1
|
Wu Y, Liu L, Zhang M, Zhan H, Wang C, Wang M, Chen S, Jia R, Yang Q, Zhu D, Liu M, Zhao X, Zhang S, Huang J, Ou X, Mao S, Gao Q, Sun D, Tian B, Cheng A. A Recombinant Duck Plague Virus Containing the ICP27 Deletion Marker Provides Robust Protection in Ducks. Microbiol Spectr 2023; 11:e0098323. [PMID: 37404171 PMCID: PMC10434260 DOI: 10.1128/spectrum.00983-23] [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: 03/08/2023] [Accepted: 06/12/2023] [Indexed: 07/06/2023] Open
Abstract
Duck plague virus (DPV) is a member of Alphaherpesvirus genus and poses a major threat to waterfowl breeding. Genetic engineered vaccines that are capable of distinguishing naturally infected from vaccine-immunized animals are useful for eradicating duck plague. In this study, reverse genetics was used to develop an ICP27-deficient strain (CHv-ΔICP27), and its potential as a marker vaccination candidate was evaluated. The results showed that the CHv-ΔICP27 generated in this study exhibited good genetic stability in vitro and was highly attenuated both in vivo and in vitro. The level of neutralizing antibody generated by CHv-ΔICP27 was comparable to that induced by a commercial DPV vaccine, suggesting that it could protect ducks from virulent DPV attack. By using molecular identification techniques such as PCR, restriction fragment length polymorphism, immunofluorescence, Western blotting, and others, it is possible to differentiate the CHv-ΔICP27 from wild-type strains. Moreover, ICP27 can also be a potential target for the genetic engineering vaccine development of alphavirus or perhaps the entire herpesvirus family members due to the highly conservative of ICP27 protein in all herpesvirus family members. IMPORTANCE The development of distinguishable marker vaccines from natural infection is a key step toward eradicating duck plague. Here, we generated a recombinant DPV that carries an ICP27 deletion marker that could be easily distinguished from wild-type strain by molecular biological methods. It was highly attenuated in vitro and in vivo and could provide comparable protection to ducks after a single dose of immunizations, as commercial vaccines did. Our findings support the use of the ICP27-deficient virus as a marker vaccine for DPV control and future eradication.
Collapse
Affiliation(s)
- Ying Wu
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, People’s Republic of China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, People’s Republic of China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, People’s Republic of China
| | - Lu Liu
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, People’s Republic of China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, People’s Republic of China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, People’s Republic of China
| | - Mengya Zhang
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, People’s Republic of China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, People’s Republic of China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, People’s Republic of China
| | - Haichuan Zhan
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, People’s Republic of China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, People’s Republic of China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, People’s Republic of China
| | - Chenjia Wang
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, People’s Republic of China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, People’s Republic of China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, People’s Republic of China
| | - Mingshu Wang
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, People’s Republic of China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, People’s Republic of China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, People’s Republic of China
| | - Shun Chen
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, People’s Republic of China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, People’s Republic of China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, People’s Republic of China
| | - Renyong Jia
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, People’s Republic of China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, People’s Republic of China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, People’s Republic of China
| | - Qiao Yang
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, People’s Republic of China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, People’s Republic of China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, People’s Republic of China
| | - Dekang Zhu
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, People’s Republic of China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, People’s Republic of China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang, People’s Republic of China
| | - Mafeng Liu
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, People’s Republic of China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, People’s Republic of China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, People’s Republic of China
| | - Xinxin Zhao
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, People’s Republic of China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, People’s Republic of China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, People’s Republic of China
| | - Shaqiu Zhang
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, People’s Republic of China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, People’s Republic of China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, People’s Republic of China
| | - Juan Huang
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, People’s Republic of China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, People’s Republic of China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, People’s Republic of China
| | - Xumin Ou
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, People’s Republic of China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, People’s Republic of China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, People’s Republic of China
| | - Sai Mao
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, People’s Republic of China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, People’s Republic of China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, People’s Republic of China
| | - Qun Gao
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, People’s Republic of China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, People’s Republic of China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, People’s Republic of China
| | - Di Sun
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, People’s Republic of China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, People’s Republic of China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, People’s Republic of China
| | - Bin Tian
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, People’s Republic of China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, People’s Republic of China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, People’s Republic of China
| | - Anchun Cheng
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, People’s Republic of China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, People’s Republic of China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang, People’s Republic of China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, People’s Republic of China
| |
Collapse
|
2
|
Koday MT, Leonard JA, Munson P, Forero A, Koday M, Bratt DL, Fuller JT, Murnane R, Qin S, Reinhart TA, Duus K, Messaoudi I, Hartman AL, Stefano-Cole K, Morrison J, Katze MG, Fuller DH. Multigenic DNA vaccine induces protective cross-reactive T cell responses against heterologous influenza virus in nonhuman primates. PLoS One 2017; 12:e0189780. [PMID: 29267331 PMCID: PMC5739435 DOI: 10.1371/journal.pone.0189780] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 12/01/2017] [Indexed: 01/19/2023] Open
Abstract
Recent avian and swine-origin influenza virus outbreaks illustrate the ongoing threat of influenza pandemics. We investigated immunogenicity and protective efficacy of a multi-antigen (MA) universal influenza DNA vaccine consisting of HA, M2, and NP antigens in cynomolgus macaques. Following challenge with a heterologous pandemic H1N1 strain, vaccinated animals exhibited significantly lower viral loads and more rapid viral clearance when compared to unvaccinated controls. The MA DNA vaccine induced robust serum and mucosal antibody responses but these high antibody titers were not broadly neutralizing. In contrast, the vaccine induced broadly-reactive NP specific T cell responses that cross-reacted with the challenge virus and inversely correlated with lower viral loads and inflammation. These results demonstrate that a MA DNA vaccine that induces strong cross-reactive T cell responses can, independent of neutralizing antibody, mediate significant cross-protection in a nonhuman primate model and further supports development as an effective approach to induce broad protection against circulating and emerging influenza strains.
Collapse
Affiliation(s)
- Merika T. Koday
- Department of Microbiology, University of Washington, Seattle, WA, United States of America
| | - Jolie A. Leonard
- Department of Microbiology, University of Washington, Seattle, WA, United States of America
| | - Paul Munson
- Department of Microbiology, University of Washington, Seattle, WA, United States of America
| | - Adriana Forero
- Department of Microbiology, University of Washington, Seattle, WA, United States of America
| | - Michael Koday
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States of America
| | - Debra L. Bratt
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States of America
| | - James T. Fuller
- Department of Microbiology, University of Washington, Seattle, WA, United States of America
| | - Robert Murnane
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States of America
| | - Shulin Qin
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Todd A. Reinhart
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Karen Duus
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, NY, United States of America
- Basic Sciences Department, College of Osteopathic Medicine, Touro University Nevada, Henderson, NV, United States of America
| | - Ilhem Messaoudi
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, OR, United States of America
| | - Amy L. Hartman
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Kelly Stefano-Cole
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Juliet Morrison
- Department of Microbiology, University of Washington, Seattle, WA, United States of America
| | - Michael G. Katze
- Department of Microbiology, University of Washington, Seattle, WA, United States of America
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States of America
| | - Deborah Heydenburg Fuller
- Department of Microbiology, University of Washington, Seattle, WA, United States of America
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States of America
- * E-mail:
| |
Collapse
|
3
|
Bagley KC, Schwartz JA, Andersen H, Eldridge JH, Xu R, Ota-Setlik A, Geltz JJ, Halford WP, Fouts TR. An Interleukin 12 Adjuvanted Herpes Simplex Virus 2 DNA Vaccine Is More Protective Than a Glycoprotein D Subunit Vaccine in a High-Dose Murine Challenge Model. Viral Immunol 2017; 30:178-195. [PMID: 28085634 DOI: 10.1089/vim.2016.0136] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Vaccination is a proven intervention against human viral diseases; however, success against Herpes Simplex Virus 2 (HSV-2) remains elusive. Most HSV-2 vaccines tested in humans to date contained just one or two immunogens, such as the virion attachment receptor glycoprotein D (gD) and/or the envelope fusion protein, glycoprotein B (gB). At least three factors may have contributed to the failures of subunit-based HSV-2 vaccines. First, immune responses directed against one or two viral antigens may lack sufficient antigenic breadth for efficacy. Second, the antibody responses elicited by these vaccines may have lacked necessary Fc-mediated effector functions. Third, these subunit vaccines may not have generated necessary protective cellular immune responses. We hypothesized that a polyvalent combination of HSV-2 antigens expressed from a DNA vaccine with an adjuvant that polarizes immune responses toward a T helper 1 (Th1) phenotype would compose a more effective vaccine. We demonstrate that delivery of DNA expressing full-length HSV-2 glycoprotein immunogens by electroporation with the adjuvant interleukin 12 (IL-12) generates substantially greater protection against a high-dose HSV-2 vaginal challenge than a recombinant gD subunit vaccine adjuvanted with alum and monophosphoryl lipid A (MPL). Our results further show that DNA vaccines targeting optimal combinations of surface glycoproteins provide better protection than gD alone and provide similar survival benefits and disease symptom reductions compared with a potent live attenuated HSV-2 0ΔNLS vaccine, but that mice vaccinated with HSV-2 0ΔNLS clear the virus much faster. Together, our data indicate that adjuvanted multivalent DNA vaccines hold promise for an effective HSV-2 vaccine, but that further improvements may be required.
Collapse
Affiliation(s)
| | | | | | | | - Rong Xu
- 3 Profectus Biosciences , Tarrytown, New York
| | | | - Joshua J Geltz
- 4 Department of Microbiology and Immunology, Southern Illinois University School of Medicine , Springfield, Illinois
| | - William P Halford
- 4 Department of Microbiology and Immunology, Southern Illinois University School of Medicine , Springfield, Illinois
| | | |
Collapse
|
4
|
He Z, Xu J, Tao W, Fu T, He F, Hu R, Jia L, Hong Y. A recombinant plasmid containing CpG motifs as a novel vaccine adjuvant for immune protection against herpes simplex virus 2. Mol Med Rep 2016; 14:1823-8. [PMID: 27357208 DOI: 10.3892/mmr.2016.5439] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 04/27/2016] [Indexed: 11/05/2022] Open
Abstract
The aim of the present study was to evaluate the efficacy of a herpes simplex virus type 2 (HSV-2) DNA vaccine co‑immunized with a plasmid adjuvant containing CpG motifs. A novel eukaryotic expression plasmid vector containing kanamycin resistance gene (pcDNA3Kan) was acquired from pET‑28a(+) and pcDNA3 plasmids. A gene encoding full length HSV‑2 glycoprotein D (gD) was amplified from the pcDNA3‑gD plasmid, which was cloned into pcDNA3Kan resulting in the construction of the recombinant plasmid pcDNA3Kan‑gD (pgD). A DNA segment containing 8 CpG motifs was synthesized, and cloned into pcDNA3Kan, resulting in the recombinant plasmid pcDNA3Kan‑CpG (pCpG). Mice were co‑inoculated with pgD (used as a DNA vaccine) and pCpG (used as an adjuvant) by bilateral intramuscular injection. Mice inoculated with pgD+pCpG showed higher titers of antibodies than those inoculated with the DNA vaccine alone (P<0.05). In addition, mice inoculated with pgD+pCpG showed the highest percentage of CD4+ T cells in the blood of all the groups (P﹤0.05). Thus, the present study demonstrated that pCpG could stimulate the HSV‑2 DNA vaccine to induce a stronger cell‑mediated immune response than the DNA vaccine alone. The aim of the present study was to evaluate the efficacy of a HSV‑2 DNA vaccine (pgD) co‑immunized with a plasmid adjuvant containing CpG motifs (pCpG). Whether the pCpG would be able to stimulate the pgD to induce a stronger immune response compared with pgD alone.
Collapse
Affiliation(s)
- Zhuojing He
- Institute of Bioengineering, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| | - Juan Xu
- Institute of Hygiene, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| | - Wei Tao
- Institute of Bioengineering, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| | - Ting Fu
- Institute of Bioengineering, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| | - Fang He
- Institute of Bioengineering, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| | - Ruxi Hu
- Institute of Bioengineering, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| | - Lan Jia
- Institute of Bioengineering, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| | - Yan Hong
- Institute of Bioengineering, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| |
Collapse
|
5
|
Bagley K, Xu R, Ota-Setlik A, Egan M, Schwartz J, Fouts T. The catalytic A1 domains of cholera toxin and heat-labile enterotoxin are potent DNA adjuvants that evoke mixed Th1/Th17 cellular immune responses. Hum Vaccin Immunother 2016; 11:2228-40. [PMID: 26042527 PMCID: PMC4635876 DOI: 10.1080/21645515.2015.1026498] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
DNA encoded adjuvants are well known for increasing the magnitude of cellular and/or humoral immune responses directed against vaccine antigens. DNA adjuvants can also tune immune responses directed against vaccine antigens to better protect against infection of the target organism. Two potent DNA adjuvants that have unique abilities to tune immune responses are the catalytic A1 domains of Cholera Toxin (CTA1) and Heat-Labile Enterotoxin (LTA1). Here, we have characterized the adjuvant activities of CTA1 and LTA1 using HIV and SIV genes as model antigens. Both of these adjuvants enhanced the magnitude of antigen-specific cellular immune responses on par with those induced by the well-characterized cytokine adjuvants IL-12 and GM-CSF. CTA1 and LTA1 preferentially enhanced cellular responses to the intracellular antigen SIVmac239-gag over those for the secreted HIVBaL-gp120 antigen. IL-12, GM-CSF and electroporation did the opposite suggesting differences in the mechanisms of actions of these diverse adjuvants. Combinations of CTA1 or LTA1 with IL-12 or GM-CSF generated additive and better balanced cellular responses to both of these antigens. Consistent with observations made with the holotoxin and the CTA1-DD adjuvant, CTA1 and LTA1 evoked mixed Th1/Th17 cellular immune responses. Together, these results show that CTA1 and LTA1 are potent DNA vaccine adjuvants that favor the intracellular antigen gag over the secreted antigen gp120 and evoke mixed Th1/Th17 responses against both of these antigens. The results also indicate that achieving a balanced immune response to multiple intracellular and extracellular antigens delivered via DNA vaccination may require combining adjuvants that have different and complementary mechanisms of action.
Collapse
|
6
|
Topical herpes simplex virus 2 (HSV-2) vaccination with human papillomavirus vectors expressing gB/gD ectodomains induces genital-tissue-resident memory CD8+ T cells and reduces genital disease and viral shedding after HSV-2 challenge. J Virol 2014; 89:83-96. [PMID: 25320297 DOI: 10.1128/jvi.02380-14] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
UNLABELLED No herpes simplex virus 2 (HSV-2) vaccine has been licensed for use in humans. HSV-2 glycoproteins B (gB) and D (gD) are targets of neutralizing antibodies and T cells, but clinical trials involving intramuscular (i.m.) injection of HSV-2 gB and gD in adjuvants have not been effective. Here we evaluated intravaginal (ivag) genetic immunization of C57BL/6 mice with a replication-defective human papillomavirus pseudovirus (HPV PsV) expressing HSV-2 gB (HPV-gB) or gD (HPV-gD) constructs to target different subcellular compartments. HPV PsV expressing a secreted ectodomain of gB (gBsec) or gD (gDsec), but not PsV expressing a cytoplasmic or membrane-bound form, induced circulating and intravaginal-tissue-resident memory CD8(+) T cells that were able to secrete gamma interferon (IFN-γ) and tumor necrosis factor alpha (TNF-α) as well as moderate levels of serum HSV neutralizing antibodies. Combined immunization with HPV-gBsec and HPV-gDsec (HPV-gBsec/gDsec) vaccines conferred longer survival after vaginal challenge with HSV-2 than immunization with HPV-gBsec or HPV-gDsec alone. HPV-gBsec/gDsec ivag vaccination was associated with a reduced severity of genital lesions and lower levels of viral shedding in the genital tract after HSV-2 challenge. In contrast, intramuscular vaccination with a soluble truncated gD protein (gD2t) in alum and monophosphoryl lipid A (MPL) elicited high neutralizing antibody titers and improved survival but did not reduce genital lesions and viral shedding. Vaccination combining ivag HPV-gBsec/gDsec and i.m. gD2t-alum-MPL improved survival and reduced genital lesions and viral shedding. Finally, high levels of circulating HSV-2-specific CD8(+) T cells, but not serum antibodies, correlated with reduced viral shedding. Taken together, our data underscore the potential of HPV PsV as a platform for a topical mucosal vaccine to control local manifestations of primary HSV-2 infection. IMPORTANCE Genital herpes is a highly prevalent chronic disease caused by HSV infection. To date, there is no licensed vaccine against HSV infection. This study describes intravaginal vaccination with a nonreplicating HPV-based vector expressing HSV glycoprotein antigens. The data presented in this study underscore the potential of HPV-based vectors as a platform for the induction of genital-tissue-resident memory T cell responses and the control of local manifestations of primary HSV infection.
Collapse
|
7
|
Sicurella M, Nicoli F, Gallerani E, Volpi I, Berto E, Finessi V, Destro F, Manservigi R, Cafaro A, Ensoli B, Caputo A, Gavioli R, Marconi PC. An attenuated herpes simplex virus type 1 (HSV1) encoding the HIV-1 Tat protein protects mice from a deadly mucosal HSV1 challenge. PLoS One 2014; 9:e100844. [PMID: 25033084 PMCID: PMC4102458 DOI: 10.1371/journal.pone.0100844] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 05/30/2014] [Indexed: 12/22/2022] Open
Abstract
Herpes simplex virus types 1 and 2 (HSV1 and HSV2) are common infectious agents in both industrialized and developing countries. They cause recurrent asymptomatic and/or symptomatic infections, and life-threatening diseases and death in newborns and immunocompromised patients. Current treatment for HSV relies on antiviral medications, which can halt the symptomatic diseases but cannot prevent the shedding that occurs in asymptomatic patients or, consequently, the spread of the viruses. Therefore, prevention rather than treatment of HSV infections has long been an area of intense research, but thus far effective anti-HSV vaccines still remain elusive. One of the key hurdles to overcome in anti-HSV vaccine development is the identification and effective use of strategies that promote the emergence of Th1-type immune responses against a wide range of epitopes involved in the control of viral replication. Since the HIV1 Tat protein has several immunomodulatory activities and increases CTL recognition of dominant and subdominant epitopes of heterologous antigens, we generated and assayed a recombinant attenuated replication-competent HSV1 vector containing the tat gene (HSV1-Tat). In this proof-of-concept study we show that immunization with this vector conferred protection in 100% of mice challenged intravaginally with a lethal dose of wild-type HSV1. We demonstrate that the presence of Tat within the recombinant virus increased and broadened Th1-like and CTL responses against HSV-derived T-cell epitopes and elicited in most immunized mice detectable IgG responses. In sharp contrast, a similarly attenuated HSV1 recombinant vector without Tat (HSV1-LacZ), induced low and different T cell responses, no measurable antibody responses and did not protect mice against the wild-type HSV1 challenge. These findings strongly suggest that recombinant HSV1 vectors expressing Tat merit further investigation for their potential to prevent and/or contain HSV1 infection and dissemination.
Collapse
Affiliation(s)
- Mariaconcetta Sicurella
- Department of Life Sciences and Biotechnology, Section of Applied Microbiology and Pathology, University of Ferrara, Ferrara, Italy
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Francesco Nicoli
- Department of Life Sciences and Biotechnology, Section of Biochemistry and Molecular Biology, University of Ferrara, Ferrara, Italy
| | - Eleonora Gallerani
- Department of Life Sciences and Biotechnology, Section of Biochemistry and Molecular Biology, University of Ferrara, Ferrara, Italy
| | - Ilaria Volpi
- Department of Life Sciences and Biotechnology, Section of Applied Microbiology and Pathology, University of Ferrara, Ferrara, Italy
| | - Elena Berto
- Department of Life Sciences and Biotechnology, Section of Applied Microbiology and Pathology, University of Ferrara, Ferrara, Italy
| | - Valentina Finessi
- Department of Life Sciences and Biotechnology, Section of Biochemistry and Molecular Biology, University of Ferrara, Ferrara, Italy
| | - Federica Destro
- Department of Life Sciences and Biotechnology, Section of Biochemistry and Molecular Biology, University of Ferrara, Ferrara, Italy
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Roberto Manservigi
- Department of Life Sciences and Biotechnology, Section of Applied Microbiology and Pathology, University of Ferrara, Ferrara, Italy
| | - Aurelio Cafaro
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy
| | - Barbara Ensoli
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy
| | - Antonella Caputo
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Riccardo Gavioli
- Department of Life Sciences and Biotechnology, Section of Biochemistry and Molecular Biology, University of Ferrara, Ferrara, Italy
| | - Peggy C. Marconi
- Department of Life Sciences and Biotechnology, Section of Applied Microbiology and Pathology, University of Ferrara, Ferrara, Italy
| |
Collapse
|
8
|
Azizi A, Tang M, Gisonni-Lex L, Mallet L. Evaluation of infectious titer in a candidate HSV type 2 vaccine by a quantitative molecular approach. BMC Microbiol 2013; 13:284. [PMID: 24313978 PMCID: PMC3878963 DOI: 10.1186/1471-2180-13-284] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 12/04/2013] [Indexed: 11/29/2022] Open
Abstract
Background One of the critical tasks in analytical testing is to monitor and assign the infectivity or potency of viral based vaccines from process development to production of final clinical lots. In this study, a high throughput RT-qPCR based approach was developed to evaluate the infectious titre in a replication-defective HSV-2 candidate vaccine, called HSV529. This assay is a combination of viral propagation and quantitative RT-PCR which measures the amount of RNA in infected cells after incubation with test samples. Results The relative infectious titre of HSV529 candidate vaccine was determined by a RT-qPCR method targeting HSV-2 gD2 gene. The data were analyzed using the parallel-line analysis as described in the European Pharmacopoeia 8th edition. The stability of HSV529 test samples were also investigated in a concordance study between RT-qPCR infectivity assay and a classical plaque assays. A suitable correlation was determined between both assays using an identical sample set in both assays. The RT-qPCR infectivity assay was further characterized by evaluating the intermediate precision and accuracy. The coefficient of variation from the six independent assays was less than 10%. The accuracy of each of the assay was also evaluated in the range of 92.91% to 120.57%. Conclusions Our data demonstrate that the developed RT-qPCR infectivity assay is a rapid high throughput approach to quantify the infectious titer or potency of live attenuated or defective viral-based vaccines, an attribute which is associated with product quality.
Collapse
Affiliation(s)
- Ali Azizi
- Microbiology & Virology Platform, Department of Analytical Research & Development North America, Sanofi Pasteur, 1755 Steeles Avenue West, Toronto, Ontario M2R 3 T4, Canada.
| | | | | | | |
Collapse
|
9
|
Platt RJ, Khodai T, Townend TJ, Bright HH, Cockle P, Perez-Tosar L, Webster R, Champion B, Hickling TP, Mirza F. CD8+ T Lymphocyte Epitopes From The Herpes Simplex Virus Type 2 ICP27, VP22 and VP13/14 Proteins To Facilitate Vaccine Design And Characterization. Cells 2013; 2:19-42. [PMID: 24709642 PMCID: PMC3972665 DOI: 10.3390/cells2010019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 11/22/2012] [Accepted: 12/27/2012] [Indexed: 11/16/2022] Open
Abstract
CD8+ T cells have the potential to control HSV-2 infection. However, limited information has been available on CD8+ T cell epitopes or the functionality of antigen specific T cells during infection or following immunization with experimental vaccines. Peptide panels from HSV-2 proteins ICP27, VP22 and VP13/14 were selected from in silico predictions of binding to human HLA-A*0201 and mouse H-2Kd, Ld and Dd molecules. Nine previously uncharacterized CD8+ T cell epitopes were identified from HSV-2 infected BALB/c mice. HSV-2 specific peptide sequences stabilized HLA-A*02 surface expression with intermediate or high affinity binding. Peptide specific CD8+ human T cell lines from peripheral blood lymphocytes were generated from a HLA-A*02+ donor. High frequencies of peptide specific CD8+ T cell responses were elicited in mice by DNA vaccination with ICP27, VP22 and VP13/14, as demonstrated by CD107a mobilization. Vaccine driven T cell responses displayed a more focused immune response than those induced by viral infection. Furthermore, vaccination with ICP27 reduced viral shedding and reduced the clinical impact of disease. In conclusion, this study describes novel HSV-2 epitopes eliciting strong CD8+ T cell responses that may facilitate epitope based vaccine design and aid immunomonitoring of antigen specific T cell frequencies in preclinical and clinical settings.
Collapse
Affiliation(s)
- Rebecca J Platt
- Biotherapeutics and Translational Research, Pharmacokinetics, Dynamics & Metabolism, Pfizer Global Research and Development, Sandwich, Kent, CT13-9NJ, UK.
| | - Tansi Khodai
- New Opportunities Unit, Pfizer Global Research and Development, Sandwich, Kent, CT13 -9NJ, UK.
| | - Tim J Townend
- Biotherapeutics and Translational Research, Pharmacokinetics, Dynamics & Metabolism, Pfizer Global Research and Development, Sandwich, Kent, CT13-9NJ, UK.
| | - Helen H Bright
- New Opportunities Unit, Pfizer Global Research and Development, Sandwich, Kent, CT13 -9NJ, UK.
| | - Paul Cockle
- Vaccine Research Unit, Pfizer Global Research and Development, Sandwich, Kent, CT13-9NJ, UK.
| | - Luis Perez-Tosar
- Biotherapeutics and Translational Research, Pharmacokinetics, Dynamics & Metabolism, Pfizer Global Research and Development, Sandwich, Kent, CT13-9NJ, UK.
| | - Rob Webster
- Biotherapeutics and Translational Research, Pharmacokinetics, Dynamics & Metabolism, Pfizer Global Research and Development, Sandwich, Kent, CT13-9NJ, UK.
| | - Brian Champion
- Vaccine Research Unit, Pfizer Global Research and Development, Sandwich, Kent, CT13-9NJ, UK.
| | - Timothy P Hickling
- Biotherapeutics and Translational Research, Pharmacokinetics, Dynamics & Metabolism, Pfizer Global Research and Development, Sandwich, Kent, CT13-9NJ, UK.
| | - Fareed Mirza
- Biotherapeutics and Translational Research, Pharmacokinetics, Dynamics & Metabolism, Pfizer Global Research and Development, Sandwich, Kent, CT13-9NJ, UK.
| |
Collapse
|
10
|
Bright H, Perez DL, Christy C, Cockle P, Eyles JE, Hammond D, Khodai T, Lang S, West K, Loudon PT. The efficacy of HSV-2 vaccines based on gD and gB is enhanced by the addition of ICP27. Vaccine 2012; 30:7529-35. [PMID: 23103198 DOI: 10.1016/j.vaccine.2012.10.046] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 09/26/2012] [Accepted: 10/13/2012] [Indexed: 01/08/2023]
Abstract
DNA vaccines expressing HSV-2 gD, gB, ICP27, VP22 and VP13/14 were shown to be immunogenic in mice; gD and gB elicited neutralising antibody, and all five antigens induced T cell responses measured by IFNγ ELISPOT. In murine HSV-2 challenge studies, gD and gB provided moderate to high levels of protection while ICP27 provided a lower level of protection depending on the model (intravaginal or intranasal) and the challenge dose. Combining vaccines expressing gB or gD with vaccines expressing ICP27 provided greater protection than any antigen alone. We conclude that the addition of ICP27 to enhance the anti-viral T cell response can improve the efficacy of gD- and gB-based vaccines.
Collapse
|
11
|
Golden JW, Josleyn M, Mucker EM, Hung CF, Loudon PT, Wu TC, Hooper JW. Side-by-side comparison of gene-based smallpox vaccine with MVA in nonhuman primates. PLoS One 2012; 7:e42353. [PMID: 22860117 PMCID: PMC3409187 DOI: 10.1371/journal.pone.0042353] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Accepted: 07/04/2012] [Indexed: 11/25/2022] Open
Abstract
Orthopoxviruses remain a threat as biological weapons and zoonoses. The licensed live-virus vaccine is associated with serious health risks, making its general usage unacceptable. Attenuated vaccines are being developed as alternatives, the most advanced of which is modified-vaccinia virus Ankara (MVA). We previously developed a gene-based vaccine, termed 4pox, which targets four orthopoxvirus antigens, A33, B5, A27 and L1. This vaccine protects mice and non-human primates from lethal orthopoxvirus disease. Here, we investigated the capacity of the molecular adjuvants GM-CSF and Escherichia coli heat-labile enterotoxin (LT) to enhance the efficacy of the 4pox gene-based vaccine. Both adjuvants significantly increased protective antibody responses in mice. We directly compared the 4pox plus LT vaccine against MVA in a monkeypox virus (MPXV) nonhuman primate (NHP) challenge model. NHPs were vaccinated twice with MVA by intramuscular injection or the 4pox/LT vaccine delivered using a disposable gene gun device. As a positive control, one NHP was vaccinated with ACAM2000. NHPs vaccinated with each vaccine developed anti-orthopoxvirus antibody responses, including those against the 4pox antigens. After MPXV intravenous challenge, all control NHPs developed severe disease, while the ACAM2000 vaccinated animal was well protected. All NHPs vaccinated with MVA were protected from lethality, but three of five developed severe disease and all animals shed virus. All five NHPs vaccinated with 4pox/LT survived and only one developed severe disease. None of the 4pox/LT-vaccinated animals shed virus. Our findings show, for the first time, that a subunit orthopoxvirus vaccine delivered by the same schedule can provide a degree of protection at least as high as that of MVA.
Collapse
Affiliation(s)
- Joseph W. Golden
- Department of Molecular Virology, Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Matthew Josleyn
- Department of Molecular Virology, Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Eric M. Mucker
- Department of Viral Therapeutics, Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Chien-Fu Hung
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States of America
| | - Peter T. Loudon
- Pfizer, Sandwich Laboratories, Sandwich, Kent, United Kingdom
| | - T. C. Wu
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States of America
| | - Jay W. Hooper
- Department of Molecular Virology, Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
- * E-mail:
| |
Collapse
|
12
|
Fuller DH, Rajakumar P, Che JW, Narendran A, Nyaundi J, Michael H, Yager EJ, Stagnar C, Wahlberg B, Taber R, Haynes JR, Cook FC, Ertl P, Tite J, Amedee AM, Murphey-Corb M. Therapeutic DNA vaccine induces broad T cell responses in the gut and sustained protection from viral rebound and AIDS in SIV-infected rhesus macaques. PLoS One 2012; 7:e33715. [PMID: 22442716 PMCID: PMC3307760 DOI: 10.1371/journal.pone.0033715] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Accepted: 02/15/2012] [Indexed: 11/18/2022] Open
Abstract
Immunotherapies that induce durable immune control of chronic HIV infection may eliminate the need for life-long dependence on drugs. We investigated a DNA vaccine formulated with a novel genetic adjuvant that stimulates immune responses in the blood and gut for the ability to improve therapy in rhesus macaques chronically infected with SIV. Using the SIV-macaque model for AIDS, we show that epidermal co-delivery of plasmids expressing SIV Gag, RT, Nef and Env, and the mucosal adjuvant, heat-labile E. coli enterotoxin (LT), during antiretroviral therapy (ART) induced a substantial 2-4-log fold reduction in mean virus burden in both the gut and blood when compared to unvaccinated controls and provided durable protection from viral rebound and disease progression after the drug was discontinued. This effect was associated with significant increases in IFN-γ T cell responses in both the blood and gut and SIV-specific CD8+ T cells with dual TNF-α and cytolytic effector functions in the blood. Importantly, a broader specificity in the T cell response seen in the gut, but not the blood, significantly correlated with a reduction in virus production in mucosal tissues and a lower virus burden in plasma. We conclude that immunizing with vaccines that induce immune responses in mucosal gut tissue could reduce residual viral reservoirs during drug therapy and improve long-term treatment of HIV infection in humans.
Collapse
Affiliation(s)
- Deborah Heydenburg Fuller
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Albany Medical College, Albany, New York, United States of America
- PowderJect Vaccines, Inc., Madison, Wisconsin, United States of America
| | - Premeela Rajakumar
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Jenny W. Che
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- PowderJect Vaccines, Inc., Madison, Wisconsin, United States of America
| | - Amithi Narendran
- Albany Medical College, Albany, New York, United States of America
| | - Julia Nyaundi
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Heather Michael
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Eric J. Yager
- Albany Medical College, Albany, New York, United States of America
| | - Cristy Stagnar
- Albany Medical College, Albany, New York, United States of America
| | - Brendon Wahlberg
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Rachel Taber
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Joel R. Haynes
- PowderJect Vaccines, Inc., Madison, Wisconsin, United States of America
| | | | - Peter Ertl
- GlaxoSmithKline, Stevenage, United Kingdom
| | - John Tite
- GlaxoSmithKline, Stevenage, United Kingdom
| | - Angela M. Amedee
- Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
| | - Michael Murphey-Corb
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
13
|
Laing KJ, Dong L, Sidney J, Sette A, Koelle DM. Immunology in the Clinic Review Series; focus on host responses: T cell responses to herpes simplex viruses. Clin Exp Immunol 2012; 167:47-58. [PMID: 22132884 PMCID: PMC3248086 DOI: 10.1111/j.1365-2249.2011.04502.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2011] [Indexed: 01/04/2023] Open
Abstract
Herpes virus infections are chronic and co-exist with acquired immune responses that generally prevent severe damage to the host, while allowing periodic shedding of virus and maintenance of its transmission in the community. Herpes simplex viruses type 1 and 2 (HSV-1, HSV-2) are typical in this regard and are representative of the viral subfamily Alphaherpesvirinae, which has a tropism for neuronal and epithelial cells. This review will emphasize recent progress in decoding the physiologically important CD8(+) and CD4(+) T cell responses to HSV in humans. The expanding data set is discussed in the context of the search for an effective HSV vaccine as therapy for existing infections and to prevent new infections.
Collapse
Affiliation(s)
- K J Laing
- Department of Medicine, University of Washington, Seattle, WA, USA
| | | | | | | | | |
Collapse
|
14
|
Bi J, Song R, Yang H, Li B, Fan J, Liu Z, Long C. Stepwise identification of HLA-A*0201-restricted CD8+ T-cell epitope peptides from herpes simplex virus type 1 genome boosted by a StepRank scheme. Biopolymers 2011; 96:328-39. [PMID: 21072852 DOI: 10.1002/bip.21564] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Identification of immunodominant epitopes is the first step in the rational design of peptide vaccines aimed at T-cell immunity. To date, however, it is yet a great challenge for accurately predicting the potent epitope peptides from a pool of large-scale candidates with an efficient manner. In this study, a method that we named StepRank has been developed for the reliable and rapid prediction of binding capabilities/affinities between proteins and genome-wide peptides. In this procedure, instead of single strategy used in most traditional epitope identification algorithms, four steps with different purposes and thus different computational demands are employed in turn to screen the large-scale peptide candidates that are normally generated from, for example, pathogenic genome. The steps 1 and 2 aim at qualitative exclusion of typical nonbinders by using empirical rule and linear statistical approach, while the steps 3 and 4 focus on quantitative examination and prediction of the interaction energy profile and binding affinity of peptide to target protein via quantitative structure-activity relationship (QSAR) and structure-based free energy analysis. We exemplify this method through its application to binding predictions of the peptide segments derived from the 76 known open-reading frames (ORFs) of herpes simplex virus type 1 (HSV-1) genome with or without affinity to human major histocompatibility complex class I (MHC I) molecule HLA-A*0201, and find that the predictive results are well compatible with the classical anchor residue theory and perfectly match for the extended motif pattern of MHC I-binding peptides. The putative epitopes are further confirmed by comparisons with 11 experimentally measured HLA-A*0201-restrcited peptides from the HSV-1 glycoproteins D and K. We expect that this well-designed scheme can be applied in the computational screening of other viral genomes as well.
Collapse
Affiliation(s)
- Jianjun Bi
- Department of Dermatology, General Hospital of Guangzhou Military Command of PLA, Guangzhou, China
| | | | | | | | | | | | | |
Collapse
|
15
|
Single and combination herpes simplex virus type 2 glycoprotein vaccines adjuvanted with CpG oligodeoxynucleotides or monophosphoryl lipid A exhibit differential immunity that is not correlated to protection in animal models. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2011; 18:1702-9. [PMID: 21852545 DOI: 10.1128/cvi.05071-11] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Despite several attempts to develop an effective prophylactic vaccine for HSV-2, all have failed to show efficacy in the clinic. The most recent of these failures was the GlaxoSmithKline (GSK) subunit vaccine based on the glycoprotein gD with the adjuvant monophosphoryl lipid A (MPL). In a phase 3 clinical trial, this vaccine failed to protect from HSV-2 disease, even though good neutralizing antibody responses were elicited. We aimed to develop a superior, novel HSV-2 vaccine containing either gD or gB alone or in combination, together with the potent adjuvant CpG oligodeoxynucleotides (CPG). The immunogenic properties of these vaccines were compared in mice. We show that gB/CPG/alum elicited a neutralizing antibody response similar to that elicited by gD/CPG/alum vaccine but a significantly greater gamma interferon (IFN-γ) T cell response. Furthermore, the combined gB-gD/CPG/alum vaccine elicited significantly greater neutralizing antibody and T cell responses than gD/MPL/alum. The efficacies of these candidate vaccines were compared in the mouse and guinea pig disease models, including a novel male guinea pig genital disease model. These studies demonstrated that increased immune response did not correlate to improved protection. First, despite a lower IFN-γ T cell response, the gD/CPG/alum vaccine was more effective than gB/CPG/alum in mice. Furthermore, the gB-gD/CPG/alum vaccine was no more effective than gD/MPL/alum in mice or male guinea pigs. We conclude that difficulties in correlating immune responses to efficacy in animal models will act as a deterrent to researchers attempting to develop effective HSV vaccines.
Collapse
|
16
|
Adjuvant activity of the catalytic A1 domain of cholera toxin for retroviral antigens delivered by GeneGun. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2011; 18:922-30. [PMID: 21508173 DOI: 10.1128/cvi.05019-11] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Most DNA-encoded adjuvants enhance immune responses to DNA vaccines in small animals but are less effective in primates. Here, we characterize the adjuvant activity of the catalytic A1 domain of cholera toxin (CTA1) for human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV) antigens in mice and macaques delivered by GeneGun. The inclusion of CTA1 with SIVmac239 Gag dramatically enhanced anti-Gag antibody responses in mice. The adjuvant effects of CTA1 for the secreted antigen HIV gp120 were much less pronounced than those for Gag, as the responses to gp120 were high in the absence of an adjuvant. CTA1 was a stronger adjuvant for Gag than was granulocyte-macrophage colony-stimulating factor (GM-CSF), and it also displayed a wider dose range than GM-CSF in mice. In macaques, CTA1 modestly enhanced the antibody responses to SIV Gag but potently primed for a recombinant Gag protein boost. The results of this study show that CTA1 is a potent adjuvant for SIV Gag when delivered by GeneGun in mice and that CTA1 provides a potent GeneGun-mediated DNA prime for a heterologous protein boost in macaques.
Collapse
|
17
|
Farley N, Bernstein DI, Bravo FJ, Earwood J, Sawtell N, Cardin RD. Recurrent vaginal shedding of herpes simplex type 2 virus in the mouse and effects of antiviral therapy. Antiviral Res 2010; 86:188-95. [PMID: 20167236 DOI: 10.1016/j.antiviral.2010.02.317] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2009] [Revised: 02/08/2010] [Accepted: 02/09/2010] [Indexed: 01/02/2023]
Abstract
A mouse model of recurrent herpes simplex type 2 (HSV-2) would improve our understanding of the immunobiology of recurrent disease and provide a useful model for evaluating antiviral treatments. We developed a model to evaluate recurrent vaginal HSV-2 shedding using high-dose acyclovir (ACV) therapy beginning at 3 days post infection (dpi). Treatment with 150mg/kg of ACV for 10 days increased survival to 80% following vaginal challenge with HSV-2 strain 186 and to 100% after challenge with strain MS. We then evaluated recurrent vaginal HSV-2 shedding in surviving mice. Although infectious virus was not detected in vaginal samples after 21dpi, viral DNA was detectable by PCR in 80% of mice (47/59) on at least 1 day, while no animal was positive for virus on every day. ACV therapy administered from day 21 to 31 significantly reduced recurrent virus shedding during this period from 7.3% (8/109 swabs) to 0.8% (1/126 swabs) (p=0.013). Lastly, ACV-rescued HSV-2-infected mice treated with cyclophosphamide at 35 and 38dpi rapidly succumbed, indicating that this model can be used to study immune control of the persistent infection. Thus, this model provides an inexpensive model for evaluating therapeutic strategies and immune control of persistent HSV.
Collapse
Affiliation(s)
- Nicholas Farley
- Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | | | | | | | | | | |
Collapse
|
18
|
Yager EJ, Dean HJ, Fuller DH. Prospects for developing an effective particle-mediated DNA vaccine against influenza. Expert Rev Vaccines 2009; 8:1205-20. [PMID: 19722894 DOI: 10.1586/erv.09.82] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Vaccine strategies capable of conferring broad protection against both seasonal and pandemic strains of influenza are urgently needed. DNA vaccines are an attractive choice owing to their capacity to induce robust humoral and cellular immune responses at low doses and because they can be developed and manufactured rapidly to more effectively meet the threat of an influenza epidemic or pandemic. Particle-mediated epidermal delivery (PMED), or the gene gun, is a DNA vaccine delivery technology shown to induce protective levels of antibody and T-cell responses in animals and humans against a wide variety of diseases, including influenza. This review focuses on current advances toward the development of an effective PMED DNA vaccine against influenza, including strategies to enhance vaccine immunogenicity, the potential for PMED-based DNA vaccines to improve protection in the vulnerable elderly population, and the prospects for a vaccine capable of providing cross-protection against both seasonal and pandemic strains of influenza.
Collapse
Affiliation(s)
- Eric J Yager
- Center for Immunology & Microbial Disease, Albany Medical College, Albany, NY 12208, USA.
| | | | | |
Collapse
|
19
|
Muller WJ, Dong L, Vilalta A, Byrd B, Wilhelm KM, McClurkan CL, Margalith M, Liu C, Kaslow D, Sidney J, Sette A, Koelle DM. Herpes simplex virus type 2 tegument proteins contain subdominant T-cell epitopes detectable in BALB/c mice after DNA immunization and infection. J Gen Virol 2009; 90:1153-1163. [PMID: 19264627 PMCID: PMC2675279 DOI: 10.1099/vir.0.008771-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2008] [Accepted: 01/08/2009] [Indexed: 11/18/2022] Open
Abstract
Cytotoxic T cells are important in controlling herpes simplex virus type 2 (HSV-2) reactivation and peripheral lesion resolution. Humans latently infected with HSV-2 have cytotoxic T cells directed against epitopes present in tegument proteins. Studies in mice of immunity to HSV have commonly focused on immunodominant responses in HSV envelope glycoproteins. These antigens have not proved to be an effective prophylactic vaccine target for most of the human population. The murine immune response against HSV tegument proteins has not been explored. We analysed cellular responses in BALB/c mice directed against the tegument proteins encoded by UL46, UL47 and UL49 and against the envelope glycoprotein gD after DNA vaccination or HSV-2 infection. After DNA vaccination, the splenocyte T-cell response to overlapping peptides from UL46 and UL47 was more than 500 gamma interferon spot-forming units per 10(6) responder cells. Peptide truncation studies, responder cell fractionation and major histocompatibility complex binding studies identified several CD8(+) and CD4(+) epitopes. Cellular responses to tegument protein epitopes were also detected after HSV-2 infection. Tegument proteins are rational candidates for further HSV-2 vaccine research.
Collapse
Affiliation(s)
- William J. Muller
- Department of Pediatrics, Division of Pediatric Infectious Diseases, University of Washington, Seattle, WA, USA
| | - Lichun Dong
- Department of Medicine, University of Washington, Seattle, WA, USA
| | | | - Benjamin Byrd
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Kai M. Wilhelm
- Department of Medicine, University of Washington, Seattle, WA, USA
| | | | | | - Chao Liu
- Department of Laboratory Medicine, University of Washington, Seattle, WA, USA
| | | | - John Sidney
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - David M. Koelle
- Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Laboratory Medicine, University of Washington, Seattle, WA, USA
- Department of Global Health Medicine, University of Washington, Seattle, WA, USA
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Benaroya Research Institute, Seattle, WA, USA
| |
Collapse
|
20
|
Jones S, Evans K, McElwaine-Johnn H, Sharpe M, Oxford J, Lambkin-Williams R, Mant T, Nolan A, Zambon M, Ellis J, Beadle J, Loudon PT. DNA vaccination protects against an influenza challenge in a double-blind randomised placebo-controlled phase 1b clinical trial. Vaccine 2009; 27:2506-12. [DOI: 10.1016/j.vaccine.2009.02.061] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2008] [Revised: 02/10/2009] [Accepted: 02/18/2009] [Indexed: 12/20/2022]
|
21
|
Engelhorn ME, Guevara-Patiño JA, Merghoub T, Liu C, Ferrone CR, Rizzuto GA, Cymerman DH, Posnett DN, Houghton AN, Wolchok JD. Mechanisms of immunization against cancer using chimeric antigens. Mol Ther 2008; 16:773-81. [PMID: 18301399 PMCID: PMC4399381 DOI: 10.1038/mt.2008.8] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Successful approaches to tumor immunotherapy must overcome the physiological state of tolerance of the immune system to self-tumor antigens. Immunization with appropriate variants of syngeneic antigens can achieve this. However, improvements in vaccine design are needed for efficient cancer immunotherapy. Here we explore nine different chimeric vaccine designs, in which the antigen of interest is expressed as an in-frame fusion with polypeptides that impact antigen processing or presentation. In DNA immunization experiments in mice, three of nine fusions elevated relevant CD8(+) T-cell responses and tumor protection relative to an unfused melanoma antigen. These fusions were: Escherichia coli outer membrane protein A (OmpA), Pseudomonas aeruginosa exotoxin A, and VP22 protein of herpes simplex virus-1. The gains of immunogenicity conferred by the latter two are independent of epitope presentation by major histocompatibility complex class II (MHC II). This finding has positive implications for immunotherapy in individuals with CD4(+) T-cell deficiencies. We present evidence that antigen instability is not a sine qua non condition for immunogenicity. Experiments using two additional melanoma antigens identified different optimal fusion partners, thereby indicating that the benefits of fusion vectors remain antigen specific. Therefore large fusion vector panels such as those presented here can provide information to promote the successful advancement of gene-based vaccines.
Collapse
Affiliation(s)
- Manuel E Engelhorn
- The Swim Across America Laboratory, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York 10021, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Sharpe M, Lynch D, Topham S, Major D, Wood J, Loudon P. Protection of mice from H5N1 influenza challenge by prophylactic DNA vaccination using particle mediated epidermal delivery. Vaccine 2007; 25:6392-8. [PMID: 17640779 DOI: 10.1016/j.vaccine.2007.06.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2006] [Revised: 05/17/2007] [Accepted: 06/06/2007] [Indexed: 11/28/2022]
Abstract
Mice were vaccinated with a DNA plasmid encoding the haemagglutinin (HA) antigen of H5N1 influenza A/Vietnam/1194/2004 by particle mediated epidermal delivery (PMED). Vaccination led to potent anti-HA serological responses that were significantly enhanced by the inclusion of a plasmid expressing the A and B subunits of Escherichia coli heat labile enterotoxin (designated DEI-LT). Mice were vaccinated with H5 or H5/DEI-LT and challenged with 100LD50 H5N1 A/Vietnam/1194/2004 virus. Vaccination provided considerable protection, and mice that received two doses (prime-boost) of H5/DEI-LT showed no symptoms of disease post vaccination, did not shed detectable virus and did not show any rise in anti-H5N1 HI titre post challenge, indicating that they were fully protected. These results demonstrate that the PMED technology may hold promise for the prophylaxis of pandemic influenza.
Collapse
Affiliation(s)
- Michaela Sharpe
- PowderMed Ltd., 4 Robert Robinson Avenue, Oxford Science Park, Oxford OX4 4GA, UK
| | | | | | | | | | | |
Collapse
|