1
|
Fu D, Wang W, Zhang Y, Zhang F, Yang P, Yang C, Tian Y, Yao R, Jian J, Sun Z, Zhang N, Ni Z, Rao Z, Zhao L, Guo Y. Self-assembling nanoparticle engineered from the ferritinophagy complex as a rabies virus vaccine candidate. Nat Commun 2024; 15:8601. [PMID: 39366932 PMCID: PMC11452399 DOI: 10.1038/s41467-024-52908-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 09/24/2024] [Indexed: 10/06/2024] Open
Abstract
Over the past decade, there has been a growing interest in ferritin-based vaccines due to their enhanced antigen immunogenicity and favorable safety profiles, with several vaccine candidates targeting various pathogens advancing to phase I clinical trials. Nevertheless, challenges associated with particle heterogeneity, improper assembly and unanticipated immunogenicity due to the bulky protein adaptor have impeded further advancement. To overcome these challenges, we devise a universal ferritin-adaptor delivery platform based on structural insights derived from the natural ferritinophagy complex of the human ferritin heavy chain (FTH1) and the nuclear receptor coactivator 4 (NCOA4). The engineered ferritinophagy (Fagy)-tag peptide demonstrate significantly enhanced binding affinity to the 24-mer ferritin nanoparticle, enabling efficient antigen presentation. Subsequently, we construct a self-assembling rabies virus (RABV) vaccine candidate by noncovalently conjugating the Fagy-tagged glycoprotein domain III (GDIII) of RABV to the ferritin nanoparticle, maintaining superior homogeneity, stability and immunogenicity. This vaccine candidate induces potent, rapid, and durable immune responses, and protects female mice against the authentic RABV challenge after single-dose administration. Furthermore, this universal, ferritin-based antigen conjugating strategy offers significant potential for developing vaccine against diverse pathogens and diseases.
Collapse
Affiliation(s)
- Dan Fu
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin, PR China
- College of Pharmacy, Nankai University, Tianjin, PR China
- Guangzhou Laboratory, Guangzhou, Guangdong, PR China
| | - Wenming Wang
- Key Laboratory of Chemical Biology and Molecular Engineering of the Education Ministry, Institute of Molecular Science, Shanxi University, Taiyuan, PR China
| | - Yan Zhang
- School of Public Health, Beihua University, Jilin, PR China
| | - Fan Zhang
- National Facility for Translational Medicine (Beijing), Medical Innovation Research Division, PLA General Hospital, Beijing, PR China
- Department of Oncology, The Fifth Medical Center, PLA General Hospital, Beijing, PR China
| | - Pinyi Yang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin, PR China
| | - Chun Yang
- College of Basic Medicine, Beihua University, Jilin, PR China
| | - Yufei Tian
- Changchun Veterinary Research Institute (CVRI), Chinese Academy of Agricultural Sciences (CAAS), Jingyue Economic Development Zone, Changchun, PR China
| | - Renqi Yao
- National Facility for Translational Medicine (Beijing), Medical Innovation Research Division, PLA General Hospital, Beijing, PR China
| | - Jingwu Jian
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin, PR China
| | - Zixian Sun
- Guangzhou Laboratory, Guangzhou, Guangdong, PR China
| | - Nan Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin, PR China
- Central Laboratory, Hebei Collaborative Innovation Center of Tumor Microecological Metabolism Regulation, Affiliated Hospital of Hebei University, Baoding, Hebei, PR China
| | - Zhiyu Ni
- Central Laboratory, Hebei Collaborative Innovation Center of Tumor Microecological Metabolism Regulation, Affiliated Hospital of Hebei University, Baoding, Hebei, PR China
| | - Zihe Rao
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin, PR China.
| | - Lei Zhao
- National Facility for Translational Medicine (Beijing), Medical Innovation Research Division, PLA General Hospital, Beijing, PR China.
- Department of Oncology, The Fifth Medical Center, PLA General Hospital, Beijing, PR China.
| | - Yu Guo
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin, PR China.
- Guangzhou Laboratory, Guangzhou, Guangdong, PR China.
| |
Collapse
|
2
|
Fegan JE, Waeckerlin RC, Tesfaw L, Islam EA, Deresse G, Dufera D, Assefa E, Woldemedhin W, Legesse A, Akalu M, Bayissa B, Nguyen QH, Ng D, Ahn SK, Schryvers AB, Tefera TA, Moraes TF, Gray-Owen SD. Developing a PmSLP3-based vaccine formulation that provides robust long-lasting protection against hemorrhagic septicemia-causing serogroup B and E strains of Pasteurella multocida in cattle. Front Immunol 2024; 15:1392681. [PMID: 38835751 PMCID: PMC11148319 DOI: 10.3389/fimmu.2024.1392681] [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: 02/27/2024] [Accepted: 04/19/2024] [Indexed: 06/06/2024] Open
Abstract
Background Pasteurella multocida is a bacterial pathogen that causes a variety of infections across diverse animal species, with one of the most devastating associated diseases being hemorrhagic septicemia. Outbreaks of hemorrhagic septicemia in cattle and buffaloes are marked by rapid progression and high mortality. These infections have particularly harmful socio-economic impacts on small holder farmers in Africa and Asia who are heavily reliant on a small number of animals kept as a means of subsistence for milk and draft power purposes. A novel vaccine target, PmSLP-3, has been identified on the surface of hemorrhagic septicemia-associated strains of P. multocida and was previously shown to elicit robust protection in cattle against lethal challenge with a serogroup B strain. Methods Here, we further investigate the protective efficacy of this surface lipoprotein, including evaluating the immunogenicity and protection upon formulation with a variety of adjuvants in both mice and cattle. Results PmSLP-3 formulated with Montanide ISA 61 elicited the highest level of serum and mucosal IgG, elicited long-lasting serum antibodies, and was fully protective against serogroup B challenge. Studies were then performed to identify the minimum number of doses required and the needed protein quantity to maintain protection. Duration studies were performed in cattle, demonstrating sustained serum IgG titres for 3 years after two doses of vaccine and full protection against lethal serogroup B challenge at 7 months after a single vaccine dose. Finally, a serogroup E challenge study was performed, demonstrating that PmSLP-3 vaccine can provide protection against challenge by the two serogroups responsible for hemorrhagic septicemia. Conclusion Together, these data indicate that PmSLP-3 formulated with Montanide ISA 61 is an immunogenic and protective vaccine against hemorrhagic septicemia-causing P. multocida strains in cattle.
Collapse
Affiliation(s)
- Jamie E Fegan
- Department of Molecular Genetics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Regula C Waeckerlin
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Liyuwork Tesfaw
- Department of Veterinary Bacteriology, National Veterinary Institute, Bishoftu/Debre Zeyit, Ethiopia
| | - Epshita A Islam
- Department of Biochemistry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Getaw Deresse
- Department of Veterinary Bacteriology, National Veterinary Institute, Bishoftu/Debre Zeyit, Ethiopia
| | - Dawit Dufera
- Department of Veterinary Bacteriology, National Veterinary Institute, Bishoftu/Debre Zeyit, Ethiopia
| | - Eyob Assefa
- Department of Veterinary Bacteriology, National Veterinary Institute, Bishoftu/Debre Zeyit, Ethiopia
| | - Wubet Woldemedhin
- Department of Veterinary Bacteriology, National Veterinary Institute, Bishoftu/Debre Zeyit, Ethiopia
| | - Abinet Legesse
- Department of Veterinary Bacteriology, National Veterinary Institute, Bishoftu/Debre Zeyit, Ethiopia
| | - Mirtneh Akalu
- Department of Veterinary Bacteriology, National Veterinary Institute, Bishoftu/Debre Zeyit, Ethiopia
| | - Berecha Bayissa
- Department of Veterinary Bacteriology, National Veterinary Institute, Bishoftu/Debre Zeyit, Ethiopia
| | - Quynh Huong Nguyen
- Department of Biochemistry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Dixon Ng
- Department of Biochemistry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Sang Kyun Ahn
- Department of Molecular Genetics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Anthony B Schryvers
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Takele A Tefera
- Department of Veterinary Bacteriology, National Veterinary Institute, Bishoftu/Debre Zeyit, Ethiopia
| | - Trevor F Moraes
- Department of Biochemistry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Scott D Gray-Owen
- Department of Molecular Genetics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
3
|
Mendoza-Guevara CC, Ramon-Gallegos E, Martinez-Escobar A, Alonso-Morales R, Ramos-Godinez MDP, Ortega J. Attachment and in vitro transfection efficiency of an anti-rabies Chitosan-DNA nanoparticle vaccine. IEEE Trans Nanobioscience 2021; 21:105-116. [PMID: 34170830 DOI: 10.1109/tnb.2021.3092307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In Mexico, urban rabies has been reduced during the last decade thanks to intensive canine control and vaccination campaigns; however, rabies transmitted by wild animals, especially by bats, has been increasing due to vampire bats feeding on livestock. Vampire bat populations has been controlled by culling with vampiricides, reducing indiscriminately other bat species. Hence, bat vaccination for rabies offers an alternative for culling. Nevertheless, available rabies vaccines are not suitable for their use in wildlife from emerging countries. This project presents an alternative for the use of plasmid vaccines using bio-nanotechnology, to create low-cost and accessible vaccines. To accomplish this goal, chitosan nanoparticles were synthesized by ionic gelation and conjugated by coacervation with a pDNA rabies vaccine to test their attachment efficiency. Also, the conjugate was functionalized with Protoporphyrin IX and Folic acid as biomarkers. The nanoparticles complex was characterized by ultraviolet visible spectroscopy, infrared spectroscopy, transmission electron microscopy, dynamic light scattering, and the Z potential was obtained. In vitro tests were performed on cell viability and transfection. The nanoparticles possessed a low polydispersity, a mean size of 118.5 ± 13.6 nm and a Z potential of 17.3 mV. The attachment efficiency was of 100% independent of pDNA added. In contrast to functionalized nanoparticles which showed a max attachment efficiency of 99.6% dependent of pDNA concentration and the method of functionalization. The conjugate did not influence the viability and they improved the transfection efficiency. Results suggest that these nanoparticles are easy to prepare, inexpensive, and exhibit potential for plasmid delivery as it improves transfection efficiency of pDNA vaccines.
Collapse
|
4
|
Fourie KR, Choudhary P, Ng SH, Obradovic M, Brownlie R, Anand SK, Wilson HL. Evaluation of immunogenicity and protection mediated by Lawsonia intracellularis subunit vaccines. Vet Immunol Immunopathol 2021; 237:110256. [PMID: 33971523 DOI: 10.1016/j.vetimm.2021.110256] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 04/30/2021] [Accepted: 05/02/2021] [Indexed: 02/06/2023]
Abstract
Lawsonia intracellularis is an economically important bacterium that causes ileitis in pigs. Current vaccines for L. intracellularis do not allow for differentiation between infected and vaccinated animals (DIVA), which is beneficial for disease tracking and surveillance. Previously, we identified five putative surface L. intracellularis proteins that were targeted by antibodies from pigs infected with L. intracellularis which could serve as antigens in a subunit vaccine. We conducted two trials to determine whether these antigens were immunogenic and provided protection against infectious challenge and whether truncated glycoprotein D could be used as a DIVA antigen. For Trial 1, 5 week-old piglets were administered intramuscular monovalent vaccines comprised of a recombinant (r) flagella subunit protein (rFliC,) and DIVA antigen (truncated glycoprotein D (TgD), a herpes virus antigen) both formulated with a combination adjuvant consisting of polyinosinic:polycytidylic acid(poly I:C), host defense peptide 1002 and polyphosphazene, referred to as Triple Adjuvant (TriAdj). Relative to control animals, animals vaccinated with rFliC and rTgD had significantly elevated antigen-specific humoral immunity in sera suggesting that rFliC and TgD are immunogenic. Control animals had negligible anti-TgD titres suggesting that TgD may be a suitable DIVA antigen for pigs. For Trial 2, piglets were immunized with a trivalent vaccine (FOG vaccine consisting of rFLiC, rOppA protein (a ABC Type dipeptide transport system) and rGroEL (a stress response protein)) and a divalent vaccine (CM vaccine consisting of rClpP (an ATP-dependent Clp protease proteolytic subunit) and rMetK (a S-adenosyl methionine synthase)) formulated with Emulsigen®. Relative to the control pigs, pigs immunized with the FOG vaccine produced robust and significantly higher serum IgG antibodies against rFliC and rGroEL, and significantly higher anti-FliC and anti-GroEL IgA antibodies in jejunal (GroEL only) and ileal intestinal mucosa. Pigs immunized with CM vaccine produced significantly higher serum antibodies against rClpP and rMetK and significantly higher anti-rClpP IgA antibodies in the ileum relative to the control pigs. Quantitative polymerase chain reaction (qPCR) analysis showed that 18 days after challenge with infectious L. intracellularis, challenged/control pigs and pigs that received the CM vaccine, but not the pigs vaccinated with the FOG vaccine, shed significantly more bacteria in feces than the unchallenged controls pigs. These data suggest that the FOG vaccinated pigs showed limited protection. While promising, more work is needed to enhance the efficiency of the intramuscular vaccine to show significant disease protection.
Collapse
Affiliation(s)
- Kezia R Fourie
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada; Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Pooja Choudhary
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Siew Hon Ng
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Milan Obradovic
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montréal, Montréal, Quebec, Canada
| | - Robert Brownlie
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | | | - Heather L Wilson
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada; Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, Saskatoon, Saskatchewan, Canada.
| |
Collapse
|
5
|
Immunogenicity and Protective Efficacy of a Non-Living Anthrax Vaccine versus a Live Spore Vaccine with Simultaneous Penicillin-G Treatment in Cattle. Vaccines (Basel) 2020; 8:vaccines8040595. [PMID: 33050254 PMCID: PMC7711464 DOI: 10.3390/vaccines8040595] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/29/2020] [Accepted: 10/01/2020] [Indexed: 11/17/2022] Open
Abstract
Sterne live spore vaccine (SLSV) is the current veterinary anthrax vaccine of choice. Unlike the non-living anthrax vaccine (NLAV) prototype, SLSV is incompatible with concurrent antibiotics use in an anthrax outbreak scenario. The NLAV candidates used in this study include a crude recombinant protective antigen (CrPA) and a purified recombinant protective antigen (PrPA) complemented by formalin-inactivated spores and Emulsigen-D®/Alhydrogel® adjuvants. Cattle were vaccinated twice (week 0 and 3) with NLAVs plus penicillin-G (Pen-G) treatment and compared to cattle vaccinated twice with SLSV alone and with Pen-G treatment. The immunogenicity was assessed using ELISA against rPA and FIS, toxin neutralisation assay (TNA) and opsonophagocytic assay. The protection was evaluated using an in vivo passive immunisation mouse model. The anti-rPA IgG titres for NLAVs plus Pen-G and SLSV without Pen-G treatment showed a significant increase, whereas the titres for SLSV plus Pen-G were insignificant compared to pre-vaccination values. A similar trend was measured for IgM, IgG1, and IgG2 and TNA titres (NT50) showed similar trends to anti-rPA titres across all vaccine groups. The anti-FIS IgG and IgM titres increased significantly for all vaccination groups at week 3 and 5 when compared to week 0. The spore opsonising capacity increased significantly in the NLAV vaccinated groups including Pen-G treatment and the SLSV without Pen-G but much less in the SLSV group with Pen-G treatment. Passive immunization of A/J mice challenged with a lethal dose of 34F2 spores indicated significant protective capacity of antibodies raised in the SLSV and the PrPA + FIS + adjuvants vaccinated and Pen-G treated groups but not for the NLAV with the CrPA + FIS + adjuvants and the SLSV vaccinated and Pen-G treated group. Our findings indicate that the PrPA + FIS + Emulsigen-D®/Alhydrogel® vaccine candidate may provide the same level of antibody responses and protective capacity as the SLSV. Advantageously, it can be used concurrently with Penicillin-G in an outbreak situation and as prophylactic treatment in feedlots and valuable breeding stocks.
Collapse
|
6
|
Jauro S, C. Ndumnego O, Ellis C, Buys A, Beyer W, van Heerden H. Immunogenicity of Non-Living Anthrax Vaccine Candidates in Cattle and Protective Efficacy of Immune Sera in A/J Mouse Model Compared to the Sterne Live Spore Vaccine. Pathogens 2020; 9:pathogens9070557. [PMID: 32664259 PMCID: PMC7400155 DOI: 10.3390/pathogens9070557] [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: 06/05/2020] [Revised: 06/29/2020] [Accepted: 06/29/2020] [Indexed: 11/16/2022] Open
Abstract
The Sterne live spore vaccine (SLSV, Bacillus anthracis strain 34F2) is the veterinary vaccine of choice against anthrax though contra-indicated for use with antimicrobials. However, the use of non-living anthrax vaccine (NLAV) candidates can overcome the SLSV limitation. In this study, cattle were vaccinated with either of the NLAV (purified recombinant PA (PrPA) or crude rPA (CrPA) and formaldehyde-inactivated spores (FIS of B. anthracis strain 34F2) and emulsigen-D®/alhydrogel® adjuvants) or SLSV. The immunogenicity of the NLAV and SLSV was assessed and the protective efficacies evaluated using a passive immunization mouse model. Polyclonal IgG (including the IgG1 subset) and IgM responses increased significantly across all vaccination groups after the first vaccination. Individual IgG subsets titres peaked significantly with all vaccines used after the second vaccination at week 5 and remained significant at week 12 when compared to week 0. The toxin neutralization (TNA) titres of the NLAV vaccinated cattle groups showed similar trends to those observed with the ELISA titres, except that the former were lower, but still significant, when compared to week 0. The opsonophagocytic assay indicated good antibody opsonizing responses with 75% (PrPA+FIS), 66% (CrPA+FIS) and 80% (SLSV) phagocytosis following spores opsonization. In the passive protection test, A/J mice transfused with purified IgG from cattle vaccinated with PrPA+FIS+Emulsigen-D®/Alhydrogel® and SLSV had 73% and 75% protection from challenge with B. anthracis strain 34F2 spores, respectively, whereas IgG from cattle vaccinated with CrPA+FIS+Emulsigen-D®/Alhydrogel® offered insignificant protection of 20%. There was no difference in protective immune response in cattle vaccinated twice with either the PrPA+FIS or SLSV. Moreover, PrPA+FIS did not show any residual side effects in vaccinated cattle. These results suggest that the immunogenicity and protective efficacy induced by the NLAV (PrPA+FIS) in the cattle and passive mouse protection test, respectively, are comparable to that induced by the standard SLSV.
Collapse
Affiliation(s)
- Solomon Jauro
- Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Onderstepoort, Pretoria 0110, South Africa;
- Department of Veterinary Microbiology, Faculty of Veterinary Medicine, University of Maiduguri, Maiduguri 600230, Nigeria
- Correspondence:
| | | | - Charlotte Ellis
- Design Biologix, Building 43b CSIR, Meiring Naude Road, Brummeria 0184, South Africa; (C.E.); (A.B.)
| | - Angela Buys
- Design Biologix, Building 43b CSIR, Meiring Naude Road, Brummeria 0184, South Africa; (C.E.); (A.B.)
| | - Wolfgang Beyer
- Department of Livestock Infectiology and Environmental Hygiene, Institute of Animal Science, University of Hohenheim, Stuttgart 70599, Germany;
| | - Henriette van Heerden
- Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Onderstepoort, Pretoria 0110, South Africa;
| |
Collapse
|
7
|
Galvez-Romero G, Salas-Rojas M, Pompa-Mera EN, Chávez-Rueda K, Aguilar-Setién Á. Addition of C3d-P28 adjuvant to a rabies DNA vaccine encoding the G5 linear epitope enhances the humoral immune response and confers protection. Vaccine 2017; 36:292-298. [PMID: 29191739 DOI: 10.1016/j.vaccine.2017.11.047] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 11/08/2017] [Accepted: 11/12/2017] [Indexed: 01/18/2023]
Abstract
Rabies DNA vaccines based on full-length glycoprotein (G) induce virus neutralizing antibody (VNA) responses and protect against the virus challenge. Although conformational epitopes of G are the main target of VNAs, some studies have shown that a polypeptide linear epitope G5 is also able to induce VNAs. However, a G5 DNA vaccine has not been explored. While multiple doses of DNA vaccines are required in order to confer a protective immune response, this could be overcome by the inclusion of C3d-P28, a molecular adjuvant is know to improve the antibody response in several anti-viral vaccine models. To induce and enhance the immune response against rabies in mice, we evaluated two DNA vaccines based on the linear epitope G5 of Rabies Virus (RABV) glycoprotein (pVaxG5 vaccine) and another vaccine consisting of G5 fused to the molecular adjuvant C3d-P28 (pVaxF1 vaccine). VNA responses were measured in mice immunized with both vaccines. The VNA levels from the group immunized with pVaxG5 decreased gradually, while those from the group vaccinated with pVaxF1 remained high throughout the experimental study. After challenge with 22 LD50 of the Challenge Virus Strain (CVS), the survival rate of mice immunized with pVaxG5 and pVaxF1 was increased by 27% and 50% respectively, in comparison to the PBS group. Furthermore, the in vitro proliferation of anti-rabies specific spleen CD4+ and CD8+ T cells from mice immunized with pVaxF1 was observed. Collectively, these results suggest that the linear G5 epitope is a potential candidate vaccine. Furthermore, the addition of a C3d-P28 adjuvant contributed to enhanced protection, the sustained production of VNAs, and a specific T-cell proliferative response.
Collapse
Affiliation(s)
- Guillermo Galvez-Romero
- Unidad de de Investigación Médica en Inmunología, UMAE Hospital de Pediatría, Centro Médico Nacional "Siglo XXI", Instituto Mexicano del Seguro Social, Ciudad de México, Mexico; Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Mónica Salas-Rojas
- Unidad de de Investigación Médica en Inmunología, UMAE Hospital de Pediatría, Centro Médico Nacional "Siglo XXI", Instituto Mexicano del Seguro Social, Ciudad de México, Mexico
| | - Ericka N Pompa-Mera
- Unidad de Investigación en Enfermedades Infecciosas y Parasitarias, Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, Mexico
| | - Karina Chávez-Rueda
- Unidad de de Investigación Médica en Inmunología, UMAE Hospital de Pediatría, Centro Médico Nacional "Siglo XXI", Instituto Mexicano del Seguro Social, Ciudad de México, Mexico
| | - Álvaro Aguilar-Setién
- Unidad de de Investigación Médica en Inmunología, UMAE Hospital de Pediatría, Centro Médico Nacional "Siglo XXI", Instituto Mexicano del Seguro Social, Ciudad de México, Mexico.
| |
Collapse
|
8
|
Garg R, Kaur M, Saxena A, Prasad R, Bhatnagar R. Alum adjuvanted rabies DNA vaccine confers 80% protection against lethal 50 LD 50 rabies challenge virus standard strain. Mol Immunol 2017; 85:166-173. [PMID: 28267643 DOI: 10.1016/j.molimm.2017.02.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 02/15/2017] [Accepted: 02/18/2017] [Indexed: 11/24/2022]
Abstract
Rabies is a serious concern world-wide. Despite availability of rabies vaccines for long; their efficacy, safety, availability and cost effectiveness has been a tremendous issue. This calls for improvement of rabies vaccination strategies. DNA vaccination has immense potential in this regard. The DNA vaccine pgp.LAMP-1 conferred 60% protection to BALB/c mice against 20 LD50 rabies challenge virus standard (CVS) strain challenge. Upon supplementation with Emulsigen-D, the vaccine formulation conferred complete protection against lethal challenge. To assess the feasibility of this vaccine formulation for human use, it was tested along with other FDA approved adjuvants, namely, Alum, Immuvac, Montanide ISA720 VG. Enhanced immune response correlated with high IgG antibody titer, Th2 biased response with a high level of rabies virus neutralizing antibodies (RVNAs) and IgG1/IgG2a ratio >1, observed upon alum supplementation of the rabies DNA vaccine. The total IgG antibody titer was 2IU/ml and total RVNA titer was observed to be 4IU/ml which is eight times higher than the minimum protective titer recommended by WHO. Furthermore, it conferred 80% protection against challenge with 50 LD50 of the rabies CVS strain, conducted in compliance with the potency test for rabies recommended by the National Institutes of Health (NIH), USA. Previously, we have established pre-clinical safety of this vaccine as per the guidelines of Schedule Y, FDA as well as The European Agency for evaluation of Medicinal Products. The vaccine showed no observable toxicity at the site of injection as well as at systemic level in Wistar rats when administered with 10X recommended dose. Therefore, supplementation of rabies DNA vaccine, pgp.LAMP-1 with alum would lead to development of a non-toxic, efficacious, stable and affordable vaccine that can be used to combat high numbers of fatal rabies infections tormenting developing countries.
Collapse
Affiliation(s)
- Rajni Garg
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067 Delhi, India; Amity Institute of Biotechnology, Amity University, Gurgaon (Manesar), 122413 Haryana, India
| | - Manpreet Kaur
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067 Delhi, India; Vaccine and Infectious Disease Research Center, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, 3(rd) Milestone, Faridabad-Gurgaon Expressway, Faridabad, 121001 Haryana, India
| | - Ankur Saxena
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067 Delhi, India; Fish Health Division, Diagnostic Virology Laboratory, ICAR-Directorate of Coldwater Fisheries Research, Anusandhan Bhawan, Industrial Area, Bhimtal 263136, District Nainital, Uttarakhand, India
| | - Rajendra Prasad
- Amity Institute of Biotechnology, Amity University, Gurgaon (Manesar), 122413 Haryana, India
| | - Rakesh Bhatnagar
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067 Delhi, India.
| |
Collapse
|
9
|
Zhu S, Guo C. Rabies Control and Treatment: From Prophylaxis to Strategies with Curative Potential. Viruses 2016; 8:v8110279. [PMID: 27801824 PMCID: PMC5127009 DOI: 10.3390/v8110279] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 10/17/2016] [Accepted: 10/20/2016] [Indexed: 12/25/2022] Open
Abstract
Rabies is an acute, fatal, neurological disease that affects almost all kinds of mammals. Vaccination (using an inactivated rabies vaccine), combined with administration of rabies immune globulin, is the only approved, effective method for post-exposure prophylaxis against rabies in humans. In the search for novel rabies control and treatment strategies, live-attenuated viruses have recently emerged as a practical and promising approach for immunizing and controlling rabies. Unlike the conventional, inactivated rabies vaccine, live-attenuated viruses are genetically modified viruses that are able to replicate in an inoculated recipient without causing adverse effects, while still eliciting robust and effective immune responses against rabies virus infection. A number of viruses with an intrinsic capacity that could be used as putative candidates for live-attenuated rabies vaccine have been intensively evaluated for therapeutic purposes. Additional novel strategies, such as a monoclonal antibody-based approach, nucleic acid-based vaccines, or small interfering RNAs (siRNAs) interfering with virus replication, could further add to the arena of strategies to combat rabies. In this review, we highlight current advances in rabies therapy and discuss the role that they might have in the future of rabies treatment. Given the pronounced and complex impact of rabies on a patient, a combination of these novel modalities has the potential to achieve maximal anti-rabies efficacy, or may even have promising curative effects in the future. However, several hurdles regarding clinical safety considerations and public awareness should be overcome before these approaches can ultimately become clinically relevant therapies.
Collapse
Affiliation(s)
- Shimao Zhu
- Shenzhen Weiguang Biological Products Co., Ltd., Shenzhen 518107, China.
| | - Caiping Guo
- Shenzhen Weiguang Biological Products Co., Ltd., Shenzhen 518107, China.
| |
Collapse
|
10
|
Schnee M, Vogel AB, Voss D, Petsch B, Baumhof P, Kramps T, Stitz L. An mRNA Vaccine Encoding Rabies Virus Glycoprotein Induces Protection against Lethal Infection in Mice and Correlates of Protection in Adult and Newborn Pigs. PLoS Negl Trop Dis 2016; 10:e0004746. [PMID: 27336830 PMCID: PMC4918980 DOI: 10.1371/journal.pntd.0004746] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 05/06/2016] [Indexed: 12/18/2022] Open
Abstract
Rabies is a zoonotic infectious disease of the central nervous system (CNS). In unvaccinated or untreated subjects, rabies virus infection causes severe neurological symptoms and is invariably fatal. Despite the long-standing existence of effective vaccines, vaccine availability remains insufficient, with high numbers of fatal infections mostly in developing countries. Nucleic acid based vaccines have proven convincingly as a new technology for the fast development of vaccines against newly emerging pathogens, diseases where no vaccine exists or for replacing already existing vaccines. We used an optimized non-replicating rabies virus glycoprotein (RABV-G) encoding messenger RNA (mRNA) to induce potent neutralizing antibodies (VN titers) in mice and domestic pigs. Functional antibody titers were followed in mice for up to one year and titers remained stable for the entire observation period in all dose groups. T cell analysis revealed the induction of both, specific CD4+ as well as CD8+ T cells by RABV-G mRNA, with the induced CD4+ T cells being higher than those induced by a licensed vaccine. Notably, RABV-G mRNA vaccinated mice were protected against lethal intracerebral challenge infection. Inhibition of viral replication by vaccination was verified by qRT-PCR. Furthermore, we demonstrate that CD4+ T cells are crucial for the generation of neutralizing antibodies. In domestic pigs we were able to induce VN titers that correlate with protection in adult and newborn pigs. This study demonstrates the feasibility of a non-replicating mRNA rabies vaccine in small and large animals and highlights the promises of mRNA vaccines for the prevention of infectious diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Lothar Stitz
- Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
- * E-mail: (BP); (LS)
| |
Collapse
|
11
|
Starodubova ES, Preobrazhenskaia OV, Kuzmenko YV, Latanova AA, Yarygina EI, Karpov VL. Rabies vaccines: Current status and prospects for development. Mol Biol 2015. [DOI: 10.1134/s0026893315040172] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
12
|
Grzywa R, Walczak M, Łupicka-Słowik A, Bobrek K, Boivin S, Brown EL, Gaweł A, Stefaniak T, Oleksyszyn J, Sieńczyk M. Adjuvant-dependent immunogenicity of Staphylococcus aureus Efb and Map proteins in chickens. Vet Immunol Immunopathol 2015; 166:50-6. [PMID: 26004944 DOI: 10.1016/j.vetimm.2015.04.009] [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: 01/22/2015] [Revised: 04/07/2015] [Accepted: 04/26/2015] [Indexed: 10/23/2022]
Abstract
The avian IgY antibodies generated in hens and isolated from egg yolk have gained in popularity as they present an alternative source of antibodies for diagnostic as well as therapeutic applications. One of the advantages of IgY technology are the large amounts of produced antibodies from a single animal combined with their high reactivity representing an attractive alternative for mammalian antibodies. Despite many known protocols for the immunization of chickens, the administration of new antigens often requires additional modification such as antigen dose or use of an adjuvant in order to elicit a significant immune response. We investigated the immunogenicity of three Staphylococcus aureus antigens including two extracellular proteins Map and Efb and one selected Efb105-124 epitope conjugated to KLH that were administered to the animals. Additionally, the immunization protocol included two adjuvant systems: Freund's complete adjuvant and Emulsigen-D. The results demonstrated a high immunostimulatory potency of Freund's complete adjuvant, especially in case of Efb compared to the immune response elicited by Emulsigen-D. However, after immunization with the KLH-Efb105-124 conjugate, the obtained antibodies showed similar reactivity regardless of adjuvant system used with the only exception being their avidity.
Collapse
Affiliation(s)
- Renata Grzywa
- Wroclaw University of Technology, Faculty of Chemistry, Division of Medicinal Chemistry and Microbiology, Wybrzeze Wyspianskiego 27, 50-370 Wroclaw, Poland
| | - Maciej Walczak
- Wroclaw University of Technology, Faculty of Chemistry, Division of Medicinal Chemistry and Microbiology, Wybrzeze Wyspianskiego 27, 50-370 Wroclaw, Poland
| | - Agnieszka Łupicka-Słowik
- Wroclaw University of Technology, Faculty of Chemistry, Division of Medicinal Chemistry and Microbiology, Wybrzeze Wyspianskiego 27, 50-370 Wroclaw, Poland
| | - Kamila Bobrek
- Wroclaw University of Environmental and Life Sciences, Faculty of Veterinary Medicine, Department of Epizootiology and Clinic of Bird and Exotic Animals, Pl. Grunwaldzki 45, 50-366 Wroclaw, Poland
| | - Stephane Boivin
- European Molecular Biology Laboratory (EMBL), Notkestraße 85 c/o DESY, Building 25A, 22603 Hamburg, Germany
| | - Eric L Brown
- Center for Infectious Diseases, University of Texas School of Public Health, Houston, TX 77030, USA
| | - Andrzej Gaweł
- Wroclaw University of Environmental and Life Sciences, Faculty of Veterinary Medicine, Department of Epizootiology and Clinic of Bird and Exotic Animals, Pl. Grunwaldzki 45, 50-366 Wroclaw, Poland
| | - Tadeusz Stefaniak
- Wroclaw University of Environmental and Life Sciences, Faculty of Veterinary Medicine, Department of Immunology, Pathophysiology and Veterinary Preventive Medicine, Norwida 31, 50-375 Wroclaw, Poland
| | - Józef Oleksyszyn
- Wroclaw University of Technology, Faculty of Chemistry, Division of Medicinal Chemistry and Microbiology, Wybrzeze Wyspianskiego 27, 50-370 Wroclaw, Poland
| | - Marcin Sieńczyk
- Wroclaw University of Technology, Faculty of Chemistry, Division of Medicinal Chemistry and Microbiology, Wybrzeze Wyspianskiego 27, 50-370 Wroclaw, Poland.
| |
Collapse
|
13
|
Kaur M, Garg R, Singh S, Bhatnagar R. Rabies vaccines: where do we stand, where are we heading? Expert Rev Vaccines 2014; 14:369-81. [PMID: 25348036 DOI: 10.1586/14760584.2015.973403] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Rabies being the most lethal zoonotic, vaccine-preventable viral disease with worldwide distribution of reservoir wild animals presents unique challenges for its diagnosis, management and control. Although vaccines available are highly effective, which had played the key role in controlling rabies in North America, western Europe and in a number of Asian and Latin American countries, the requirement of multiple doses along with boosters, associated cost to reduce the incidence in wild animals and prophylactic human vaccination has remained a major impediment towards achieving the same goals in poorer parts of the world such as sub-Saharan Africa and southeast Asia. Current efforts to contain rabies worldwide are directed towards the development of more safe, cheaper and efficacious vaccines along with anti-rabies antibodies for post-exposure prophylaxis. The work presented here provides an overview of the advances made towards controlling the human rabies, particularly in last 10 years, and future perspective.
Collapse
Affiliation(s)
- Manpreet Kaur
- BSL3 Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi - 110067, Delhi, India
| | | | | | | |
Collapse
|
14
|
Park ME, Lee SY, Kim RH, Ko MK, Lee KN, Kim SM, Kim BK, Lee JS, Kim B, Park JH. Enhanced immune responses of foot-and-mouth disease vaccine using new oil/gel adjuvant mixtures in pigs and goats. Vaccine 2014; 32:5221-7. [PMID: 25066738 DOI: 10.1016/j.vaccine.2014.07.040] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 06/09/2014] [Accepted: 07/09/2014] [Indexed: 10/25/2022]
Abstract
The immunity and protective capability produced by vaccines can vary remarkably according to the kinds of adjuvants being used. In the case of foot-and-mouth disease (FMD) vaccines in pigs, only oil-adjuvant vaccines have been used, and these tend to show lower immunity in pigs than in cattle. New adjuvants for these vaccines are therefore needed. We made different experimental FMD vaccines using new adjuvants (ISA 201, Carbigen, Emulsigen-D) and well-known adjuvants (ISA 206, aluminum hydroxide gel) and then conducted tests to compare the enhancement in pig immunity. More effective immune responses and protection against challenge were observed with the new adjuvants Emulsigen-D and ISA 201 compared to existing adjuvants. In the case of dairy goats, a mixture of Emulsigen-D and aluminum hydroxide gel produced rapid neutralizing antibody responses that were similar to results from tests conducted with pigs.
Collapse
Affiliation(s)
- Min-Eun Park
- Animal and Plant Quarantine Agency, 175 Anyang-ro, Manangu, Anyang city, Gyeonggido 430-757, Republic of Korea; Veterinary College, Chungnam National University, Yuseonggu, Daejeon 305-764, Republic of Korea
| | - Seo-Yong Lee
- Animal and Plant Quarantine Agency, 175 Anyang-ro, Manangu, Anyang city, Gyeonggido 430-757, Republic of Korea; Veterinary College, Chungnam National University, Yuseonggu, Daejeon 305-764, Republic of Korea
| | - Rae-Hyung Kim
- Animal and Plant Quarantine Agency, 175 Anyang-ro, Manangu, Anyang city, Gyeonggido 430-757, Republic of Korea
| | - Mi-Kyeong Ko
- Animal and Plant Quarantine Agency, 175 Anyang-ro, Manangu, Anyang city, Gyeonggido 430-757, Republic of Korea
| | - Kwang-Nyeong Lee
- Animal and Plant Quarantine Agency, 175 Anyang-ro, Manangu, Anyang city, Gyeonggido 430-757, Republic of Korea
| | - Su-Mi Kim
- Animal and Plant Quarantine Agency, 175 Anyang-ro, Manangu, Anyang city, Gyeonggido 430-757, Republic of Korea
| | - Byoung-Kwan Kim
- MJ Biologics, 1961 Premier Drive Suite 402, Mankato, MN 56001, USA
| | - Jong-Soo Lee
- Veterinary College, Chungnam National University, Yuseonggu, Daejeon 305-764, Republic of Korea
| | - Byounghan Kim
- Animal and Plant Quarantine Agency, 175 Anyang-ro, Manangu, Anyang city, Gyeonggido 430-757, Republic of Korea
| | - Jong-Hyeon Park
- Animal and Plant Quarantine Agency, 175 Anyang-ro, Manangu, Anyang city, Gyeonggido 430-757, Republic of Korea.
| |
Collapse
|
15
|
Dairy cows produce cytokine and cytotoxic T cell responses following vaccination with an antigenic fraction from Streptococcus uberis. Vet Immunol Immunopathol 2014; 160:51-60. [DOI: 10.1016/j.vetimm.2014.03.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Revised: 03/23/2014] [Accepted: 03/24/2014] [Indexed: 11/24/2022]
|
16
|
Koraka P, Bosch BJ, Cox M, Chubet R, Amerongen GV, Lövgren-Bengtsson K, Martina BEE, Roose J, Rottier PJM, Osterhaus ADME. A recombinant rabies vaccine expressing the trimeric form of the glycoprotein confers enhanced immunogenicity and protection in outbred mice. Vaccine 2014; 32:4644-50. [PMID: 24962755 DOI: 10.1016/j.vaccine.2014.06.058] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 06/02/2014] [Accepted: 06/11/2014] [Indexed: 02/07/2023]
Abstract
Rabies is a disease characterized by an invariably lethal encephalitis of viral origin that can be controlled by preventive vaccination programs of wildlife, domestic animals and humans in areas with a high risk of exposure. Currently available vaccines are expensive, cumbersome to produce and require intensive immunization and booster schemes to induce and maintain protective immunity. In the present study, we describe the development of candidate recombinant subunit rabies vaccines based on the glycoprotein G of the prototype rabies virus (RABV-G) expressed either as a monomer (RABV-mG) or in its native trimeric configuration (RABV-tG), with or without Matrix-M™ adjuvant. Immunogenicity and protective efficacy of the respective candidate vaccines were tested in outbred NIH Swiss albino mice. The RABV-tG candidate vaccine proved to be superior to the RABV-mG vaccine candidate both in terms of immunogenicity and efficacy. The relatively poor immunogenicity of the RABV-mG vaccine candidate was greatly improved by the addition of the adjuvant. A single, low dose of RABV-tG in combination with Matrix-M™ induced high levels of high avidity neutralizing antibodies and protected all mice against challenge with a lethal dose of RABV. Consequently RABV-tG used in combination with Matrix-M™ is a promising vaccine candidate that overcomes the limitations of currently used vaccines.
Collapse
Affiliation(s)
- Penelope Koraka
- Department of Viroscience, Erasmus Medical Centre, PO Box 2040, 3000 CA Rotterdam The Netherlands.
| | - Berend-Jan Bosch
- Virology Division, Department of Infectious Diseases & Immunology, Faculty of Veterinary Medicine, Utrecht University, 3508TD Utrecht, The Netherlands
| | - Manon Cox
- Protein Sciences Corp, 1000 Research Parkway, Meriden, CT 06450-7159, USA
| | - Rick Chubet
- Protein Sciences Corp, 1000 Research Parkway, Meriden, CT 06450-7159, USA
| | - Geert van Amerongen
- Department of Viroscience, Erasmus Medical Centre, PO Box 2040, 3000 CA Rotterdam The Netherlands; Institute for Translational Immunology, PO Box 450, 3720 AL Bilthoven, The Netherlands
| | | | - Byron E E Martina
- Department of Viroscience, Erasmus Medical Centre, PO Box 2040, 3000 CA Rotterdam The Netherlands
| | - Jouke Roose
- Department of Viroscience, Erasmus Medical Centre, PO Box 2040, 3000 CA Rotterdam The Netherlands
| | - Peter J M Rottier
- Virology Division, Department of Infectious Diseases & Immunology, Faculty of Veterinary Medicine, Utrecht University, 3508TD Utrecht, The Netherlands
| | - Albert D M E Osterhaus
- Department of Viroscience, Erasmus Medical Centre, PO Box 2040, 3000 CA Rotterdam The Netherlands
| |
Collapse
|
17
|
Ullas PT, Desai A, Madhusudana SN. Immunogenicity and efficacy of a plasmid DNA rabies vaccine incorporating Myd88 as a genetic adjuvant. Clin Exp Vaccine Res 2014; 3:202-11. [PMID: 25003094 PMCID: PMC4083073 DOI: 10.7774/cevr.2014.3.2.202] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 04/29/2014] [Accepted: 05/01/2014] [Indexed: 11/15/2022] Open
Abstract
PURPOSE Myeloid differentiation factor 88 (Myd88), a ubiquitous Toll-like receptor adaptor molecule, has been reported to play important roles in B cell responses to infections and vaccination. The present study evaluated the effects of genetic adjuvanting with Myd88 on the immune responses to a plasmid DNA rabies vaccine. MATERIALS AND METHODS Plasmids encoding rabies glycoprotein alone (pIRES-Rgp) or a fragment of Myd88 gene in addition (pIRES-Rgp-Myd) were constructed and administered intramuscularly or intrademally in Swiss albino mice (on days 0, 7, and 21). Rabies virus neutralizing antibody (RVNA) titres were estimated in the mice sera on days 14 and 28 by rapid fluorescent focus inhibition test. The protective efficacy of the constructs was evaluated by an intracerebral challenge with challenge virus standard virus on day 35. RESULTS Co-expression of Myd88 increased RVNA responses to pIRES-Rgp by 3- and 2-folds, following intramuscular and intradermal immunization, respectively. pIRES-Rgp protected 80% of the mice following intramuscular and intradermal immunizations, while pIRES-Rgp-Myd afforded 100% protection following similar administrations. CONCLUSION Genetic adjuvanting with Myd88 enhanced the RVNA responses and protective efficacy of a plasmid DNA rabies vaccine. This strategy might be useful for rabies vaccination of canines in the field, and needs further evaluation.
Collapse
Affiliation(s)
| | - Anita Desai
- Department of Neurovirology, National Institute of Mental Health and Neurosciences, Bangalore, Karnataka, India
| | - Shampur Narayan Madhusudana
- Department of Neurovirology, National Institute of Mental Health and Neurosciences, Bangalore, Karnataka, India
| |
Collapse
|
18
|
Blome S, Gabriel C, Beer M. Modern adjuvants do not enhance the efficacy of an inactivated African swine fever virus vaccine preparation. Vaccine 2014; 32:3879-82. [PMID: 24877766 DOI: 10.1016/j.vaccine.2014.05.051] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Revised: 05/09/2014] [Accepted: 05/15/2014] [Indexed: 11/18/2022]
Abstract
African swine fever (ASF) is among the most devastating viral diseases of pigs. In recent years, the disease has spread alarmingly. Despite intensive research activities, promising vaccine candidates are still lacking. For this reason, a study was undertaken to re-assess inactivated ASFV preparations with state-of-the-art adjuvants. Inactivated preparations of ASF virus (ASFV) "Armenia08" were adjuvanted with either Polygen™ or Emulsigen(®)-D, respectively, and used to immunize six weaner pigs two times with a three-week interval. Six weeks after the first immunization, animals were challenged with the homologues highly virulent ASFV. Although ASFV-specific antibodies were detectable in all but one vaccinated animal prior to challenge, no protective effect of immunization was observed. All animals developed acute-lethal ASF and had to be euthanized within eleven days post challenge. A slightly accelerated clinical course in vaccinees could even indicate an antibody dependent enhancement, which could also influence efficacy of other vaccine approaches.
Collapse
Affiliation(s)
- Sandra Blome
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Südufer 10, 17493 Greifswald, Insel Riems, Germany.
| | - Claudia Gabriel
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Südufer 10, 17493 Greifswald, Insel Riems, Germany.
| | - Martin Beer
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Südufer 10, 17493 Greifswald, Insel Riems, Germany.
| |
Collapse
|
19
|
Immune responses of mice against recombinant bovine herpesvirus 5 glycoprotein D. Vaccine 2014; 32:2413-9. [PMID: 24657716 DOI: 10.1016/j.vaccine.2014.03.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 03/03/2014] [Accepted: 03/07/2014] [Indexed: 11/23/2022]
Abstract
Glycoprotein D (gD) is essential for attachment and penetration of Bovine herpesvirus 5 (BoHV-5) into permissive cells, and is a major target of the host immune system, inducing strong humoral and cellular immune responses. The aim of this study was to evaluate in mice the immunogenicity of recombinant BoHV-5 gD (rgD5) expressed in Pichia pastoris. Vaccines formulated with rgD5 alone or adjuvanted with Montanide 50 ISA V2; Emulsigen or Emulsigen-DDA was administered intramuscularly or subcutaneously. Almost all formulations stimulated a humoral immune response after the first inoculation. The only exception was observed when the rgD5 was administered subcutaneously without adjuvant, in this case, the antibodies were observed after three doses. Higher titers of neutralizing antibodies were obtained with the three oil-based adjuvant formulations when compared to non-adjuvanted vaccine formulations. The rgD5 vaccine stimulated high mRNA expression levels of Th1 (INF-γ) and pro-inflammatory cytokines (IL-17, GM-CSF). The results demonstrated that the recombinant gD from BoHV-5 conserved important epitopes for viral neutralization from native BoHV-5 gD and was able to elicit mixed Th1/Th2 immune response in mice.
Collapse
|
20
|
Ullas PT, Madhusudana SN, Desai A, Sagar BKC, Jayamurugan G, Rajesh YBRD, Jayaraman N. Enhancement of immunogenicity and efficacy of a plasmid DNA rabies vaccine by nanoformulation with a fourth-generation amine-terminated poly(ether imine) dendrimer. Int J Nanomedicine 2014; 9:627-34. [PMID: 24501540 PMCID: PMC3912024 DOI: 10.2147/ijn.s53415] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Purpose Delayed onset of, and low magnitude of, protective immune responses are major drawbacks limiting the practical utility of plasmid vaccination against rabies. In this study we evaluated whether nanoformulation with the novel poly(ether imine) (PETIM) dendrimer can enhance the immunogenicity and efficacy of a plasmid-based rabies vaccine. Materials and methods A plasmid vaccine construct (pIRES-Rgp) was prepared by cloning the full-length rabies virus glycoprotein gene into pIRES vector. Drawing upon the results of our previous study, a dendriplex (dendrimer-DNA complex) of pIRES-Rgp was made with PETIM dendrimer (10:1 w/w, PETIM:pIRES-Rgp). In vitro transfection was done on baby hamster kidney (BHK)-21 cells to evaluate expression of glycoprotein gene from pIRES-Rgp and PETIM-pIRES-Rgp. Subsequently, groups of Swiss albino mice were immunized intramuscularly with pIRES-Rgp or PETIM-pIRES-Rgp. A commercially available cell culture rabies vaccine was included for comparison. Rabies virus neutralizing antibody (RVNA) titers in the immune sera were evaluated on days 14, 28, and 90 by rapid fluorescent focus inhibition test. Finally, an intracerebral challenge study using a challenge virus standard strain of rabies virus was done to evaluate the protective efficacy of the formulations. Results Protective levels of RVNA titer (≥0.5 IU/mL) were observed by day 14 in animals immunized with pIRES-Rgp and its dendriplex. Notably, PETIM-pIRES-Rgp produced 4.5-fold higher RVNA titers compared to pIRES-Rgp at this time point. All mice immunized with the PETIM-pIRES-Rgp survived the intracerebral rabies virus challenge, compared with 60% in the group which received pIRES-Rgp. Conclusion Our results suggest that nanoformulation with PETIM dendrimer can produce an earlier onset of a high-titered protective antibody response to a plasmid-based rabies vaccine. PETIM dendriplexing appears to be an efficacious nonviral delivery strategy to enhance genetic vaccination.
Collapse
Affiliation(s)
| | | | - Anita Desai
- Department of Neurovirology, National Institute of Mental Health and Neurosciences, Bangalore, India
| | | | | | | | | |
Collapse
|
21
|
Garg R, Kaur M, Saxena A, Bhatnagar R. DNA vaccination for rabies: Evaluation of preclinical safety and toxicology. ACTA ACUST UNITED AC 2014. [DOI: 10.1016/j.trivac.2014.04.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
22
|
A new rabies vaccine based on a recombinant ORF virus (parapoxvirus) expressing the rabies virus glycoprotein. J Virol 2012; 87:1618-30. [PMID: 23175365 DOI: 10.1128/jvi.02470-12] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The present study describes the generation of a new Orf virus (ORFV) recombinant, D1701-V-RabG, expressing the rabies virus (RABV) glycoprotein that is correctly presented on the surface of infected cells without the need of replication or production of infectious recombinant virus. One single immunization with recombinant ORFV can stimulate high RABV-specific virus-neutralizing antibody (VNA) titers in mice, cats, and dogs, representing all nonpermissive hosts for the ORFV vector. The protective immune response against severe lethal challenge infection was analyzed in detail in mice using different dosages, numbers, and routes for immunization with the ORFV recombinant. Long-term levels of VNA could be elicited that remained greater than 0.5 IU per ml serum, indicative for the protective status. Single applications of higher doses (10(7) PFU) can be sufficient to confer complete protection against intracranial (i.c.) challenge, whereas booster immunization was needed for protection by the application of lower dosages. Anamnestic immune responses were achieved by each of the seven tested routes of inoculation, including oral application. Finally, in vivo antibody-mediated depletion of CD4-positive and/or CD8-posititve T cell subpopulations during immunization and/or challenge infection attested the importance of CD4 T cells for the induction of protective immunity by D1701-V-RabG. This report demonstrates another example of the potential of the ORFV vector and also indicates the capability of the new recombinant for vaccination of animals.
Collapse
|
23
|
Bharati K, Vrati S. Viral Vaccines in India: An Overview. PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, INDIA. SECTION B 2012; 82:181-198. [PMID: 32226202 PMCID: PMC7100346 DOI: 10.1007/s40011-011-0014-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Accepted: 11/14/2011] [Indexed: 01/27/2023]
Abstract
Viruses cause a large number of diseases in humans, some of which are fatal, while others are highly debilitating. A majority of viral diseases attack infants and young children, while others strike people in their prime. Development of preventive measures against viral diseases is, therefore, of paramount importance. Vaccination is the most cost-effective medical intervention for preventing mortality and morbidity against infectious diseases. A number of effective and safe vaccines are currently available against several viral diseases of significant medical importance. Many of these manufactured in India, are at par with international standards and are affordable. For many other viral diseases, for which vaccines are currently not available, research is underway at various national laboratories, as well as in the private sector companies in India. The present overview highlights the various vaccine preventable viral diseases that are of special importance to India and aims to provide a glimpse of the various vaccines that are currently available, or are under development in India.
Collapse
Affiliation(s)
| | - Sudhanshu Vrati
- Vaccine and Infectious Disease Research Centre, Translational Health Science and Technology Institute, 496, Udyog Vihar Phase III, Gurgaon, 122016 India
- National Institute of Immunology, New Delhi, 110067 India
| |
Collapse
|
24
|
Ullas PT, Desai A, Madhusudana SN. Rabies DNA Vaccines: Current Status and Future. ACTA ACUST UNITED AC 2012. [DOI: 10.4236/wjv.2012.21005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|