 243122 India
| | - Mukesh Bhatt
- CADRAD, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh 243122 India
| | - Chris Einstien
- Division of Biological Products, ICAR-Indian Veterinary Research Institute (ICAR-IVRI), Izatnagar, Bareilly, Uttar Pradesh 243122 India
| | - Annepu Revathi
- Division of Biological Products, ICAR-Indian Veterinary Research Institute (ICAR-IVRI), Izatnagar, Bareilly, Uttar Pradesh 243122 India
| | - Ravi Thakur
- Division of Biological Products, ICAR-Indian Veterinary Research Institute (ICAR-IVRI), Izatnagar, Bareilly, Uttar Pradesh 243122 India
| | - Dhaval J Kamothi
- Division of Pharmacology and Toxicology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh 243122 India
| | - P V S S Dedeepya
- Division of Biological Products, ICAR-Indian Veterinary Research Institute (ICAR-IVRI), Izatnagar, Bareilly, Uttar Pradesh 243122 India
| | - H H Savsani
- Veterinary College, Kamdhenu University, Junagadh, Gujarat 362001 India
| |
Collapse
|
3
|
Wang W, Wang S, Meng X, Zhao Y, Li N, Wang T, Feng N, Yan F, Xia X. A virus-like particle candidate vaccine based on CRISPR/Cas9 gene editing technology elicits broad-spectrum protection against SARS-CoV-2. Antiviral Res 2024; 225:105854. [PMID: 38447647 DOI: 10.1016/j.antiviral.2024.105854] [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: 10/29/2023] [Revised: 02/08/2024] [Accepted: 03/02/2024] [Indexed: 03/08/2024]
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants with frequent mutations has seriously damaged the effectiveness of the 2019 coronavirus disease (COVID-19) vaccine. There is an urgent need to develop a broad-spectrum vaccine while elucidating the underlying immune mechanisms. Here, we developed a SARS-CoV-2 virus-like particles (VLPs) vaccine based on the Canarypox-virus vector (ALVAC-VLPs) using CRISPR/Cas9. Immunization with ALVAC-VLPs showed the effectively induce SARS-CoV-2 specific T and B cell responses to resist the lethal challenge of mouse adaptive strains. Notably, ALVAC-VLPs conferred protection in golden hamsters against SARS-CoV-2 Wuhan-Hu-1 (wild-type, WT) and variants (Beta, Delta, Omicron BA.1, and BA.2), as evidenced by the prevention of weight loss, reduction in lung and turbinate tissue damage, and decreased viral load. Further investigation into the mechanism of immune response induced by ALVAC-VLPs revealed that toll-like receptor 4 (TLR4) mediates the recruitment of dendritic cells (DCs) to secondary lymphoid organs, thereby initiating follicle assisted T (Tfh) cell differentiation, the proliferation of germinal center (GC) B cells and plasma cell production. These findings demonstrate the immunogenicity and efficacy of the safe ALVAC-VLPs vaccine against SARS-CoV-2 and provide valuable insight into the development of COVID-19 vaccine strategies.
Collapse
Affiliation(s)
- Weiqi Wang
- College of Veterinary Medicine, Jilin University, Changchun, 130062, Jilin, China; Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, Jilin, China
| | - Shen Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, Jilin, China
| | - Xianyong Meng
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, Jilin, China; College of Veterinary Medicine, Jilin Agricultural University, Changchun, 130118, Jilin, China
| | - Yongkun Zhao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, Jilin, China
| | - Nan Li
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, Jilin, China
| | - Tiecheng Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, Jilin, China
| | - Na Feng
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, Jilin, China.
| | - Feihu Yan
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, Jilin, China.
| | - Xianzhu Xia
- College of Veterinary Medicine, Jilin University, Changchun, 130062, Jilin, China; Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, Jilin, China.
| |
Collapse
|
4
|
Weidinger AK, Bergmann M, König M, Zablotski Y, Hartmann K. Anti-rabies humoral immune response in cats after concurrent vs separate vaccination against rabies and feline leukaemia virus using canarypox-vectored vaccines. J Feline Med Surg 2024; 26:1098612X231218643. [PMID: 38358295 PMCID: PMC10911302 DOI: 10.1177/1098612x231218643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2024]
Abstract
OBJECTIVES Some expert groups recommend that cats should be vaccinated with non-adjuvanted feline leukaemia virus (FeLV) and rabies vector vaccines, which, in the European Union, are currently not licensed for concurrent use and have to be administered at least 14 days apart (different from the USA) and thus at separate visits, which is associated with more stress for cats and owners. The aim of this study was to assess the anti-rabies antibody response in cats after vaccination against rabies and FeLV at concurrent vs separate (4 weeks apart) visits using two canarypox-vectored vaccines (Purevax Rabies and Purevax FeLV; Boehringer Ingelheim) and to evaluate the occurrence of vaccine-associated adverse events (VAAEs). METHODS Healthy FeLV antigen-negative client-owned kittens (n = 106) were prospectively included in this randomised study. All kittens received primary vaccinations against rabies (week 0) and FeLV (weeks 4 and 8). After 1 year, the study group (n = 52) received booster vaccinations against rabies and FeLV concurrently at the same visit (weeks 50-52). The control group (n = 54) received booster vaccinations against rabies (weeks 50-52) and FeLV (weeks 54-56) separately. Anti-rabies virus antibodies (anti-RAV Ab) were determined by fluorescent antibody virus neutralisation assay at weeks 4, 50-52 and 54-56, and compared between both groups using a Mann-Whitney U-test. RESULTS Four weeks after the first rabies vaccination, 87/106 (82.1%) kittens had a titre ⩾0.5 IU/ml and 19/106 (17.9%) had a titre <0.5 IU/ml. Four weeks after the 1-year rabies booster, all cats had adequate anti-RAV Ab according to the World Organisation for Animal Health (⩾0.5 IU/ml), and the titres of the study group (median = 14.30 IU/ml) and the control group (median = 21.39 IU/ml) did not differ significantly (P = 0.141). VAAEs were observed in 7/106 (6.6%) cats. CONCLUSIONS AND RELEVANCE Concurrent administration of Purevax FeLV and Purevax Rabies vector vaccines at the 1-year booster does not interfere with the development of anti-RAV Ab or cause more adverse effects and thus represents a better option than separate vaccination visits for cats and owners.
Collapse
Affiliation(s)
- Anna-Karina Weidinger
- LMU Small Animal Clinic, Centre for Clinical Veterinary Medicine, LMU Munich, Munich, Germany
| | - Michèle Bergmann
- LMU Small Animal Clinic, Centre for Clinical Veterinary Medicine, LMU Munich, Munich, Germany
| | - Matthias König
- Institute of Virology, Faculty of Veterinary Medicine, Justus Liebig University, Giessen, Hessen, Germany
| | - Yury Zablotski
- LMU Small Animal Clinic, Centre for Clinical Veterinary Medicine, LMU Munich, Munich, Germany
| | - Katrin Hartmann
- LMU Small Animal Clinic, Centre for Clinical Veterinary Medicine, LMU Munich, Munich, Germany
| |
Collapse
|
5
|
Zhang Y, Mo R, Sun S, Cui Z, Liang B, Li E, Wang T, Feng Y, Yang S, Yan F, Zhao Y, Xia X. Bacillus subtilis vector based oral rabies vaccines induced potent immune response and protective efficacy in mice. Front Microbiol 2023; 14:1126533. [PMID: 36846792 PMCID: PMC9948087 DOI: 10.3389/fmicb.2023.1126533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 01/09/2023] [Indexed: 02/11/2023] Open
Abstract
Introduction Rabies is a worldwide epidemic that poses a serious threat to global public health. At present, rabies in domestic dogs, cats, and some pets can be effectively prevented and controlled by intramuscular injection of rabies vaccine. But for some inaccessible animals, especially stray dogs, and wild animals, it is difficult to prevent with intramuscular injection. Therefore, it is necessary to develop a safe and effective oral rabies vaccine. Methods We constructed recombinant Bacillus subtilis (B. subtilis) expressing two different strains of rabies virus G protein, named CotG-E-G and CotG-C-G, immunogenicity was studied in mice. Results The results showed that CotG-E-G and CotG-C-G could significantly increase the specific SIgA titers in feces, serum IgG titers, and neutralizing antibodies. ELISpot experiments showed that CotG-E-G and CotG-C-G could also induce Th1 and Th2 to mediate the secretion of immune-related IFN-γ and IL-4. Collectively, our results suggested that recombinant B. subtilis CotG-E-G and CotG-C-G have excellent immunogenicity and are expected to be novel oral vaccine candidates for the prevention and control of wild animal rabies.
Collapse
Affiliation(s)
- Ying Zhang
- Northeast Forestry University College of Wildlife and Protected Area, Harbin, China,Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| | - Ruo Mo
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China,College of Veterinary Medicine, Jilin Agricultural University, Changchun, Jilin, China
| | - Sheng Sun
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| | - Zhanding Cui
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Bo Liang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| | - Entao Li
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Tiecheng Wang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| | - Ye Feng
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| | - Songtao Yang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| | - Feihu Yan
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China,Feihu Yan,✉
| | - Yongkun Zhao
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China,Yongkun Zhao,✉
| | - Xianzhu Xia
- Northeast Forestry University College of Wildlife and Protected Area, Harbin, China,Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China,*Correspondence: Xianzhu Xia,✉
| |
Collapse
|
6
|
El-Hage C, Hartley C, Savage C, Watson J, Gilkerson J, Paillot R. Assessment of Humoral and Long-Term Cell-Mediated Immune Responses to Recombinant Canarypox-Vectored Equine Influenza Virus Vaccination in Horses Using Conventional and Accelerated Regimens Respectively. Vaccines (Basel) 2022; 10:vaccines10060855. [PMID: 35746463 PMCID: PMC9229645 DOI: 10.3390/vaccines10060855] [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: 04/26/2022] [Revised: 05/19/2022] [Accepted: 05/25/2022] [Indexed: 02/05/2023] Open
Abstract
During Australia's first and only outbreak of equine influenza (EI), which was restricted to two northeastern states, horses were strategically vaccinated with a recombinant canarypox-vectored vaccine (rCP-EIV; ProteqFlu™, Merial P/L). The vaccine encoded for haemagglutinin (HA) belonging to two equine influenza viruses (EIVs), including an American and Eurasian lineage subtype that predated the EIV responsible for the outbreak (A/equine/Sydney/07). Racehorses in Victoria (a southern state that remained free of EI) were vaccinated prophylactically. Although the vaccine encoded for (HA) belonged to two EIVs of distinct strains of the field virus, clinical protection was reported in vaccinated horses. Our aim is to assess the extent of humoral immunity in one group of vaccinated horses and interferon-gamma ((EIV)-IFN-γ)) production in the peripheral blood mononuclear cells (PBMCs) of a second population of vaccinated horses. Twelve racehorses at work were monitored for haemagglutination inhibition antibodies to three antigenically distinct equine influenza viruses (EIVs) The EIV antigens included two H3N8 subtypes: A/equine/Sydney/07) A/equine/Newmarket/95 (a European lineage strain) and an H7N7 subtype (A/equine/Prague1956). Cell-mediated immune responses of: seven racehorses following an accelerated vaccination schedule, two horses vaccinated using a conventional regimen, and six unvaccinated horses were evaluated by determining (EIV)-IFN-γ levels. Antibody responses following vaccination with ProteqFlu™ were cross-reactive in nature, with responses to both H3N8 EIV strains. Although (EIV)IFN-γ was clearly detected following the in vitro re-stimulation of PBMC, there was no significant difference between the different groups of horses. Results of this study support reports of clinical protection of Australian horses following vaccination with Proteq-Flu™ with objective evidence of humoral cross-reactivity to the outbreak viral strain A/equine/Sydney/07.
Collapse
Affiliation(s)
- Charles El-Hage
- Centre for Equine Infectious Diseases, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC 3010, Australia; (C.H.); (C.S.); (J.G.)
- Correspondence: ; Tel.: +61-417166029
| | - Carol Hartley
- Centre for Equine Infectious Diseases, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC 3010, Australia; (C.H.); (C.S.); (J.G.)
| | - Catherine Savage
- Centre for Equine Infectious Diseases, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC 3010, Australia; (C.H.); (C.S.); (J.G.)
| | - James Watson
- Australian Centre for Disease Preparedness, CSIRO, Geelong, VIC 3216, Australia;
| | - James Gilkerson
- Centre for Equine Infectious Diseases, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC 3010, Australia; (C.H.); (C.S.); (J.G.)
| | - Romain Paillot
- School of Equine and Veterinary Physiotherapy, Writtle University College, Lordship Road, Writtle, Chelmsford CM1 3RR, UK;
| |
Collapse
|
7
|
Elakov AL. [Anti-rabies vaccines applied in the Russian Federation and perspectives for their improvement]. Vopr Virusol 2022; 67:107-114. [PMID: 35521983 DOI: 10.36233/0507-4088-102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 05/05/2022] [Indexed: 12/25/2022]
Abstract
Rabies is almost ubiquitous (except in certain areas) and poses a significant danger to both animals and humans. Every year around 55,000 people die from this disease worldwide. In the Russian Federation alone 400,000- 450,000 patients annually apply for anti-rabies treatment. In the absolute majority of cases human infection is caused by contact with infected animals. In RF, a number of cultured inactivated anti-rabies vaccines for medical and veterinary purposes have been developed, registered and used for specific prevention of rabies. These vaccine preparations have shown high effectiveness in preventing infection in domestic and farm animals. At the same time, the main reservoir of the rabies virus (Mononegavirales: Rhabdoviridae: Lyssavirus) (RV) are wild carnivores (Mammalia: Carnivora). For the purpose of their oral immunization, live virus vaccines from attenuated (fixed) strains of RV that are little resistant in the external environment are used. In Western Europe and North America there is successful experience with recombinant anti-rabies vaccine preparations containing a viral glycoprotein gene (G-protein). Such vaccines are safe for humans and animals. In Russia also had been developed a vector anti-rabies vaccine based on adenovirus (Adenoviridae), which can be used to combat this infection. Currently, in addition to classical rabies, diseases caused by new, previously unknown lyssaviruses (Lyssavirus) are becoming increasingly important. Bats (Mammalia: Microchiroptera) are their vectors. Cases of illness and death after contact with these animals have been described. In the near future, we should expect the development of new vaccines that will provide protection not only against RV, but also against other lyssaviruses.
Collapse
Affiliation(s)
- A L Elakov
- FSBSI «Federal Scientific Center - All-Russian Scientific Research Institute of Experimental Veterinary Medicine named after K.I. Skryabin and Ya.R. Kovalenko of the Russian Academy of Sciences»
| |
Collapse
|
8
|
Unger H, Kangethe RT, Liaqat F, Viljoen GJ. Advances in Irradiated Livestock Vaccine Research and Production Addressing the Unmet Needs for Farmers and Veterinary Services in FAO/IAEA Member States. Front Immunol 2022; 13:853874. [PMID: 35418985 PMCID: PMC8997582 DOI: 10.3389/fimmu.2022.853874] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 03/07/2022] [Indexed: 12/13/2022] Open
Abstract
The Animal Production and Health section (APH) of the Joint FAO/IAEA Centre of Nuclear Techniques in Food and Agriculture at the International Atomic Energy Agency has over the last 58 years provided technical and scientific support to more than 100 countries through co-ordinated research activities and technical co-operation projects in peaceful uses of nuclear technologies. A key component of this support has been the development of irradiated vaccines targeting diseases that are endemic to participating countries. APH laboratories has over the last decade developed new techniques and has put in place a framework that allows researchers from participating member states to develop relevant vaccines targeting local diseases while using irradiation as a tool for improving livestock resources.
Collapse
Affiliation(s)
- Hermann Unger
- Animal Production and Health Section, Joint FAO/IAEA Centre of Nuclear Techniques in Food and Agriculture, Department of Nuclear Sciences and Applications, International Atomic Energy Agency (IAEA), Vienna, Austria
| | - Richard T Kangethe
- Animal Production and Health Section, Joint FAO/IAEA Centre of Nuclear Techniques in Food and Agriculture, Department of Nuclear Sciences and Applications, International Atomic Energy Agency (IAEA), Vienna, Austria
| | - Fatima Liaqat
- Animal Production and Health Section, Joint FAO/IAEA Centre of Nuclear Techniques in Food and Agriculture, Department of Nuclear Sciences and Applications, International Atomic Energy Agency (IAEA), Vienna, Austria
| | - Gerrit J Viljoen
- Animal Production and Health Section, Joint FAO/IAEA Centre of Nuclear Techniques in Food and Agriculture, Department of Nuclear Sciences and Applications, International Atomic Energy Agency (IAEA), Vienna, Austria
| |
Collapse
|
9
|
Saba C, Eggleston R, Parks A, Peroni J, Sjoberg E, Rice S, Tyma J, Williams J, Grosenbaugh D, Leard AT. ALVAC-fIL2, a feline interleukin-2 immunomodulator, as a treatment for sarcoids in horses: A pilot study. J Vet Intern Med 2022; 36:1179-1184. [PMID: 35416353 PMCID: PMC9151478 DOI: 10.1111/jvim.16425] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 03/16/2022] [Accepted: 03/25/2022] [Indexed: 12/19/2022] Open
Abstract
Background Sarcoid tumors are common in horses and may negatively impact the performance and value of the horse. No known treatment is reliably successful. Hypotheses/Objectives To determine tolerability, overall response rate, time to response, and progression‐free survival of horses with biopsy‐confirmed or suspected sarcoids treated with ALVAC‐fIL2. Animals Client‐owned horses with measurable, presumed‐ or biopsy‐confirmed sarcoid tumors. Methods Prospective pilot study. One milliliter of ALVAC‐fIL2 was injected into 4 to 5 areas of the sarcoid(s) in each horse (week 0); this treatment was repeated in weeks 1, 3, and 7. Sarcoids were measured at each visit, and response to treatment was determined according to the Response Evaluation Criteria in Solid Tumors for dogs (v1.0). After the final treatment, horses were reassessed and sarcoids remeasured every 3 months until tumor progression or for a minimum of 1 year if progression was not documented. Results Fourteen horses were included. Tumor size decreased in 86% of the horses, and the median time to first response was 89 days (range, 34‐406 days). Median time to best response was 211 days (range, 56‐406 days), but 3 of the sarcoids still were decreasing in size at the time of final evaluation. The median progression‐free interval was not reached. Adverse events were minimal and included transient focal inflammation in 2 horses. Conclusions and Clinical Importance Intratumoral injection of ALVAC‐fIL2 has promise as a well‐tolerated and effective, tissue‐sparing treatment for horses with sarcoid tumors.
Collapse
Affiliation(s)
- Corey Saba
- Department of Large Animal Medicine and Surgery, University of Georgia College of Veterinary Medicine, Athens, Georgia, USA
| | - Randall Eggleston
- Department of Large Animal Medicine and Surgery, University of Georgia College of Veterinary Medicine, Athens, Georgia, USA
| | - Andrew Parks
- Department of Large Animal Medicine and Surgery, University of Georgia College of Veterinary Medicine, Athens, Georgia, USA
| | - John Peroni
- Department of Large Animal Medicine and Surgery, University of Georgia College of Veterinary Medicine, Athens, Georgia, USA
| | - Eric Sjoberg
- Maggie's Menagerie Veterinary Services, Ila, Georgia, USA
| | - Shelbe Rice
- Maggie's Menagerie Veterinary Services, Ila, Georgia, USA
| | - Jesse Tyma
- Department of Large Animal Medicine and Surgery, University of Georgia College of Veterinary Medicine, Athens, Georgia, USA
| | - Jarred Williams
- Department of Large Animal Medicine and Surgery, University of Georgia College of Veterinary Medicine, Athens, Georgia, USA
| | | | | |
Collapse
|
10
|
Development of Plant-Based Vaccines for Prevention of Avian Influenza and Newcastle Disease in Poultry. Vaccines (Basel) 2022; 10:vaccines10030478. [PMID: 35335110 PMCID: PMC8952014 DOI: 10.3390/vaccines10030478] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/10/2022] [Accepted: 03/16/2022] [Indexed: 02/01/2023] Open
Abstract
Viral diseases, including avian influenza (AI) and Newcastle disease (ND), are an important cause of morbidity and mortality in poultry, resulting in significant economic losses. Despite the availability of commercial vaccines for the major viral diseases of poultry, these diseases continue to pose a significant risk to global food security. There are multiple factors for this: vaccine costs may be prohibitive, cold chain storage for attenuated live-virus vaccines may not be achievable, and commercial vaccines may protect poorly against local emerging strains. The development of transient gene expression systems in plants provides a versatile and robust tool to generate a high yield of recombinant proteins with superior speed while managing to achieve cost-efficient production. Plant-derived vaccines offer good stability and safety these include both subunit and virus-like particle (VLP) vaccines. VLPs offer potential benefits compared to currently available traditional vaccines, including significant reductions in virus shedding and the ability to differentiate between infected and vaccinated birds (DIVA). This review discusses the current state of plant-based vaccines for prevention of the AI and ND in poultry, challenges in their development, and potential for expanding their use in low- and middle-income countries.
Collapse
|
11
|
Lemon JL, McMenamy MJ. A Review of UK-Registered and Candidate Vaccines for Bovine Respiratory Disease. Vaccines (Basel) 2021; 9:vaccines9121403. [PMID: 34960149 PMCID: PMC8703677 DOI: 10.3390/vaccines9121403] [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: 08/26/2021] [Revised: 11/10/2021] [Accepted: 11/22/2021] [Indexed: 01/11/2023] Open
Abstract
Vaccination is widely regarded as a cornerstone in animal or herd health and infectious disease management. Nineteen vaccines against the major pathogens implicated in bovine respiratory disease are registered for use in the UK by the Veterinary Medicines Directorate (VMD). However, despite annual prophylactic vaccination, bovine respiratory disease is still conservatively estimated to cost the UK economy approximately £80 million per annum. This review examines the vaccine types available, discusses the surrounding literature and scientific rationale of the limitations and assesses the potential of novel vaccine technologies.
Collapse
Affiliation(s)
- Joanne L. Lemon
- Sustainable Agri-Food and Sciences Division, Agri-Food and Bioscience Institute, Newforge Lane, Belfast BT9 5PX, UK
- Correspondence:
| | - Michael J. McMenamy
- Veterinary Sciences Division, Agri-Food and Bioscience Institute, Stormont, Belfast BT4 3SD, UK;
| |
Collapse
|
12
|
Cid R, Bolívar J. Platforms for Production of Protein-Based Vaccines: From Classical to Next-Generation Strategies. Biomolecules 2021; 11:1072. [PMID: 34439738 PMCID: PMC8394948 DOI: 10.3390/biom11081072] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/16/2021] [Accepted: 07/17/2021] [Indexed: 12/12/2022] Open
Abstract
To date, vaccination has become one of the most effective strategies to control and reduce infectious diseases, preventing millions of deaths worldwide. The earliest vaccines were developed as live-attenuated or inactivated pathogens, and, although they still represent the most extended human vaccine types, they also face some issues, such as the potential to revert to a pathogenic form of live-attenuated formulations or the weaker immune response associated with inactivated vaccines. Advances in genetic engineering have enabled improvements in vaccine design and strategies, such as recombinant subunit vaccines, have emerged, expanding the number of diseases that can be prevented. Moreover, antigen display systems such as VLPs or those designed by nanotechnology have improved the efficacy of subunit vaccines. Platforms for the production of recombinant vaccines have also evolved from the first hosts, Escherichia coli and Saccharomyces cerevisiae, to insect or mammalian cells. Traditional bacterial and yeast systems have been improved by engineering and new systems based on plants or insect larvae have emerged as alternative, low-cost platforms. Vaccine development is still time-consuming and costly, and alternative systems that can offer cost-effective and faster processes are demanding to address infectious diseases that still do not have a treatment and to face possible future pandemics.
Collapse
Affiliation(s)
- Raquel Cid
- ADL Bionatur Solutions S.A., Av. del Desarrollo Tecnológico 11, 11591 Jerez de la Frontera, Spain
| | - Jorge Bolívar
- Department of Biomedicine, Biotechnology and Public Health-Biochemistry and Molecular Biology, Campus Universitario de Puerto Real, University of Cadiz, 11510 Puerto Real, Spain
| |
Collapse
|
13
|
Volkmann A, Williamson AL, Weidenthaler H, Meyer TPH, Robertson JS, Excler JL, Condit RC, Evans E, Smith ER, Kim D, Chen RT. The Brighton Collaboration standardized template for collection of key information for risk/benefit assessment of a Modified Vaccinia Ankara (MVA) vaccine platform. Vaccine 2021; 39:3067-3080. [PMID: 33077299 PMCID: PMC7568176 DOI: 10.1016/j.vaccine.2020.08.050] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 08/18/2020] [Indexed: 12/25/2022]
Abstract
The Brighton Collaboration Viral Vector Vaccines Safety Working Group (V3SWG) was formed to evaluate the safety and characteristics of live, recombinant viral vector vaccines. The Modified Vaccinia Ankara (MVA) vector system is being explored as a platform for development of multiple vaccines. This paper reviews the molecular and biological features specifically of the MVA-BN vector system, followed by a template with details on the safety and characteristics of an MVA-BN based vaccine against Zaire ebolavirus and other filovirus strains. The MVA-BN-Filo vaccine is based on a live, highly attenuated poxviral vector incapable of replicating in human cells and encodes glycoproteins of Ebola virus Zaire, Sudan virus and Marburg virus and the nucleoprotein of the Thai Forest virus. This vaccine has been approved in the European Union in July 2020 as part of a heterologous Ebola vaccination regimen. The MVA-BN vector is attenuated following over 500 serial passages in eggs, showing restricted host tropism and incompetence to replicate in human cells. MVA has six major deletions and other mutations of genes outside these deletions, which all contribute to the replication deficiency in human and other mammalian cells. Attenuation of MVA-BN was demonstrated by safe administration in immunocompromised mice and non-human primates. In multiple clinical trials with the MVA-BN backbone, more than 7800 participants have been vaccinated, demonstrating a safety profile consistent with other licensed, modern vaccines. MVA-BN has been approved as smallpox vaccine in Europe and Canada in 2013, and as smallpox and monkeypox vaccine in the US in 2019. No signal for inflammatory cardiac disorders was identified throughout the MVA-BN development program. This is in sharp contrast to the older, replicating vaccinia smallpox vaccines, which have a known risk for myocarditis and/or pericarditis in up to 1 in 200 vaccinees. MVA-BN-Filo as part of a heterologous Ebola vaccination regimen (Ad26.ZEBOV/MVA-BN-Filo) has undergone clinical testing including Phase III in West Africa and is currently in use in large scale vaccination studies in Central African countries. This paper provides a comprehensive picture of the MVA-BN vector, which has reached regulatory approvals, both as MVA-BN backbone for smallpox/monkeypox, as well as for the MVA-BN-Filo construct as part of an Ebola vaccination regimen, and therefore aims to provide solutions to prevent disease from high-consequence human pathogens.
Collapse
Affiliation(s)
| | - Anna-Lise Williamson
- Institute of Infectious Disease and Molecular Medicine at the University of Cape Town, South Africa
| | | | | | | | | | - Richard C Condit
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, USA
| | - Eric Evans
- 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.
| | - Denny Kim
- Janssen Pharmaceuticals, Titusville, NJ, USA
| | - Robert T Chen
- Brighton Collaboration, a Program of the Task Force for Global Health, Decatur, GA, USA
| |
Collapse
|
14
|
Loschelder-Ostrowski J, Winter JC, Merle R, Klopfleisch R, Gehlen H. Treatment of equine sarcoids using recombinant poxviruses expressing feline interleukin-2. Vet Dermatol 2021; 32:283-e77. [PMID: 33728715 DOI: 10.1111/vde.12941] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 10/14/2020] [Accepted: 11/22/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND Interleukin (IL)-2 stimulates antitumour immunity and is successfully used for the treatment of different neoplasias. HYPOTHESIS/OBJECTIVES Canarypox virus locally expressing feline IL-2 is safe and can be used to treat equine sarcoids. ANIMALS Twenty horses of different breeds with a median age of eight years (interquartile range 6.0-13.3 years) and a total number of 59 sarcoids were included in the study. METHODS In this prospective clinical trial, sarcoids were injected twice seven days apart, with a recombinant canarypox virus expressing feline IL-2. Complete blood counts (CBC) and fibrinogen levels were measured before treatment and on days 1, 2, 7 and 8. RESULTS Complete regression was achieved in eight horses (40%) and partial regression in two horses (10%). No change in sarcoid size was observed in two horses (10%) and the disease progressed in five horses (25%). Sarcoids of three horses (15%) showed initial response followed by tumour growth. There were no significant changes in CBC and fibrinogen levels after either injection. One horse developed a mild fever the day after each injection, which subsided without treatment the following day. CONCLUSIONS Treatment of equine sarcoids with recombinant canarypox virus expressing feline IL-2 seems to be a safe therapy option. Although the expression of IL-2 after vector injection and its biological activity in horses were not proven in this study, the treatment resulted in regression and partial regression in 50% of the cases. Further studies are necessary to verify these findings and to establish a treatment protocol.
Collapse
Affiliation(s)
| | | | - Roswitha Merle
- Department of Epidemiology, Free University of Berlin, Königsweg 67, Berlin, 14163, Germany
| | - Robert Klopfleisch
- Department of Veterinary Pathology, Free University of Berlin, Robert-von-Ostertag-Str. 15, Berlin, 14163, Germany
| | - Heidrun Gehlen
- Clinic for Horses, Free University of Berlin, Oertzenweg 19b, Berlin, 14163, Germany
| |
Collapse
|
15
|
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: 70] [Impact Index Per Article: 14.0] [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
|
16
|
Construction and evaluation of recombinant Lactobacillus plantarum NC8 delivering one single or two copies of G protein fused with a DC-targeting peptide (DCpep) as novel oral rabies vaccine. Vet Microbiol 2020; 251:108906. [PMID: 33160196 DOI: 10.1016/j.vetmic.2020.108906] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 10/20/2020] [Indexed: 11/24/2022]
Abstract
Rabies remains an important public health threat in most developing countries. To develop a more effective and safe oral vaccine against rabies, we constructed recombinant Lactobacillus plantarum NC8 carrying one or two copies of the G gene with a dendritic cell-targeting peptide (DCpep) fused at the C-terminal designated NC8-pSIP409-sRVG or NC8-pSIP409-dRVG, respectively. The immunogenicity and protective efficacy of these recombinant Lactobacillus plantarum against RABV were evaluated by oral administration in a mouse model. The results showed that recombinant NC8-pSIP409-dRVG possessed more G protein, resulting in more functional maturation of DCs. After three cycle of oral immunization, NC8-pSIP409-dRVG induced significantly higher levels of specific IgG antibody and mixed Th1/Th2 with a strong Th1-biasd immune response in mice. Most importantly, although the titers of RABV neutralizing antibody (VNA) were below the threshold of 0.5 IU/mL, the NC8-pSIP409-dRVG could protect 60 % of inoculated mice against lethal RABV challenge. These data reveal that recombinant NC8-pSIP409-dRVG may be a novel and promising oral vaccine candidate to prevent and control of animal rabies.
Collapse
|
17
|
Romanutti C, Keller L, Zanetti FA. Current status of virus-vectored vaccines against pathogens that affect poultry. Vaccine 2020; 38:6990-7001. [PMID: 32951939 DOI: 10.1016/j.vaccine.2020.09.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 08/12/2020] [Accepted: 09/02/2020] [Indexed: 01/04/2023]
Abstract
The most effective strategies for the control of disease in poultry are vaccination and biosecurity. Vaccines useful against pathogens affecting poultry must be safe, effective with a single dose, inexpensive, applicable by mass vaccination methods, and able to induce a protective immune response in the presence of maternal antibodies. Viral vector meet some of these characteristics and if the attenuated virus used as vector infects birds, the vaccine will have the advantage of being bivalent. Thus, viral vectors are currently a tool of choice for the development of new poultry vaccines. This review describes the main viruses used as vectors for the delivery and in vivo expression of antigens of poultry pathogens. It also presents the methodologies most frequently used to obtain recombinant viral vectors and summarizes the state-of-the-art related to vectored vaccines in poultry (some of them currently licensed), the pathogens targeted and their antigens, and the ability of these vaccines to induce an effective immune response. Finally, the review discusses the results of a few studies comparing recombinant viral vector vaccines and live-attenuated vaccines in vaccine matching challenges, and mentions strategies and future researches that can help to improve the efficacy of vectored vaccines in poultry birds.
Collapse
Affiliation(s)
- Carina Romanutti
- Centro de Virología Animal (CEVAN), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Saladillo 2468 (C1440FFX), Ciudad Autónoma de Buenos Aires, Argentina.
| | - Leticia Keller
- Instituto de Ciencia y Tecnología "Dr. Cesar Milstein", CONICET, Saladillo 2468 (C1440FFX), Ciudad Autónoma de Buenos Aires, Argentina.
| | - Flavia Adriana Zanetti
- Instituto de Ciencia y Tecnología "Dr. Cesar Milstein", CONICET, Saladillo 2468 (C1440FFX), Ciudad Autónoma de Buenos Aires, Argentina.
| |
Collapse
|
18
|
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has resulted in a global pandemic, prompting unprecedented efforts to contain the virus. Many developed countries have implemented widespread testing and have rapidly mobilized research programmes to develop vaccines and therapeutics. However, these approaches may be impractical in Africa, where the infrastructure for testing is poorly developed and owing to the limited manufacturing capacity to produce pharmaceuticals. Furthermore, a large burden of HIV-1 and tuberculosis in Africa could exacerbate the severity of infection and may affect vaccine immunogenicity. This Review discusses global efforts to develop diagnostics, therapeutics and vaccines, with these considerations in mind. We also highlight vaccine and diagnostic production platforms that are being developed in Africa and that could be translated into clinical development through appropriate partnerships for manufacture. The COVID-19 pandemic has prompted unparalleled progress in the development of vaccines and therapeutics in many countries, but it has also highlighted the vulnerability of resource-limited countries in Africa. Margolin and colleagues review global efforts to develop SARS-CoV-2 diagnostics, therapeutics and vaccines, with a focus on the opportunities and challenges in Africa.
Collapse
|
19
|
Abstract
This brief review discusses some recent advances in vaccine technologies with particular reference to their application within veterinary medicine. It highlights some of the key inactivated/killed approaches to vaccination, including natural split-product and subunit vaccines, recombinant subunit and protein vaccines, and peptide vaccines. It also covers live/attenuated vaccine strategies, including modified live marker/differentiating infected from vaccinated animals vaccines, live vector vaccines, and nucleic acid vaccines.
Collapse
Affiliation(s)
- Michael James Francis
- BioVacc Consulting Ltd, The Red House, 10 Market Square, Amersham, Buckinghamshire HP7 0DQ, UK.
| |
Collapse
|
20
|
Sharma C, Rokana N, Chandra M, Singh BP, Gulhane RD, Gill JPS, Ray P, Puniya AK, Panwar H. Antimicrobial Resistance: Its Surveillance, Impact, and Alternative Management Strategies in Dairy Animals. Front Vet Sci 2018; 4:237. [PMID: 29359135 PMCID: PMC5766636 DOI: 10.3389/fvets.2017.00237] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 12/15/2017] [Indexed: 01/10/2023] Open
Abstract
Antimicrobial resistance (AMR), one among the most common priority areas identified by both national and international agencies, is mushrooming as a silent pandemic. The advancement in public health care through introduction of antibiotics against infectious agents is now being threatened by global development of multidrug-resistant strains. These strains are product of both continuous evolution and un-checked antimicrobial usage (AMU). Though antibiotic application in livestock has largely contributed toward health and productivity, it has also played significant role in evolution of resistant strains. Although, a significant emphasis has been given to AMR in humans, trends in animals, on other hand, are not much emphasized. Dairy farming involves surplus use of antibiotics as prophylactic and growth promoting agents. This non-therapeutic application of antibiotics, their dosage, and withdrawal period needs to be re-evaluated and rationally defined. A dairy animal also poses a serious risk of transmission of resistant strains to humans and environment. Outlining the scope of the problem is necessary for formulating and monitoring an active response to AMR. Effective and commendably connected surveillance programs at multidisciplinary level can contribute to better understand and minimize the emergence of resistance. Besides, it requires a renewed emphasis on investments into research for finding alternate, safe, cost effective, and innovative strategies, parallel to discovery of new antibiotics. Nevertheless, numerous direct or indirect novel approaches based on host-microbial interaction and molecular mechanisms of pathogens are also being developed and corroborated by researchers to combat the threat of resistance. This review places a concerted effort to club the current outline of AMU and AMR in dairy animals; ongoing global surveillance and monitoring programs; its impact at animal human interface; and strategies for combating resistance with an extensive overview on possible alternates to current day antibiotics that could be implemented in livestock sector.
Collapse
Affiliation(s)
- Chetan Sharma
- Department of Dairy Microbiology, College of Dairy Science and Technology, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, India
| | - Namita Rokana
- Department of Dairy Microbiology, College of Dairy Science and Technology, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, India
| | - Mudit Chandra
- Department of Veterinary Microbiology, College of Veterinary Science, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, India
| | - Brij Pal Singh
- Department of Dairy Microbiology, College of Dairy Science and Technology, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, India
| | - Rohini Devidas Gulhane
- Department of Dairy Microbiology, College of Dairy Science and Technology, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, India
| | - Jatinder Paul Singh Gill
- School of Public Health and Zoonoses, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, India
| | - Pallab Ray
- Department of Medical Microbiology, Post Graduate Institute for Medical Education and Research (PGIMER), Chandigarh, India
| | - Anil Kumar Puniya
- Department of Dairy Microbiology, College of Dairy Science and Technology, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, India
| | - Harsh Panwar
- Department of Dairy Microbiology, College of Dairy Science and Technology, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, India
| |
Collapse
|
21
|
Grosenbaugh DA, Frances-Duvert V, Abedi S, Feilmeier B, Ru H, Poulet H. Efficacy of a nonadjuvanted recombinant FeLV vaccine and two inactivated FeLV vaccines when subject to consistent virulent FeLV challenge conditions. Biologicals 2017; 49:76-80. [PMID: 28734742 DOI: 10.1016/j.biologicals.2016.10.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 09/19/2016] [Accepted: 10/17/2016] [Indexed: 11/25/2022] Open
Abstract
The purpose of this study was to compare the efficacy of three FeLV vaccines, under identical conditions in a laboratory challenge model that closely mimics natural infection. Four groups of cats (n = 20 per group) were administered two doses of vaccine, 21 days apart, starting at 9-10 weeks of age (Purevax® FeLV, Versifel® FeLV, Nobivac® feline 2-FeLV, and a placebo). Cats were challenged 3 weeks later with a virulent, heterologous FeLV isolate. FeLV antigenemia was determined at weekly intervals from 3 to 15 weeks postchallenge. Circulating proviral DNA was determined on terminal PBMC samples. Following challenge, persistent antigenemia developed in 15 (75%) placebo-vaccinated cats, 3 (15%) cats in the Versifel FeLV vaccinated group, and 1 cat (5%) each in the Purevax FeLV and the Nobivac FeLV vaccinated groups. The prevented fractions for three vaccine groups were 93%, 93%, and 80% respectively. The adjusted p-values for all vaccine group comparisons fail to approach statistical significance. There was excellent agreement between proviral FeLV DNA in circulating PBMCs and persistent antigenemia. It is shown that when cats are managed under the same conditions during a virulent challenge, via the normal route of infection, the tested vaccines all show a comparable degree of protection.
Collapse
Affiliation(s)
| | - Valérie Frances-Duvert
- Merial, Research and Development, Lyon Gerland Laboratories, 254 rue Marcel Mérieux, 69007, Lyon, France
| | - Shabnam Abedi
- Merial, Research and Development, 1730 Olympic Dr., Athens, GA 30601, USA
| | - Bradley Feilmeier
- Merial, Research and Development, 1730 Olympic Dr., Athens, GA 30601, USA
| | - Hongyu Ru
- Merial, Research and Development, 1730 Olympic Dr., Athens, GA 30601, USA
| | - Hervé Poulet
- Merial, Research and Development, Lyon Gerland Laboratories, 254 rue Marcel Mérieux, 69007, Lyon, France
| |
Collapse
|
22
|
Xiao X, Zhang Y, Wei Q, Yin X. Flagellin FljB as an adjuvant to the recombinant adenovirus rabies glycoprotein vaccine increases immune responses against rabies in mice. Arch Virol 2017; 162:2655-2665. [PMID: 28550434 DOI: 10.1007/s00705-017-3413-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 04/12/2017] [Indexed: 12/25/2022]
Abstract
Rabies virus (RABV) causes an acute progressive viral encephalitis. Although currently licensed vaccines have an excellent safety and efficacy record, the development of a safer and more cost-effective vaccine is still being sought. An E1-deleted, replication-defective human adenovirus type 5 (HAd5) vector expressing RABV glycoprotein (HAd5-G) is thought to be a promising candidate vaccine for immune prophylaxis against rabies. Salmonella enterica serovar Typhimurium (S. Typhimurium) flagellin is a well-known immune adjuvant. In this work, we have researched the adjuvant effect of flagellins (FljB and FliC) for HAd5 in mice for the first time. We found that the recombinant HAd5 expressing RABV glycoprotein and FljB (HAd5-GB), if administered intramuscularly, but not orally, could induce stronger immune responses and provide better protection against rabies than HAd5-G or the recombinant HAd5 expressing glycoprotein and FliC (HAd5-GC). These results suggest that the recombinant HAd5-GB has potential for development as a promising rabies vaccine.
Collapse
Affiliation(s)
- Xingxing Xiao
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Grazing Animal Diseases, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, 730046, Gansu, People's Republic of China
| | - Yun Zhang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Grazing Animal Diseases, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, 730046, Gansu, People's Republic of China
| | - Qiaolin Wei
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Grazing Animal Diseases, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, 730046, Gansu, People's Republic of China
| | - Xiangping Yin
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Grazing Animal Diseases, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, 730046, Gansu, People's Republic of China.
| |
Collapse
|
23
|
Garanzini D, Del Médico-Zajac MP, Calamante G. Development of Recombinant Canarypox Viruses Expressing Immunogens. Methods Mol Biol 2017; 1581:15-28. [PMID: 28374241 DOI: 10.1007/978-1-4939-6869-5_2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Canarypox viruses (CNPV) are excellent candidates to develop recombinant vector vaccines due to both their capability to induce protective immune responses and their incompetence to replicate in mammalian cells (safety profile). In addition, CNPV and the derived recombinants can be manipulated under biosafety level 1 conditions. There is no commercially available system to obtain recombinant CNPV; however, the methodology and tools required to develop recombinant vaccinia virus (VV), prototype of the Poxviridae family, can be easily adapted. This chapter provides protocols for the generation, plaque isolation, molecular characterization, amplification and purification of recombinant CNPV.
Collapse
Affiliation(s)
- Débora Garanzini
- Instituto de Biotecnología, CICVyA-INTA, N. Repetto y de los Reseros, Hurlingham, Buenos Aires, Argentina
- Instituto Nacional de Producción de Biológicos, ANLIS "Dr. Carlos G. Malbrán", Av. Vélez Sarsfield, 563, Ciudad Autónoma de Buenos Aires, Argentina
| | - María Paula Del Médico-Zajac
- Instituto de Biotecnología, CICVyA-INTA, N. Repetto y de los Reseros, Hurlingham, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Godoy Cruz, 2290, Ciudad Autónoma de Buenos Aires, Argentina
| | - Gabriela Calamante
- Instituto de Biotecnología, CICVyA-INTA, N. Repetto y de los Reseros, Hurlingham, Buenos Aires, Argentina.
| |
Collapse
|
24
|
McCoy CR, Jackson NL, Day J, Clinton SM. Genetic predisposition to high anxiety- and depression-like behavior coincides with diminished DNA methylation in the adult rat amygdala. Behav Brain Res 2016; 320:165-178. [PMID: 27965039 DOI: 10.1016/j.bbr.2016.12.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 11/22/2016] [Accepted: 12/07/2016] [Indexed: 12/17/2022]
Abstract
Understanding biological mechanisms that shape vulnerability to emotional dysfunction is critical for elucidating the neurobiology of psychiatric illnesses like anxiety and depression. To elucidate molecular and epigenetic alterations in the brain that contribute to individual differences in emotionality, our laboratory utilized a rodent model of temperamental differences. Rats bred for low response to novelty (Low Responders, LRs) are inhibited in novel situations and display high anxiety, helplessness, and diminished sociability compared to High Novelty Responder (HR) rats. Our current transcriptome profiling experiment identified widespread gene expression differences in the amygdala of adult HR/LR rats; we hypothesize that HR/LR gene expression and downstream behavioral differences stem from distinct epigenetic (specifically DNA methylation) patterning in the HR/LR brain. Although we found similar levels of DNA methyltransferase proteins in the adult HR/LR amygdala, next-generation sequencing analysis of the methylome revealed 793 differentially methylated genomic sites between the groups. Most of the differentially methylated sites were hypermethylated in HR versus LR, so we next tested the hypothesis that enhancing DNA methylation in LRs would improve their anxiety/depression-like phenotype. We found that increasing DNA methylation in LRs (via increased dietary methyl donor content) improved their anxiety-like behavior and decreased their typically high levels of Forced Swim Test (FST) immobility; however, dietary methyl donor depletion exacerbated LRs' high FST immobility. These data are generally consistent with findings in depressed patients showing that treatment with DNA methylation-promoting agents improves depressive symptoms, and highlight epigenetic mechanisms that may contribute to individual differences in risk for emotional dysfunction.
Collapse
Affiliation(s)
- Chelsea R McCoy
- School of Neuroscience, Virginia Tech University, Blacksburg, VA 24060, USA
| | - Nateka L Jackson
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham AL, USA
| | - Jeremy Day
- Department of Neurobiology, University of Alabama at Birmingham AL, USA
| | - Sarah M Clinton
- School of Neuroscience, Virginia Tech University, Blacksburg, VA 24060, USA.
| |
Collapse
|
25
|
Guillaume-Vasselin V, Lemaitre L, Dhondt KP, Tedeschi L, Poulard A, Charreyre C, Horvat B. Protection from Hendra virus infection with Canarypox recombinant vaccine. NPJ Vaccines 2016; 1:16003. [PMID: 29263849 PMCID: PMC5707888 DOI: 10.1038/npjvaccines.2016.3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Revised: 04/14/2016] [Accepted: 05/25/2016] [Indexed: 11/18/2022] Open
Abstract
Hendra virus (HeV) is an emerging zoonotic pathogen, which causes severe respiratory illness and encephalitis in humans and horses. Since its first appearance in 1994, spillovers of HeV from its natural reservoir fruit bats occur on almost an annual basis. The high mortality rate in both humans and horses and the wide-ranging reservoir distribution are making HeV a serious public health problem, especially for people exposed to sick horses. This study has aimed to develop an efficient low-cost HeV vaccine for horses based on Canarypox recombinant vector expressing HeV glycoproteins, attachment glycoprotein (G) and fusion protein (F). This vaccine was used to immunise hamsters and then challenged intraperitoneally with HeV 3 weeks later. The higher tested dose of the vaccine efficiently prevented oropharyngeal virus shedding and protected animals from clinical disease and virus-induced mortality. Vaccine induced generation of seroneutralising antibodies and prevented virus-induced histopathological changes and a production of viral RNA and antigens in animal tissues. Interestingly, some vaccinated animals, including those immunised at a lower dose, were protected in the absence of detectable specific antibodies, suggesting the induction of an efficient virus-specific cellular immunity. Finally, ponies immunised using the same vaccination protocol as hamsters developed strong seroneutralising titres against both HeV and closely related Nipah virus, indicating that this vaccine may have the ability to induce cross-protection against Henipavirus infection. These data suggest that Canarypox-based vectors encoding for HeV glycoproteins present very promising new vaccine candidate to prevent infection and shedding of the highly lethal HeV.
Collapse
Affiliation(s)
- Vanessa Guillaume-Vasselin
- CIRI, International Center for Infectiology Research, Lyon, France.,Inserm, U1111, Lyon, France.,CNRS, UMR5308, Lyon, France.,Université Lyon 1, Lyon, France.,Ecole Normale Supérieure de Lyon, Lyon, France
| | | | - Kévin P Dhondt
- CIRI, International Center for Infectiology Research, Lyon, France.,Inserm, U1111, Lyon, France.,CNRS, UMR5308, Lyon, France.,Université Lyon 1, Lyon, France.,Ecole Normale Supérieure de Lyon, Lyon, France
| | | | | | | | - Branka Horvat
- CIRI, International Center for Infectiology Research, Lyon, France.,Inserm, U1111, Lyon, France.,CNRS, UMR5308, Lyon, France.,Université Lyon 1, Lyon, France.,Ecole Normale Supérieure de Lyon, Lyon, France
| |
Collapse
|
26
|
Ramezanpour B, de Foucauld J, Kortekaas J. Emergency deployment of genetically engineered veterinary vaccines in Europe. Vaccine 2016; 34:3435-40. [PMID: 27208587 DOI: 10.1016/j.vaccine.2016.05.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 04/29/2016] [Accepted: 05/05/2016] [Indexed: 11/30/2022]
Abstract
On the 9th of November 2015, preceding the World Veterinary Vaccine Congress, a workshop was held to discuss how veterinary vaccines can be deployed more rapidly to appropriately respond to future epizootics in Europe. Considering their potential and unprecedented suitability for surge production, the workshop focussed on vaccines based on genetically engineered viruses and replicon particles. The workshop was attended by academics and representatives from leading pharmaceutical companies, regulatory experts, the European Medicines Agency and the European Commission. We here outline the present regulatory pathways for genetically engineered vaccines in Europe and describe the incentive for the organization of the pre-congress workshop. The participants agreed that existing European regulations on the deliberate release of genetically engineered vaccines into the environment should be updated to facilitate quick deployment of these vaccines in emergency situations.
Collapse
Affiliation(s)
- Bahar Ramezanpour
- Vrije Universiteit Amsterdam, Athena Institute, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | - Jean de Foucauld
- Ceva Santé Animale, 10 avenue de la Ballastière, 33500 Libourne, France
| | - Jeroen Kortekaas
- Department of Virology, Central Veterinary Institute (CVI-Lelystad), part of Wageningen University and Research Centre, P.O. Box 65, 8200 AB Lelystad, The Netherlands.
| |
Collapse
|
27
|
Status of vaccine research and development of vaccines for Nipah virus. Vaccine 2016; 34:2971-2975. [DOI: 10.1016/j.vaccine.2015.12.075] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 12/22/2015] [Indexed: 01/29/2023]
|
28
|
Yaghchi CA, Zhang Z, Alusi G, Lemoine NR, Wang Y. Vaccinia virus, a promising new therapeutic agent for pancreatic cancer. Immunotherapy 2015; 7:1249-58. [PMID: 26595180 PMCID: PMC4976866 DOI: 10.2217/imt.15.90] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The poor prognosis of pancreatic cancer patients signifies a need for radically new therapeutic strategies. Tumor-targeted oncolytic viruses have emerged as attractive therapeutic candidates for cancer treatment due to their inherent ability to specifically target and lyse tumor cells as well as induce antitumor effects by multiple action mechanisms. Vaccinia virus has several inherent features that make it particularly suitable for use as an oncolytic agent. In this review, we will discuss the potential of vaccinia virus in the management of pancreatic cancer in light of our increased understanding of cellular and immunological mechanisms involved in the disease process as well as our extending knowledge in the biology of vaccinia virus.
Collapse
Affiliation(s)
- Chadwan Al Yaghchi
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, UK
| | - Zhongxian Zhang
- National Centre for International Research in Cell & Gene Therapy, Sino-British Research Centre for Molecular Oncology, Zhengzhou University, China
| | - Ghassan Alusi
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, UK
| | - Nicholas R Lemoine
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, UK
- National Centre for International Research in Cell & Gene Therapy, Sino-British Research Centre for Molecular Oncology, Zhengzhou University, China
| | - Yaohe Wang
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, UK
- National Centre for International Research in Cell & Gene Therapy, Sino-British Research Centre for Molecular Oncology, Zhengzhou University, China
| |
Collapse
|
29
|
Deng S, Martin C, Patil R, Zhu F, Zhao B, Xiang Z, He Y. Vaxvec: The first web-based recombinant vaccine vector database and its data analysis. Vaccine 2015; 33:6938-46. [PMID: 26403370 DOI: 10.1016/j.vaccine.2015.07.113] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 07/23/2015] [Indexed: 01/12/2023]
Abstract
A recombinant vector vaccine uses an attenuated virus, bacterium, or parasite as the carrier to express a heterologous antigen(s). Many recombinant vaccine vectors and related vaccines have been developed and extensively investigated. To compare and better understand recombinant vectors and vaccines, we have generated Vaxvec (http://www.violinet.org/vaxvec), the first web-based database that stores various recombinant vaccine vectors and those experimentally verified vaccines that use these vectors. Vaxvec has now included 59 vaccine vectors that have been used in 196 recombinant vector vaccines against 66 pathogens and cancers. These vectors are classified to 41 viral vectors, 15 bacterial vectors, 1 parasitic vector, and 1 fungal vector. The most commonly used viral vaccine vectors are double-stranded DNA viruses, including herpesviruses, adenoviruses, and poxviruses. For example, Vaxvec includes 63 poxvirus-based recombinant vaccines for over 20 pathogens and cancers. Vaxvec collects 30 recombinant vector influenza vaccines that use 17 recombinant vectors and were experimentally tested in 7 animal models. In addition, over 60 protective antigens used in recombinant vector vaccines are annotated and analyzed. User-friendly web-interfaces are available for querying various data in Vaxvec. To support data exchange, the information of vaccine vectors, vaccines, and related information is stored in the Vaccine Ontology (VO). Vaxvec is a timely and vital source of vaccine vector database and facilitates efficient vaccine vector research and development.
Collapse
Affiliation(s)
- Shunzhou Deng
- Department of Veterinary Medicine, Jiangxi Agricultural University, Nanchang, Jiangxi 330045, China; Unit for Laboratory Animal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Carly Martin
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI 48109, USA
| | - Rasika Patil
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI 48109, USA
| | - Felix Zhu
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI 48109, USA
| | - Bin Zhao
- Unit for Laboratory Animal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA; School of Information, University of Michigan, Ann Arbor, MI 48109, USA
| | - Zuoshuang Xiang
- Unit for Laboratory Animal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Yongqun He
- Unit for Laboratory Animal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Center for Computational Medicine and Biology, University of Michigan, Ann Arbor, MI 48109, USA; Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
30
|
Abstract
Inactivated and attenuated vaccines have contributed to the control or even the eradication of significant animal pathogens. However, these traditional vaccine technologies have limitations and disadvantages. Inactivated vaccines lack efficacy against certain pathogens, while attenuated vaccines are not always as safe. New technology vaccines, namely DNA and recombinant viral vector vaccines, are being developed and tested against pathogens of small ruminants. These vaccines induce both humoral and cellular immune responses, are safe to manufacture and use and can be utilized in strategies for differentiation of infected from vaccinated animals. Although there are more strict regulatory requirements for the safety standards of these vaccines, once a vaccine platform is evaluated and established, effective vaccines can be rapidly produced and deployed in the field to prevent spread of emerging pathogens. The present article offers an introduction to these next generation technologies and examples of vaccines that have been tested against important diseases of sheep and goats.
Collapse
Affiliation(s)
- C S Kyriakis
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA.
| |
Collapse
|
31
|
Loomis RJ, Johnson PR. Emerging Vaccine Technologies. Vaccines (Basel) 2015; 3:429-47. [PMID: 26343196 PMCID: PMC4494353 DOI: 10.3390/vaccines3020429] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Revised: 05/05/2015] [Accepted: 05/18/2015] [Indexed: 12/13/2022] Open
Abstract
Vaccination has proven to be an invaluable means of preventing infectious diseases by reducing both incidence of disease and mortality. However, vaccines have not been effectively developed for many diseases including HIV-1, hepatitis C virus (HCV), tuberculosis and malaria, among others. The emergence of new technologies with a growing understanding of host-pathogen interactions and immunity may lead to efficacious vaccines against pathogens, previously thought impossible.
Collapse
Affiliation(s)
- Rebecca J Loomis
- The Children's Hospital of Philadelphia Research Institute, Philadelphia, PA 19104, USA.
| | - Philip R Johnson
- The Children's Hospital of Philadelphia Research Institute, Philadelphia, PA 19104, USA.
| |
Collapse
|
32
|
|
33
|
Zanetti FA, Grand MDC, Mitarotonda RC, Taboga OA, Calamante G. Canarypox virus expressing infectious bursal disease VP2 protein as immunogen for chickens. Braz J Microbiol 2014; 45:231-4. [PMID: 24948937 PMCID: PMC4059302 DOI: 10.1590/s1517-83822014000100032] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Accepted: 09/09/2013] [Indexed: 02/07/2023] Open
Abstract
Canarypox viruses (CNPV) carrying the coding sequence of VP2 protein from infectious bursal disease virus (IBDV) were obtained. These viruses were able to express VP2 protein in vitro and to induce IBDV-neutralizing antibodies when inoculated in specific pathogen-free chickens demonstrating that CNPV platform is usefulness to develop immunogens for chickens.
Collapse
Affiliation(s)
- Flavia Adriana Zanetti
- Consejo Nacional de Investigaciones Científicas y Tecnológicas Ciudad Autónoma de Buenos Aires Argentina
| | - María Daniela Conte Grand
- Consejo Nacional de Investigaciones Científicas y Tecnológicas Ciudad Autónoma de Buenos Aires Argentina
| | - Romina Cristina Mitarotonda
- Instituto de Biotecnología Centro de Investigaciones en Ciencias Veterinarias y Agronómicas Instituto Nacional de Tecnología Agropecuaria Castelar, Buenos Aires Argentina
| | - Oscar Alberto Taboga
- Consejo Nacional de Investigaciones Científicas y Tecnológicas Ciudad Autónoma de Buenos Aires Argentina ; Instituto de Biotecnología Centro de Investigaciones en Ciencias Veterinarias y Agronómicas Instituto Nacional de Tecnología Agropecuaria Castelar, Buenos Aires Argentina
| | - Gabriela Calamante
- Instituto de Biotecnología Centro de Investigaciones en Ciencias Veterinarias y Agronómicas Instituto Nacional de Tecnología Agropecuaria Castelar, Buenos Aires Argentina
| |
Collapse
|
34
|
Delany I, Rappuoli R, De Gregorio E. Vaccines for the 21st century. EMBO Mol Med 2014; 6:708-20. [PMID: 24803000 PMCID: PMC4203350 DOI: 10.1002/emmm.201403876] [Citation(s) in RCA: 285] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 03/20/2014] [Accepted: 04/07/2014] [Indexed: 12/18/2022] Open
Abstract
In the last century, vaccination has been the most effective medical intervention to reduce death and morbidity caused by infectious diseases. It is believed that vaccines save at least 2-3 million lives per year worldwide. Smallpox has been eradicated and polio has almost disappeared worldwide through global vaccine campaigns. Most of the viral and bacterial infections that traditionally affected children have been drastically reduced thanks to national immunization programs in developed countries. However, many diseases are not yet preventable by vaccination, and vaccines have not been fully exploited for target populations such as elderly and pregnant women. This review focuses on the state of the art of recent clinical trials of vaccines for major unmet medical needs such as HIV, malaria, TB, and cancer. In addition, we describe the innovative technologies currently used in vaccine research and development including adjuvants, vectors, nucleic acid vaccines, and structure-based antigen design. The hope is that thanks to these technologies, more diseases will be addressed in the 21st century by novel preventative and therapeutic vaccines.
Collapse
|
35
|
Angenvoort J, Fischer D, Fast C, Ziegler U, Eiden M, de la Fuente JG, Lierz M, Groschup MH. Limited efficacy of West Nile virus vaccines in large falcons (Falco spp.). Vet Res 2014; 45:41. [PMID: 24708385 PMCID: PMC4021075 DOI: 10.1186/1297-9716-45-41] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2013] [Accepted: 03/13/2014] [Indexed: 11/30/2022] Open
Abstract
West Nile virus (WNV) can lead to fatal diseases in raptor species. Unfortunately, there is no vaccine which has been designed specifically for use in breeding stocks of falcons. Therefore the immunogenicity and protective capacity of two commercially available WNV vaccines, both approved for use in horses, were evaluated in large falcons. One vaccine contained adjuvanted inactivated WNV lineage 1 immunogens, while the second represented a canarypox recombinant live virus vector vaccine. The efficacy of different vaccination regimes for these two vaccines was assessed serologically and by challenging the falcons with a WNV strain of homologous lineage 1. Our studies show that the recombinant vaccine conveys a slightly better protection than the inactivated vaccine, but moderate (recombinant vaccine) or weak (inactivated vaccine) side effects were observed at the injection sites. Using the recommended 2-dose regimen, both vaccines elicited only sub-optimal antibody responses and gave only partial protection following WNV challenge. Better results were obtained for both vaccines after a third dose, i.e. alleviation of clinical signs, absence of fatalities and reduction of virus shedding and viraemia. Therefore the consequences of WNV infections in falcons can be clearly alleviated by vaccination, especially if the amended triple administration scheme is used, although side effects at the vaccination site must be accepted.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Martin H Groschup
- Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Institute of Novel and Emerging Infectious Diseases, Südufer 10, 17493 Greifswald, Insel Riems, Germany.
| |
Collapse
|
36
|
Mastelic B, Garçon N, Del Giudice G, Golding H, Gruber M, Neels P, Fritzell B. Predictive markers of safety and immunogenicity of adjuvanted vaccines. Biologicals 2013; 41:458-68. [PMID: 24071553 DOI: 10.1016/j.biologicals.2013.08.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 08/29/2013] [Accepted: 08/31/2013] [Indexed: 01/08/2023] Open
Abstract
Vaccination represents one of the greatest public health triumphs; in part due to the effect of adjuvants that have been included in vaccine preparations to boost the immune responses through different mechanisms. Although a variety of novel adjuvants have been under development, only a limited number have been approved by regulatory authorities for human vaccines. This report reflects the conclusions of a group of scientists from academia, regulatory agencies and industry who attended a conference on the current state of the art in the adjuvant field. Held at the U.S. Pharmacopeial Convention (USP) in Rockville, Maryland, USA, from 18 to 19 April 2013 and organized by the International Association for Biologicals (IABS), the conference focused particularly on the future development of effective adjuvants and adjuvanted vaccines and on overcoming major hurdles, such as safety and immunogenicity assessment, as well as regulatory scrutiny. More information on the conference output can be found on the IABS website, http://www.iabs.org/.
Collapse
Affiliation(s)
- Beatris Mastelic
- WHO Center for Vaccinology and Neonatal Immunology, University of Geneva, CMU, 1 rue Michel-Servet, 1211 Geneva 4, Switzerland.
| | | | | | | | | | | | | |
Collapse
|
37
|
Draper SJ, Cottingham MG, Gilbert SC. Utilizing poxviral vectored vaccines for antibody induction-progress and prospects. Vaccine 2013; 31:4223-30. [PMID: 23746455 PMCID: PMC7131268 DOI: 10.1016/j.vaccine.2013.05.091] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Accepted: 05/22/2013] [Indexed: 02/06/2023]
Abstract
Poxviral vectors are now regarded as robust tools for B cell and antibody induction. Antibody responses can be induced against the vector as well as a transgene. Increasing application is seen in heterologous prime–boost immunization regimes. Effective veterinary poxviral vaccine products are now licensed. Promising results of antibody induction are being reported in human clinical trials.
Over the last decade, poxviral vectors emerged as a mainstay approach for the induction of T cell-mediated immunity by vaccination, and their suitability for human use has led to widespread clinical testing of candidate vectors against infectious intracellular pathogens and cancer. In contrast, poxviruses have been widely perceived in the vaccine field as a poor choice of vector for the induction of humoral immunity. However, a growing body of data, from both animal models and recent clinical trials, now suggests that these vectors can be successfully utilized to prime and boost B cells and effective antibody responses. Significant progress has been made in the context of heterologous prime–boost immunization regimes, whereby poxviruses are able to boost responses primed by other vectors, leading to the induction of high-titre antigen-specific antibody responses. In other cases, poxviral vectors have been shown to stimulate humoral immunity against both themselves and encoded transgenes, in particular viral surface proteins such as influenza haemagglutinin. In the veterinary field, recombinant poxviral vectors have made a significant impact with numerous vectors licensed for use against a variety of animal viruses. On-going studies continue to explore the potential of poxviral vectors to modulate qualitative aspects of the humoral response, as well as their amenability to adjuvantation seeking to improve quantitative antibody immunogenicity. Nevertheless, the underlying mechanisms of B cell induction by recombinant poxviruses remain poorly defined, and further work is necessary to help guide the rational optimization of future poxviral vaccine candidates aiming to induce antibodies.
Collapse
Affiliation(s)
- Simon J Draper
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Headington, Oxford OX3 7DQ, UK.
| | | | | |
Collapse
|
38
|
Cottingham MG, Carroll MW. Recombinant MVA vaccines: dispelling the myths. Vaccine 2013; 31:4247-51. [PMID: 23523407 DOI: 10.1016/j.vaccine.2013.03.021] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Accepted: 03/11/2013] [Indexed: 12/22/2022]
Abstract
Diseases such as HIV/AIDS, tuberculosis, malaria and cancer are prime targets for prophylactic or therapeutic vaccination, but have proven partially or wholly resistant to traditional approaches to vaccine design. New vaccines based on recombinant viral vectors expressing a foreign antigen are under intense development for these and other indications. One of the most advanced and most promising vectors is the attenuated, non-replicating poxvirus MVA (modified vaccinia virus Ankara), a safer derivative of the uniquely successful smallpox vaccine. Despite the ability of recombinant MVA to induce potent humoral and cellular immune responses against transgenic antigen in humans, especially when used as the latter element of a heterologous prime-boost regimen, doubts are occasionally expressed about the ultimate feasibility of this approach. In this review, five common misconceptions over recombinant MVA are discussed, and evidence is cited to show that recombinant MVA is at least sufficiently genetically stable, manufacturable, safe, and immunogenic (even in the face of prior anti-vector immunity) to warrant reasonable hope over the feasibility of large-scale deployment, should useful levels of protection against target pathogens, or therapeutic benefit for cancer, be demonstrated in efficacy trials.
Collapse
Affiliation(s)
- Matthew G Cottingham
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, OX3 7DQ, UK.
| | | |
Collapse
|
39
|
Animal Health Markets and Opportunities: Companion Animal Landscape. ADVANCES IN DELIVERY SCIENCE AND TECHNOLOGY 2013. [DOI: 10.1007/978-1-4614-4439-8_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
|
40
|
Yang DK, Kim HH, Lee KW, Song JY. The present and future of rabies vaccine in animals. Clin Exp Vaccine Res 2013; 2:19-25. [PMID: 23596586 PMCID: PMC3623496 DOI: 10.7774/cevr.2013.2.1.19] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Revised: 10/31/2012] [Accepted: 11/05/2012] [Indexed: 11/15/2022] Open
Abstract
An effective strategy for preventing rabies consists of controlling rabies in the host reservoir with vaccination. Rabies vaccine has proven to be the most effective weapon for coping with this fatal viral zoonotic disease of warm-blooded animals, including human. Natural rabies infection of an individual is always associated with exposure to rabid animals, and the duration of clinical signs can vary from days to months. The incubation period for the disease depends on the site of the bite, severity of injury, and the amount of infecting virus at the time of exposure. The mortality of untreated cases in humans is 100%. Over the last 100 years, various rabies vaccines have been developed and used to prevent or control rabies in animals, such as modified live vaccine, inactivated rabies vaccine, and oral modified live vaccine. These have proved safe and efficacious worldwide. New-generation rabies vaccines, including recombinant rabies virus-based vaccines, vectored vaccines, DNA-based vaccines, and plant vaccines, have been explored to overcome the limitations of conventional rabies vaccines. This article discusses current and next-generation rabies vaccines in animals.
Collapse
Affiliation(s)
- Dong-Kun Yang
- Viral Disease Division, Animal, Plant and Fishery Quarantine Inspection Agency, Anyang, Korea
| | - Ha-Hyun Kim
- Viral Disease Division, Animal, Plant and Fishery Quarantine Inspection Agency, Anyang, Korea
| | - Kyung-Woo Lee
- Viral Disease Division, Animal, Plant and Fishery Quarantine Inspection Agency, Anyang, Korea
| | - Jae-Young Song
- Viral Disease Division, Animal, Plant and Fishery Quarantine Inspection Agency, Anyang, Korea
| |
Collapse
|
41
|
Three-year duration of immunity in cats vaccinated with a canarypox-vectored recombinant rabies virus vaccine. Vaccine 2012; 30:6991-6. [PMID: 23059358 DOI: 10.1016/j.vaccine.2012.09.068] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Revised: 09/24/2012] [Accepted: 09/26/2012] [Indexed: 11/21/2022]
Abstract
Despite the availability of efficacious vaccines for animals and humans, rabies is still a major zoonosis. Prevention of rabies in dogs and cats is key for reducing the risk of transmission of this deadly disease to humans. Most veterinary vaccines are adjuvanted inactivated vaccines and have been shown to provide one to four-year duration of immunity. In response to debates about the safety of adjuvanted vaccines in cats, a non-adjuvanted feline rabies vaccine with one-year duration of immunity claim was specifically developed using the canarypoxvirus vector technology. The objective of this study was to validate a vaccination program based on primary vaccination, revaccination one year later and boosters every three years. Seronegative cats were vaccinated at 12 weeks of age and received a booster vaccination one year later. This vaccination regimen induced a strong and sustained antibody response, and all vaccinated animals were protected against virulent rabies challenge carried out 3 years after vaccination. These results validated 3-year duration of immunity after a complete basic vaccination program consisting in primary vaccination from 12 weeks of age followed by revaccination one year later with a non-adjuvanted canarypox-vectored vaccine.
Collapse
|
42
|
Ryan MP, Walsh G. Veterinary-based biopharmaceuticals. Trends Biotechnol 2012; 30:615-20. [PMID: 22995556 DOI: 10.1016/j.tibtech.2012.08.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Revised: 08/22/2012] [Accepted: 08/23/2012] [Indexed: 10/27/2022]
Affiliation(s)
- Michael P Ryan
- Industrial Biochemistry Program, Materials and Surface Sciences Institute, University of Limerick, Limerick, Ireland
| | | |
Collapse
|
43
|
Construction and immunogenicity of recombinant swinepox virus expressing capsid protein of PCV2. Vaccine 2012; 30:6307-13. [DOI: 10.1016/j.vaccine.2012.07.082] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Revised: 07/21/2012] [Accepted: 07/30/2012] [Indexed: 11/17/2022]
|
44
|
El Garch H, Crafford JE, Amouyal P, Durand PY, Edlund Toulemonde C, Lemaitre L, Cozette V, Guthrie A, Minke JM. An African horse sickness virus serotype 4 recombinant canarypox virus vaccine elicits specific cell-mediated immune responses in horses. Vet Immunol Immunopathol 2012; 149:76-85. [PMID: 22763149 DOI: 10.1016/j.vetimm.2012.06.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2011] [Revised: 05/30/2012] [Accepted: 06/06/2012] [Indexed: 12/24/2022]
Abstract
A recombinant canarypox virus vectored vaccine co-expressing synthetic genes encoding outer capsid proteins, VP2 and VP5, of African horse sickness virus (AHSV) serotype 4 (ALVAC(®)-AHSV4) has been demonstrated to fully protect horses against homologous challenge with virulent field virus. Guthrie et al. (2009) detected weak and variable titres of neutralizing antibody (ranging from <10 to 40) 8 weeks after vaccination leading us to hypothesize that there could be a participation of cell mediated immunity (CMI) in protection against AHSV4. The present study aimed at characterizing the CMI induced by the experimental ALVAC(®)-AHSV4 vaccine. Six horses received two vaccinations twenty-eight days apart and three horses remained unvaccinated. The detection of VP2/VP5 specific IFN-γ responses was assessed by enzyme linked immune spot (ELISpot) assay and clearly demonstrated that all ALVAC(®)-AHSV4 vaccinated horses developed significant IFN-γ production compared to unvaccinated horses. More detailed immune responses obtained by flow cytometry demonstrated that ALVAC(®)-AHSV4 vaccinations induced immune cells, mainly CD8(+) T cells, able to recognize multiple T-epitopes through all VP2 and only the N-terminus sequence of VP5. Neither VP2 nor VP5 specific IFN-γ responses were detected in unvaccinated horses. Overall, our data demonstrated that an experimental recombinant canarypox based vaccine induced significant CMI specific for both VP2 and VP5 proteins of AHSV4.
Collapse
|
45
|
Employing Live Microbes for Vaccine Delivery. DEVELOPMENT OF NOVEL VACCINES 2012. [PMCID: PMC7123214 DOI: 10.1007/978-3-7091-0709-6_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/29/2022]
|
46
|
Lin HX, Huang DY, Wang Y, Lu CP, Fan HJ. A novel vaccine against Streptococcus equi ssp. zooepidemicus infections: the recombinant swinepox virus expressing M-like protein. Vaccine 2011; 29:7027-34. [PMID: 21807055 DOI: 10.1016/j.vaccine.2011.07.074] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Revised: 06/21/2011] [Accepted: 07/08/2011] [Indexed: 11/30/2022]
Abstract
To develop a safer, more immunogenic and efficacious vaccine against Streptococcus equi ssp. zooepidemicus (SEZ) infections, the gene of M-like protein (SzP) was placed under the strong vaccinia virus promoter P28 and then inserted into swinepox virus (SPV) genome. The recombinant swinepox virus (rSPV-szp) was isolated in a non-selective medium by the co-expression of Escherichia coli LacZ gene and verified by PCR, western blotting and immunofluorescence assays. To evaluate the immunogenicity of this rSPV-szp, ICR mice were immunized with the rSPV-szp, inactivated SEZ vaccine (positive control), wild type SPV (negative control), or PBS (challenge control). All mice were intraperitoneally challenged with 5 LD(50) of homogenous ATCC 35246 strain 14 days post-vaccination. The results showed that at least 70% mice in rSPV-szp-vaccinated group were protected against homogenous ATCC 35246 challenge, the survival rate was significantly higher compared with mice in the negative control group and the challenge control group (P<0.001). The antibody titers of the rSPV-szp-vaccinated group were significantly higher (P<0.05) than the other three groups. Passive immune protection assays showed that the hyperimmune sera against M-like protein could provide mice with complete protection against challenge of ATCC 35246. Semi-quantitative RT-PCR analysis showed a marked increased in levels of IL-4 and IFN-γ mRNA in immunized mice. The results suggested that the recombinant rSPV-szp provided mice with significant protection from the SEZ infections. It is a promising candidate for the vaccine development against SEZ infections.
Collapse
Affiliation(s)
- Hui-xing Lin
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | | | | | | | | |
Collapse
|
47
|
Koppers-Lalic D, Hoeben RC. Non-human viruses developed as therapeutic agent for use in humans. Rev Med Virol 2011; 21:227-39. [PMID: 21560181 PMCID: PMC7169051 DOI: 10.1002/rmv.694] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Revised: 03/28/2011] [Accepted: 03/29/2011] [Indexed: 11/08/2022]
Abstract
Viruses usually infect a restricted set of host species, and only in rare cases does productive infection occur outside the natural host range. Infection of a new host species can manifest as a distinct disease. In this respect, the use of non‐human viruses in clinical therapy may be a cause for concern. It could provide the opportunity for the viruses to adapt to the new host and be transferred to the recipient's relatives or medical caretakers, or even to the normal host species. Such environmental impact is evidently undesirable. To forecast future clinical use of non‐human viruses, a literature study was performed to identify the viruses that are being considered for application as therapeutic agents for use in humans. Twenty‐seven non‐human virus species were identified that are in (pre)clinical development, mainly as oncolytic agents. For risk management, it is essential that the potential environmental consequences are assessed before initiating clinical use, even if the virus is not formally classified as a genetically modified organism. To aid such assessment, each of these viruses was classified in one of five relative environmental risk categories, ranging from “Negligible” to “Very High”. Canary pox virus and the Autographa californica baculovirus were assigned a “Negligible” classification, and Seneca Valley virus, murine leukemia virus, and Maraba virus to the “High” category. A complicating factor in the classification is the scarcity of publicly available information on key aspects of virus biology in some species. In such cases the relative environmental risk score was increased as a precaution. Copyright © 2011 John Wiley & Sons, Ltd.
Collapse
|
48
|
Toro, H, Suarez, DL, Tang, DCC, van Ginkel, FW, Breedlove C. Avian Influenza Mucosal Vaccination in Chickens with Replication-Defective Recombinant Adenovirus Vaccine. Avian Dis 2011; 55:43-7. [DOI: 10.1637/9516-090210-reg.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
49
|
Humoral and cell-mediated immune responses of old horses following recombinant canarypox virus vaccination and subsequent challenge infection. Vet Immunol Immunopathol 2011; 139:128-40. [DOI: 10.1016/j.vetimm.2010.09.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2010] [Revised: 09/20/2010] [Accepted: 09/28/2010] [Indexed: 01/21/2023]
|
50
|
Stittelaar KJ, Lacombe V, van Lavieren R, van Amerongen G, Simon J, Cozette V, Swayne DE, Poulet H, Osterhaus ADME. Cross-clade immunity in cats vaccinated with a canarypox-vectored avian influenza vaccine. Vaccine 2010; 28:4970-6. [PMID: 20566392 DOI: 10.1016/j.vaccine.2010.05.028] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2010] [Revised: 05/10/2010] [Accepted: 05/11/2010] [Indexed: 11/25/2022]
Abstract
Several felid species have been shown to be susceptible to infection with highly pathogenic avian influenza (HPAI) viruses of the H5N1 subtype. Infection of felids by H5N1 HPAI virus is often fatal, and cat-to-cat transmission has been documented. Domestic cats may then be involved in the transmission of infection to other animals but also to humans. A particular concern is the hypothetical role of the cat in the adaptation of the virus to mammalian species, thus increasing the pandemic risk. Therefore, the development of a HPAI vaccine for domestic cats should be considered a veterinary and also a public health priority. Here we show that vaccination of cats with a recombinant canarypox (ALVAC)(1)) virus, expressing the hemagglutinin (HA) of influenza virus A/chicken/Indonesia/03 (H5N1) confers protection against challenge infection with two antigenically distinct H5N1 virus isolates from humans. Despite low hemagglutination inhibiting (HI) antibody titers at the time of challenge, all vaccinated cats were protected against mortality and had reduced histopathological changes in the lungs. Importantly, viral shedding was reduced in vaccinated cats as compared to controls, suggesting that vaccination of cats could reduce the risk of viral transmission. In conclusion this study showed that the recombinant canarypox virus protected cats against homologous and heterologous H5N1 HPAI virus challenges.
Collapse
Affiliation(s)
- Koert J Stittelaar
- ViroClinics Biosciences B.V., Dr. Molewaterplein 50, 3015 GE Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|