1
|
Zhang X, Goedegebuure SP, Chen MY, Mishra R, Zhang F, Yu YY, Singhal K, Li L, Gao F, Myers NB, Vickery T, Hundal J, McLellan MD, Sturmoski MA, Kim SW, Chen I, Davidson JT, Sankpal NV, Myles S, Suresh R, Ma CX, Foluso A, Wang-Gillam A, Davies S, Hagemann IS, Mardis ER, Griffith O, Griffith M, Miller CA, Hansen TH, Fleming TP, Schreiber RD, Gillanders WE. Neoantigen DNA vaccines are safe, feasible, and induce neoantigen-specific immune responses in triple-negative breast cancer patients. Genome Med 2024; 16:131. [PMID: 39538331 PMCID: PMC11562513 DOI: 10.1186/s13073-024-01388-3] [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: 11/26/2023] [Accepted: 09/20/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Neoantigen vaccines can induce or enhance highly specific antitumor immune responses with minimal risk of autoimmunity. We have developed a neoantigen DNA vaccine platform capable of efficiently presenting both HLA class I and II epitopes and performed a phase 1 clinical trial in triple-negative breast cancer patients with persistent disease on surgical pathology following neoadjuvant chemotherapy, a patient population at high risk of disease recurrence. METHODS Expressed somatic mutations were identified by tumor/normal exome sequencing and tumor RNA sequencing. The pVACtools software suite of neoantigen prediction algorithms was used to identify and prioritize cancer neoantigens and facilitate vaccine design for manufacture in an academic GMP facility. Neoantigen DNA vaccines were administered via electroporation in the adjuvant setting (i.e., following surgical removal of the primary tumor and completion of standard of care therapy). Vaccines were monitored for safety and immune responses via ELISpot, intracellular cytokine production via flow cytometry, and TCR sequencing. RESULTS Eighteen subjects received three doses of a neoantigen DNA vaccine encoding on average 11 neoantigens per patient (range 4-20). The vaccinations were well tolerated with relatively few adverse events. Neoantigen-specific T cell responses were induced in 14/18 patients as measured by ELISpot and flow cytometry. At a median follow-up of 36 months, recurrence-free survival was 87.5% (95% CI: 72.7-100%) in the cohort of vaccinated patients. CONCLUSION Our study demonstrates neoantigen DNA vaccines are safe, feasible, and capable of inducing neoantigen-specific immune responses. CLINICAL TRIAL REGISTRATION NUMBER NCT02348320.
Collapse
Affiliation(s)
- Xiuli Zhang
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - S Peter Goedegebuure
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, Saint Louis, MO, USA
| | - Michael Y Chen
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Rashmi Mishra
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Felicia Zhang
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Yik Yeung Yu
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Kartik Singhal
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Lijin Li
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Feng Gao
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Nancy B Myers
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Tammi Vickery
- Bursky Center for Human Immunology & Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Jasreet Hundal
- McDonnell Genome Institute, Washington University School of Medicine, Saint Louis, MO, USA
| | - Michael D McLellan
- McDonnell Genome Institute, Washington University School of Medicine, Saint Louis, MO, USA
| | - Mark A Sturmoski
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Samuel W Kim
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Ina Chen
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Jesse T Davidson
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Narendra V Sankpal
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Stephanie Myles
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, Saint Louis, MO, USA
| | - Rama Suresh
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, Saint Louis, MO, USA
| | - Cynthia X Ma
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, Saint Louis, MO, USA
| | - Ademuyiwa Foluso
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, Saint Louis, MO, USA
| | - Andrea Wang-Gillam
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, Saint Louis, MO, USA
| | - Sherri Davies
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Ian S Hagemann
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Elaine R Mardis
- McDonnell Genome Institute, Washington University School of Medicine, Saint Louis, MO, USA
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, Saint Louis, MO, USA
- Current Affiliation: Department of Pediatrics, Nationwide Children's Hospital and The Ohio State University College of Medicine, Columbus, OH, USA
| | - Obi Griffith
- McDonnell Genome Institute, Washington University School of Medicine, Saint Louis, MO, USA
| | - Malachi Griffith
- McDonnell Genome Institute, Washington University School of Medicine, Saint Louis, MO, USA
| | - Christopher A Miller
- McDonnell Genome Institute, Washington University School of Medicine, Saint Louis, MO, USA
| | - Ted H Hansen
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Timothy P Fleming
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, Saint Louis, MO, USA
| | - Robert D Schreiber
- Bursky Center for Human Immunology & Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, Saint Louis, MO, USA
| | - William E Gillanders
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA.
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
2
|
Huo C, Huang D, Ma Z, Li G, Li T, Lin W, Jiang N, Xing W, Xu G, Yu H, Luo L, Sun H. Specificity of DNA Vaccines against the Genogroup J and U Infectious Hematopoietic Necrosis Virus Strains Prevalent in China. Viruses 2022; 14:v14122707. [PMID: 36560709 PMCID: PMC9780822 DOI: 10.3390/v14122707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 11/23/2022] [Accepted: 11/29/2022] [Indexed: 12/09/2022] Open
Abstract
Infectious hematopoietic necrosis virus (IHNV) is the most important pathogen threatening the aquaculture of salmonid fish in China. In addition to the common genogroup J IHNV, genogroup U has been newly discovered in China. However, there is no effective DNA vaccine to fight against this emerging genogroup U IHNV in China. In this study, DNA vaccines encoding the IHNV viral glycoprotein (G) gene of the GS2014 (genogroup J) and BjLL (genogroup U) strains isolated from northern China were successfully developed, which were identified by restriction analysis and IFA. The expression of the Mx-1 gene and G gene in the spleens and muscles of the injection site as well as the titers of the serum antibodies were measured to evaluate the vaccine efficacy by RT-qPCR and ELISA. We found that DNA vaccine immunization could activate Mx1 gene expression and upregulate G gene expression, and the mRNA levels of the Mx1 gene in the muscles were significantly higher than those in the spleens. Notably, DNA vaccine immunization might not promote the serum antibody in fish at the early stage of immunization. Furthermore, the efficacy of the constructed vaccines was tested in intra- and cross-genogroup challenges by a viral challenge in vivo. It seemed that the DNA vaccines were able to provide great immune protection against IHNV infection. In addition, the genogroup J IHNV-G DNA vaccine showed better immune efficacy than the genogroup U IHNV-G or divalent vaccine, which could provide cross-immune protection against the genogroup U IHNV challenge. Therefore, this is the first study to construct an IHNV DNA vaccine using the G gene from an emerging genogroup U IHNV strain in China. The results provide great insight into the advances of new prophylactic strategies to fight both the genogroup J and U IHNV in China.
Collapse
Affiliation(s)
- Caiyun Huo
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, No. 9 Shuguang Huayuan Zhonglu, Haidian District, Beijing 100097, China
| | - Dandan Huang
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, No. 9 Shuguang Huayuan Zhonglu, Haidian District, Beijing 100097, China
| | - Zhihong Ma
- Beijing Fisheries Research Institute, Beijing Academy of Agriculture and Forestry Sciences, No. 18 Jiaomen Road, Fengtai District, Beijing 100068, China
| | - Guiping Li
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, No. 9 Shuguang Huayuan Zhonglu, Haidian District, Beijing 100097, China
| | - Tieliang Li
- Beijing Fisheries Research Institute, Beijing Academy of Agriculture and Forestry Sciences, No. 18 Jiaomen Road, Fengtai District, Beijing 100068, China
| | - Wutong Lin
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, No. 9 Shuguang Huayuan Zhonglu, Haidian District, Beijing 100097, China
| | - Na Jiang
- Beijing Fisheries Research Institute, Beijing Academy of Agriculture and Forestry Sciences, No. 18 Jiaomen Road, Fengtai District, Beijing 100068, China
| | - Wei Xing
- Beijing Fisheries Research Institute, Beijing Academy of Agriculture and Forestry Sciences, No. 18 Jiaomen Road, Fengtai District, Beijing 100068, China
| | - Guanling Xu
- Beijing Fisheries Research Institute, Beijing Academy of Agriculture and Forestry Sciences, No. 18 Jiaomen Road, Fengtai District, Beijing 100068, China
| | - Huanhuan Yu
- Beijing Fisheries Research Institute, Beijing Academy of Agriculture and Forestry Sciences, No. 18 Jiaomen Road, Fengtai District, Beijing 100068, China
| | - Lin Luo
- Beijing Fisheries Research Institute, Beijing Academy of Agriculture and Forestry Sciences, No. 18 Jiaomen Road, Fengtai District, Beijing 100068, China
- Correspondence: (L.L.); (H.S.)
| | - Huiling Sun
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, No. 9 Shuguang Huayuan Zhonglu, Haidian District, Beijing 100097, China
- Correspondence: (L.L.); (H.S.)
| |
Collapse
|
3
|
Ulmer JB, Liu MA. Path to Success and Future Impact of Nucleic Acid Vaccines: DNA and mRNA. MOLECULAR FRONTIERS JOURNAL 2021. [DOI: 10.1142/s2529732521400022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The rapid development of mRNA vaccines for COVID-19 has both astonished the world and raised concerns about their safety, perhaps because many people do not realize the decades’ long efforts for nucleic acid vaccines, both mRNA and DNA vaccines, including the licensure of several veterinary DNA vaccines. This manuscript traces the milestones for nucleic acid vaccine research and development (R&D), with a focus on the immune and safety issues they both raised and answered. The characteristics of the two entities are compared, demonstrating the similarities and differences between them, the advantages and disadvantages, which might lead toward using one or the other technology for different indications. In addition, as the SARS-CoV-2 pandemic has once again highlighted the importance of One Health, that is, the interactions between animal and human pathogens, focus will also be given to how DNA vaccine utilization and studies both in large domestic animals and in wildlife pave the way for more integrated approaches for vaccines to respond quickly to, and prevent, the global impacts of emerging diseases.
Collapse
|
4
|
Li L, Zhang X, Wang X, Kim SW, Herndon JM, Becker-Hapak MK, Carreno BM, Myers NB, Sturmoski MA, McLellan MD, Miller CA, Johanns TM, Tan BR, Dunn GP, Fleming TP, Hansen TH, Goedegebuure SP, Gillanders WE. Optimized polyepitope neoantigen DNA vaccines elicit neoantigen-specific immune responses in preclinical models and in clinical translation. Genome Med 2021; 13:56. [PMID: 33879241 PMCID: PMC8059244 DOI: 10.1186/s13073-021-00872-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 03/17/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Preclinical studies and early clinical trials have shown that targeting cancer neoantigens is a promising approach towards the development of personalized cancer immunotherapies. DNA vaccines can be rapidly and efficiently manufactured and can integrate multiple neoantigens simultaneously. We therefore sought to optimize the design of polyepitope DNA vaccines and test optimized polyepitope neoantigen DNA vaccines in preclinical models and in clinical translation. METHODS We developed and optimized a DNA vaccine platform to target multiple neoantigens. The polyepitope DNA vaccine platform was first optimized using model antigens in vitro and in vivo. We then identified neoantigens in preclinical breast cancer models through genome sequencing and in silico neoantigen prediction pipelines. Optimized polyepitope neoantigen DNA vaccines specific for the murine breast tumor E0771 and 4T1 were designed and their immunogenicity was tested in vivo. We also tested an optimized polyepitope neoantigen DNA vaccine in a patient with metastatic pancreatic neuroendocrine tumor. RESULTS Our data support an optimized polyepitope neoantigen DNA vaccine design encoding long (≥20-mer) epitopes with a mutant form of ubiquitin (Ubmut) fused to the N-terminus for antigen processing and presentation. Optimized polyepitope neoantigen DNA vaccines were immunogenic and generated robust neoantigen-specific immune responses in mice. The magnitude of immune responses generated by optimized polyepitope neoantigen DNA vaccines was similar to that of synthetic long peptide vaccines specific for the same neoantigens. When combined with immune checkpoint blockade therapy, optimized polyepitope neoantigen DNA vaccines were capable of inducing antitumor immunity in preclinical models. Immune monitoring data suggest that optimized polyepitope neoantigen DNA vaccines are capable of inducing neoantigen-specific T cell responses in a patient with metastatic pancreatic neuroendocrine tumor. CONCLUSIONS We have developed and optimized a novel polyepitope neoantigen DNA vaccine platform that can target multiple neoantigens and induce antitumor immune responses in preclinical models and neoantigen-specific responses in clinical translation.
Collapse
Affiliation(s)
- Lijin Li
- Department of Surgery, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Xiuli Zhang
- Department of Surgery, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Xiaoli Wang
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Samuel W Kim
- Department of Surgery, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - John M Herndon
- Department of Surgery, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | | | - Beatriz M Carreno
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
- Present Address: Parker Institute for Cancer Immunotherapy, Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nancy B Myers
- Department of Surgery, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Mark A Sturmoski
- Department of Surgery, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Michael D McLellan
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
| | - Christopher A Miller
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, St Louis, MO, USA
| | - Tanner M Johanns
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Benjamin R Tan
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Gavin P Dunn
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Timothy P Fleming
- Department of Surgery, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, St Louis, MO, USA
- Present Address: Norton Thoracic Institute, St. Joseph Hospital and Medical Center, Phoenix, AZ, USA
| | - Ted H Hansen
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - S Peter Goedegebuure
- Department of Surgery, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, St Louis, MO, USA
| | - William E Gillanders
- Department of Surgery, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA.
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, St Louis, MO, USA.
| |
Collapse
|
5
|
Rong X, Ailing F, Xiaodong L, Jie H, Min L. Monitoring hepatitis B by using point-of-care testing: biomarkers, current technologies, and perspectives. Expert Rev Mol Diagn 2021; 21:195-211. [PMID: 33467927 DOI: 10.1080/14737159.2021.1876565] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Introduction: Liver diseases caused by hepatitis B virus (HBV) are pandemic infectious diseases that seriously endanger human health, conventional diagnosis methods can not meet the requirements in resource-limited areas. The point of acre detection methods can easily resolve those problems. Herein, we review the most recent advances in POC-based hepatitis B detection methods and present some recommendations for future development. It aims to provide ideas for future research.Areas covered: Epidemiological data on Hepatitis B, conventional diagnostic methods for hepatitis B detection, some latest point of care detection methods for hepatitis B detection and list out the recommendations for future development.Expert opinion: This manuscript summarized traditional biomarkers of different hepatitis B stages and recent-developed POCT platforms (including microfluidic platforms and lateral-flow strips) and discuss the challenges associated with their use. Some emerging biomarkers that can be used in hepatitis B diagnosis are also listed. This manuscript has certain guiding significance to the development of hepatitis B detection.
Collapse
Affiliation(s)
- Xu Rong
- Institute of Physics & Optoelectronics Technology, Baoji University of Arts and Sciences, Baoji, China
| | - Feng Ailing
- Institute of Physics & Optoelectronics Technology, Baoji University of Arts and Sciences, Baoji, China
| | - Li Xiaodong
- Institute of Physics & Optoelectronics Technology, Baoji University of Arts and Sciences, Baoji, China
| | - Hu Jie
- Suzhou DiYinAn Biotech Co., Ltd. & Suzhou Innovation Center for Life Science and Technology, Suzhou, China
| | - Lin Min
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
6
|
Lokaj‐Berisha V, Gacaferri‐Lumezi B, Berisha N. Serum levels of stress hormones in Albanian patients with allergic rhinitis. Int Forum Allergy Rhinol 2019; 9:1077-1082. [DOI: 10.1002/alr.22370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 05/22/2019] [Accepted: 05/30/2019] [Indexed: 11/10/2022]
Affiliation(s)
- Violeta Lokaj‐Berisha
- Department of Physiology & Immunology, Faculty of MedicineUniversity of Prishtina Republic of Kosova
| | - Besa Gacaferri‐Lumezi
- Department of Physiology & Immunology, Faculty of MedicineUniversity of Prishtina Republic of Kosova
| | - Naser Berisha
- Department of Obstetrics and GynecologyUniversity Clinical Centre Prishtina Republic of Kosova
| |
Collapse
|
7
|
Fynan EF, Lu S, Robinson HL. One Group's Historical Reflections on DNA Vaccine Development. Hum Gene Ther 2018; 29:966-970. [PMID: 30129778 PMCID: PMC6152846 DOI: 10.1089/hum.2018.066] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 06/30/2018] [Indexed: 12/02/2022] Open
Abstract
DNA vaccines were pioneered by several groups in the early 1990s. This article presents the reflections of one of these groups on their work with retroviral vectors in chickens that contributed to the discovery and early development of DNA vaccines. Although the findings were initially met with skepticism, the work presented here combined with that of others founded a new method of vaccination: the direct inoculation of purified DNA encoding the target antigen.
Collapse
Affiliation(s)
- Ellen F. Fynan
- Department of Biology, Worcester State College, Worcester, Massachusetts
| | - Shan Lu
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | | |
Collapse
|
8
|
Minz S, Pandey RS. Development of Adjuvanted Solid Fat Nanoemulsions for Pulmonary Hepatitis B Vaccination. J Pharm Sci 2018; 107:1701-1712. [PMID: 29454622 DOI: 10.1016/j.xphs.2018.02.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 02/04/2018] [Accepted: 02/06/2018] [Indexed: 10/18/2022]
Abstract
Pulmonary vaccination is one of the most promising routes for immunization owing to its noninvasive nature and induction of strong mucosal immunity and systemic response. In the present study, recombinant hepatitis B surface antigen loaded solid fat nanoemulsions (SFNs) as carrier system and monophosphoryl lipid A as an adjuvant-carrier system was prepared and evaluated as multiadjuvanted vaccine system for deep pulmonary vaccination. Deposition and clearance from the deep lung of rats were determined by gamma scintigraphy. Biodistribution of SFNs was determined by the live animal imaging system. SFNs dispersion showed slower clearance as compared with sodium pertechnetate control solution (∗∗∗p <0.001) from the pulmonary region due to the virtue of particulate and hydrophobic nature of formulations. Humoral (sIgA and IgG) and cellular (IL-2 and IF-γ) immune responses were found to be significant (∗∗∗p <0.001) when compared with naïve antigen (recombinant surface antigen without any excipient) solution. Data indicate that deep pulmonary immunization offers a stronger immune response with balanced humoral, mucosal, and cellular immunization, which further needs to be tested in higher animals to support this hypothesis for clinical translation of this so far neglected yet potential target tissue for immunization.
Collapse
Affiliation(s)
- Sunita Minz
- Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, Madhya Pradesh, India 484887; SLT Institute of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya, Bilaspur, Chhattisgarh, India 495009
| | - Ravi Shankar Pandey
- SLT Institute of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya, Bilaspur, Chhattisgarh, India 495009.
| |
Collapse
|
9
|
Abstract
INTRODUCTION Traditional inactivated and protein vaccines generate strong antibodies, but struggle to generate T cell responses. Attenuated pathogen vaccines generate both, but risk causing the disease they aim to prevent. Newer gene-based vaccines drive both responses and avoid the risk of infection. While these replication-defective (RD) vaccines work well in small animals, they can be weak in humans because they do not replicate antigen genes like more potent replication-competent (RC) vaccines. RC vaccines generate substantially stronger immune responses, but also risk causing their own infections. To circumvent these problems, we developed single-cycle adenovirus (SC-Ad) vectors that amplify vaccine genes, but that avoid the risk of infection. This review will discuss these vectors and their prospects for use as vaccines. AREAS COVERED This review provides a background of different types of vaccines. The benefits of gene-based vaccines and their ability to replicate antigen genes are described. Adenovirus vectors are discussed and compared to other vaccine types. Replication-defective, single-cycle, and replication-competent Ad vaccines are compared. EXPERT COMMENTARY The potential utility of these vaccines are discussed when used against infectious diseases and as cancer vaccines. We propose a move away from replication-defective vaccines towards more robust replication-competent or single-cycle vaccines.
Collapse
Affiliation(s)
- Michael Barry
- a Division of Infectious Diseases, Department of Medicine, Department of Immunology, Department of Molecular Medicine , Mayo Clinic , Rochester , MN , USA
| |
Collapse
|
10
|
Mahajan V, Gaymalov Z, Alakhova D, Gupta R, Zucker IH, Kabanov AV. Horizontal gene transfer from macrophages to ischemic muscles upon delivery of naked DNA with Pluronic block copolymers. Biomaterials 2015; 75:58-70. [PMID: 26480472 DOI: 10.1016/j.biomaterials.2015.10.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 09/30/2015] [Accepted: 10/01/2015] [Indexed: 12/31/2022]
Abstract
Intramuscular administration of plasmid DNA (pDNA) with non-ionic Pluronic block copolymers increases gene expression in injected muscles and lymphoid organs. We studied the role of immune cells in muscle transfection upon inflammation. Local inflammation in murine hind limb ischemia model (MHLIM) drastically increased DNA, RNA and expressed protein levels in ischemic muscles injected with pDNA/Pluronic. The systemic inflammation (MHLIM or peritonitis) also increased expression of pDNA/Pluronic in the muscles. When pDNA/Pluronic was injected in ischemic muscles the reporter gene, Green Fluorescent Protein (GFP) co-localized with desmin(+) muscle fibers and CD11b(+) macrophages (MØs), suggesting transfection of MØs along with the muscle cells. P85 enhanced (∼ 4 orders) transfection of MØs with pDNA in vitro. Moreover, adoptively transferred MØs were shown to pass the transgene to inflamed muscle cells in MHLIM. Using a co-culture of myotubes (MTs) and transfected MØs expressing a reporter gene under constitutive (cmv-luciferase) or muscle specific (desmin-luciferase) promoter we demonstrated that P85 enhances horizontal gene transfer from MØ to MTs. Therefore, MØs can play an important role in muscle transfection with pDNA/Pluronic during inflammation, with both inflammation and Pluronic contributing to the increased gene expression. pDNA/Pluronic has potential for therapeutic gene delivery in muscle pathologies that involve inflammation.
Collapse
Affiliation(s)
- Vivek Mahajan
- Division of Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA; Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-5850, USA
| | - Zagit Gaymalov
- Division of Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA; Department of Pharmaceutical Sciences and Center for Drug Delivery and Nanomedicine, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198-5850, USA
| | - Daria Alakhova
- Division of Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Richa Gupta
- Division of Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Irving H Zucker
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198-5850, USA
| | - Alexander V Kabanov
- Division of Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA; Department of Pharmaceutical Sciences and Center for Drug Delivery and Nanomedicine, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198-5850, USA; Laboratory of Chemical Design of Bionanomaterials, Faculty of Chemistry, M.V. Lomonosov Moscow State University, 119899 Moscow, Russia.
| |
Collapse
|
11
|
Gebbing M, Bergmann T, Schulz E, Ehrhardt A. Gene therapeutic approaches to inhibit hepatitis B virus replication. World J Hepatol 2015; 7:150-164. [PMID: 25729471 PMCID: PMC4342598 DOI: 10.4254/wjh.v7.i2.150] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 10/23/2014] [Accepted: 11/19/2014] [Indexed: 02/06/2023] Open
Abstract
Acute and chronic hepatitis B virus (HBV) infections remain to present a major global health problem. The infection can be associated with acute symptomatic or asymptomatic hepatitis which can cause chronic inflammation of the liver and over years this can lead to cirrhosis and the development of hepatocellular carcinomas. Currently available therapeutics for chronically infected individuals aim at reducing viral replication and to slow down or stop the progression of the disease. Therefore, novel treatment options are needed to efficiently combat and eradicate this disease. Here we provide a state of the art overview of gene therapeutic approaches to inhibit HBV replication. We discuss non-viral and viral approaches which were explored to deliver therapeutic nucleic acids aiming at reducing HBV replication. Types of delivered therapeutic nucleic acids which were studied since many years include antisense oligodeoxynucleotides and antisense RNA, ribozymes and DNAzymes, RNA interference, and external guide sequences. More recently designer nucleases gained increased attention and were exploited to destroy the HBV genome. In addition we mention other strategies to reduce HBV replication based on delivery of DNA encoding dominant negative mutants and DNA vaccination. In combination with available cell culture and animal models for HBV infection, in vitro and in vivo studies can be performed to test efficacy of gene therapeutic approaches. Recent progress but also challenges will be specified and future perspectives will be discussed. This is an exciting time to explore such approaches because recent successes of gene therapeutic strategies in the clinic to treat genetic diseases raise hope to find alternative treatment options for patients chronically infected with HBV.
Collapse
|
12
|
Nezam FS, Hosseini SM, Kheiri MT, Abdoli A, Memarnejadian A, Shenagari M, Gholami S, Sohani H, Rahmatollahi H, Jamali A. Suppressive Effects of Chronic Stress on Influenza Virus Protection after Vaccination with Plasmid DNA-Encoded Nucleoprotein. Neuroimmunomodulation 2015; 22:322-7. [PMID: 25765110 DOI: 10.1159/000371354] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 11/24/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Influenza is a highly infectious and acute respiratory disease caused by an infection of the host respiratory tract mucosa by the influenza virus. The use of DNA vaccines that express conserved genes such as nucleoprotein (NP) represents a new method of vaccination against influenza. In this study, the effect of chronic stress on the efficiency of this type of vaccine has been evaluated in a mouse model. METHODS The NP DNA vaccine was administered intradermally 3 times on days 0, 3 and 6 to stressed and nonstressed male BALB/c mice. Two weeks after the last immunization, half of these mice were challenged with A/Puerto Rico/8/34 (PR8) influenza virus and were weighed for 12 days, and their mortality rate was assessed during this period. The cellular immune response of the other half of the mice was evaluated by cytotoxicity assay. RESULTS The results indicate a significant reduction in the cytotoxic T-lymphocyte response of stressed mice in comparison with unstressed mice. Also, the percentage of weight loss and mortality after the challenge in stressed mice was significantly increased compared to the other group. CONCLUSION These results indicate that the NP DNA vaccine is not able to induce any effective cytotoxic T-lymphocyte response against influenza virus in stressed mice and cannot induce protective immunity against influenza infection in this group of mice.
Collapse
Affiliation(s)
- Fatemeh Sadat Nezam
- Department of Microbiology, Faculty of Biological Sciences, Shahid-Beheshti University, Tehran, Iran
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Wu Y, Zhang F, Ma W, Song J, Huang Q, Zhang H. A Plasmid Encoding Japanese Encephalitis Virus PrM and E Proteins Elicits Protective Immunity in Suckling Mice. Microbiol Immunol 2013; 48:585-90. [PMID: 15322338 DOI: 10.1111/j.1348-0421.2004.tb03555.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
A plasmid encoding Japanese encephalitis virus (JEV) prM and E proteins was constructed, and its efficacy as a candidate vaccine against JEV was evaluated in suckling mice. Groups of 10 BALB/c mice (5-7 days old) were immunized twice via muscular injection with this DNA vaccine, an empty vector or PBS at an interval of 3 weeks, and were challenged with a lethal dose of JEV 3 weeks after the second inoculation. Both cellular and humoral immune responses were examined before the challenge. Two animals from each group were sacrificed to detect the JEV-specific cytotoxic T lymphocyte activity. JEV-specific lactate dehydrogenase release in the DNA vaccine, empty vector and PBS groups was 37.5%, 18% and 8.5% respectively. JEV-specific antibody was detected in 8 of 10 animals in DNA vaccine group with a geometrical mean titer of 1: 28.3. The pooled serum from the same group also showed a neutralizing activity. Six of 8 mice in the DNA vaccine group survived the challenge, with a protection rate of 75%, but all the mice died in the two control groups. These results show that this JEV prM and E DNA vaccine is immunogenic and protective against JEV infection in the mouse model.
Collapse
MESH Headings
- Animals
- Animals, Suckling
- Antibodies, Viral/blood
- Encephalitis Virus, Japanese/genetics
- Encephalitis Virus, Japanese/immunology
- Encephalitis, Japanese/immunology
- Encephalitis, Japanese/prevention & control
- Immunization
- Japanese Encephalitis Vaccines/administration & dosage
- Japanese Encephalitis Vaccines/genetics
- Japanese Encephalitis Vaccines/immunology
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/immunology
- Mice
- Mice, Inbred BALB C
- Plasmids/genetics
- Plasmids/immunology
- T-Lymphocytes, Cytotoxic/immunology
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/immunology
- Viral Envelope Proteins/genetics
- Viral Envelope Proteins/immunology
Collapse
Affiliation(s)
- Yushui Wu
- Department of Microbiology, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | | | | | | | | | | |
Collapse
|
14
|
Han L, Zhen YH, Liang AX, Zhang J, Riaz H, Xiong JJ, Guo AZ, Yang LG. Oral vaccination with inhibin DNA delivered using attenuated Salmonella choleraesuis for improving reproductive traits in mice. J Basic Microbiol 2013; 54:962-8. [PMID: 24123188 DOI: 10.1002/jobm.201300052] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Accepted: 06/29/2013] [Indexed: 01/11/2023]
Abstract
The objective of this study was to examine the efficacy and safety of a novel inhibin vaccine containing inhibin α (1-32) fragments in mice. A recombinant plasmid pVAX-asd-IS was constructed by inserting recombinant inhibin α (1-32) and the hepatitis B surface antigen S into the plasmid in which the asd gene, rather than the kanamycin gene, was a selection marker. Ninety Kuming mice were divided into six groups consisting of 15 mice each. First group was (C1) injected with 200 µl of PBS, second (C2) received 1 × 10(10) CFU of crp(-) /asd(-) C500/pVAX-asd and served as vector control, third did not receive any treatment (C3), while fourth, fifth, and sixth group received 1 × 10(10) , 1 × 10(9) , 1 × 10(8) CFU of the recombinant inhibin vaccine crp(-) /asd(-) C500/pVAX-asd-IS (group T1, T2, T3), respectively. Western blotting demonstrated that recombinant expressed inhibin protein possessed immune function and that this plasmid could replicate for up to 40 generations stably. Vaccination with this strain at a dose of 1 × 10(10) CFU/200 µl per mouse induced high anti-inhibin antibody levels, significantly increased large-follicle production in T1 group (p < 0.05) and average litter size (p > 0.05) compared with control groups. Integration studies showed no evidence of inhibin fusion gene integrated into mice's genome 2-month after immunization. These results suggest that the vaccine described in the present study may provide a safe method to improve reproductive traits in animals. A trend towards increased litter size and significant increase in large follicle population depict that this vaccine may have direct application in large animal industry.
Collapse
Affiliation(s)
- Li Han
- Laboratory of Animal Infectious Diseases, State Key Laboratory of Agricultural Microbiology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; College of Animal Science and Technology and College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Zhao Y, Bao Y, Zhang L, Chang L, Jiang L, Liu Y, Zhang L, Qin J. Biosafety of the plasmid pcDNA3-1E of Eimeria acervulina in chicken. Exp Parasitol 2012; 133:231-6. [PMID: 23247194 DOI: 10.1016/j.exppara.2012.11.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2012] [Revised: 11/30/2012] [Accepted: 11/30/2012] [Indexed: 11/19/2022]
Abstract
To evaluate the biosafety of the plasmid pcDNA3-1E of Eimeria acervulina in chicken, two-week-old chickens were injected intramuscularly with the plasmid pcDNA3-1E at dose of 50 μg/chicken. At the 15 days post-injection, the tissue samples were collected, the total DNA was extracted, and the 3-1E gene was amplified by PCR. Genomic DNA was first purified away from free plasmid using gel electrophoresis, and then assayed for integrated plasmid using PCR amplification of the 3-1E gene. Simultaneously, the environmental dejection samples were collected, the total bacterial DNA was extracted and then transfer of the pcDNA3-1E gene was detected by PCR amplification of the 3-1E gene. Two-week-old chickens were injected intramuscularly with the plasmid pcDNA3-1E with three dosage groups of 100 μg, 500 μg and 2500 μg/chicken for 14 days respectively, and with physiological saline at dose of 2500 μL/chicken as control group for acute toxicity test. A target band of 583 bp was obtained by PCR with chicken genomic DNA as template. If the chicken genomic DNA was purified, no target band could be obtained. It showed that the recombinant plasmid pcDNA3-1E existed in tissues, and no genomic integration of DNA plasmid was detected in the immunized chickens. No target band was found by PCR with environmental dejection bacteria genomic DNA as template. It showed that integration and transfer phenomenon did not exist in environment. The acute toxicity results showed the typical clinical symptoms did not occur in the inoculated chickens, the blood biochemical indices and viscera configuration were not affected significantly in the inoculated group and control group (P>0.05). The results showed that the plasmid pcDNA3-1E was safe and suitable for chicken clinical trials.
Collapse
Affiliation(s)
- Yuelan Zhao
- College of Veterinary Medicine , Agricultural University of Hebei, Baoding 071001, China
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Niedre-Otomere B, Bogdanova A, Skrastina D, Zajakina A, Bruvere R, Ose V, Gerlich WH, Garoff H, Pumpens P, Glebe D, Kozlovska T. Recombinant Semliki Forest virus vectors encoding hepatitis B virus small surface and pre-S1 antigens induce broadly reactive neutralizing antibodies. J Viral Hepat 2012; 19:664-73. [PMID: 22863271 DOI: 10.1111/j.1365-2893.2012.01594.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Most hepatitis B virus (HBV) vaccines consist of viral small surface (S) protein subtype adw2 expressed in yeast cells. In spite of good efficacy, HBV-genotype and subtype differences, escape mutants and insufficient Th1 activation remain potential problems. To address these problems, we generated recombinant Semliki Forest virus (rSFV) vectors encoding S protein, subtype adw2 or ayw2, or a fragment of the large surface protein, amino acids 1-48 of the pre-S1 domain, fused to S (pre-S1.1-48/S). The antigen loop in S protein and the selected pre-S1 sequences are known targets of neutralizing antibodies. BALB/c mice were immunized intravenously with 10(7) rSFV particles and 10(8) rSFV particles 3 weeks later. Antibodies induced by rSFV encoding S proteins reacted preferentially with subtype determinants of yeast-derived S antigen but equally well with patient-derived S antigen. Immunization with rSFV encoding pre-S1.1-48/S resulted in formation of pre-S1- and S-specific immunoglobulin G (IgG), while immunization with the isogenic mutant without S start codon induced pre-S1 antibodies only. Neutralizing antibodies were determined by mixing with plasma-derived HBV/ayw2 and subsequent inoculation of susceptible primary hepatocyte cultures from Tupaia belangeri. S/adw2 antisera neutralized HBV/ayw2 as effectively as antisera raised with S/ayw2. The pre-S1 antibodies also completely neutralized HBV infectivity. The IgG1/IgG2a ratios ranged from 0.28 to 0.88 in the four immunized groups and were lowest for the pre-S1.1-48/S vector, indicating the strongest Th1 response. This vector type may induce subtype-independent and S-escape-resistant neutralizing antibodies against HBV.
Collapse
Affiliation(s)
- B Niedre-Otomere
- Biomedical Research and Study Centre, University of Latvia, Riga, Latvia, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Wang SL, Han L, Ahmad S, Cao SX, Xue LQ, Xing ZF, Feng JZ, Liang AX, Yang LG. Effect of a DNA vaccine harboring two copies of inhibin α (1-32) fragments on immune response, hormone concentrations and reproductive performance in rats. Theriogenology 2012; 78:393-401. [PMID: 22592078 DOI: 10.1016/j.theriogenology.2012.02.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2011] [Revised: 02/18/2012] [Accepted: 02/21/2012] [Indexed: 10/28/2022]
Abstract
The objective was to investigate the effects of a novel DNA vaccine (pcISI) harboring two copies of inhibin α (1-32) fragments on immune response, hormone concentrations and reproductive performance in rats. Female Wistar rats (n=18 per group) were immunized (twice, 4 wk apart) with 10, 50, or 100 μg (T1, T2 and T3, respectively), of the pcISI plasmid. At 4 wk after the second immunization, plasma antibody titers were higher (P<0.05) in T3 than in either T1 or T2 (0.341±0.123, 0.236±0.068, and 0.251±0.077, respectively, mean±SD). Concurrrently, plasma concentrations of FSH and estradiol were highest (P<0.05) in T3, and were higher (P<0.05) in T1 and T2 than in control groups. For antibody-positive rats, there was a correlation (P<0.01) between antibody titer and FSH concentrations after two pcISI immunizations. The number of mature follicles in the T3 group (46.00±4.65) was higher (P<0.05) than in two control groups (29.25±3.72 and 27.92±3.48), and also higher (P<0.05) than in T1 and T2 (37.17±4.99 and 38.75±7.09). Antibody-positive rats had more mature ovarian follicles than negative rats (46.75±4.23 vs. 35.60±3.38, P<0.05). Moreover, litter size and number of placentas were increased (P<0.05) in the pcISI immunization groups, except for the T1 group, compared to the control groups. In conclusion, the pcISI DNA vaccine successfully induced a humoral immune response, improved reproductive hormone concentrations, stimulated follicular development, and increased number of placentas and litter size. Furthermore, 100 μg yielded the best immune response.
Collapse
Affiliation(s)
- Shui-Lian Wang
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Zhou H, Min J, Zhao Q, Gu Q, Cong H, Li Y, He S. Protective immune response against Toxoplasma gondii elicited by a recombinant DNA vaccine with a novel genetic adjuvant. Vaccine 2012; 30:1800-6. [PMID: 22240340 DOI: 10.1016/j.vaccine.2012.01.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Revised: 12/05/2011] [Accepted: 01/02/2012] [Indexed: 12/11/2022]
Abstract
Previous immunological studies from our laboratory have demonstrated the potential role of Toxoplasma gondii antigens SAG1 and GRA2 as vaccine candidates. To further evaluate the vaccine's effects, a series of recombinant DNA vaccines pVAX1-SAG1, pVAX1-GRA2 and pVAX1-SAG1-GRA2, termed pSAG1, pGRA2 and pSAG1-GRA2, respectively, were constructed. A plasmid pVAX1-S/PreS2, termed pSPreS2 encoding hepatitis B virus (HBV) surface antigen (HBsAg) S and PreS2 as a novel genetic adjuvant, was also constructed. The expression abilities of those DNA plasmids were examined in HFF cells by Western blotting. Then BALB/c mice were intramuscularly immunized with DNA plasmids and followed by challenging with the highly virulent T. gondii RH strain. The results demonstrated that the recombinant DNA vaccine pSAG1-GRA2 was capable of eliciting high levels of antibodies, a Th1 type of immune response with significant production of IFN-γ and low levels of IL-4 or IL-10 in BALB/c mice, and partial protection against the acute phase of toxoplasmosis as compared to pSAG1, pGRA2 and controls. In addition, the adjuvant pSPreS2 formulated with DNA vaccine induced a Th1 type of immune response and therefore might be a novel genetic adjuvant to DNA vaccine for further investigation.
Collapse
Affiliation(s)
- Huaiyu Zhou
- Department of Parasitology, Shandong University School of Medicine, Jinan, Shandong, PR China.
| | | | | | | | | | | | | |
Collapse
|
19
|
Cancer genome sequencing and its implications for personalized cancer vaccines. Cancers (Basel) 2011; 3:4191-211. [PMID: 24213133 PMCID: PMC3763418 DOI: 10.3390/cancers3044191] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2011] [Revised: 10/31/2011] [Accepted: 11/09/2011] [Indexed: 12/31/2022] Open
Abstract
New DNA sequencing platforms have revolutionized human genome sequencing. The dramatic advances in genome sequencing technologies predict that the $1,000 genome will become a reality within the next few years. Applied to cancer, the availability of cancer genome sequences permits real-time decision-making with the potential to affect diagnosis, prognosis, and treatment, and has opened the door towards personalized medicine. A promising strategy is the identification of mutated tumor antigens, and the design of personalized cancer vaccines. Supporting this notion are preliminary analyses of the epitope landscape in breast cancer suggesting that individual tumors express significant numbers of novel antigens to the immune system that can be specifically targeted through cancer vaccines.
Collapse
|
20
|
Abstract
This review provides a detailed look at the attributes and immunologic mechanisms of plasmid DNA vaccines and their utility as laboratory tools as well as potential human vaccines. The immunogenicity and efficacy of DNA vaccines in a variety of preclinical models is used to illustrate how they differ from traditional vaccines in novel ways due to the in situ antigen production and the ease with which they are constructed. The ability to make new DNA vaccines without needing to handle a virulent pathogen or to adapt the pathogen for manufacturing purposes demonstrates the potential value of this vaccine technology for use against emerging and epidemic pathogens. Similarly, personalized anti-tumor DNA vaccines can also readily be made from a biopsy. Because DNA vaccines bias the T-helper (Th) cell response to a Th1 phenotype, DNA vaccines are also under development for vaccines against allergy and autoimmune diseases. The licensure of four animal health products, including two prophylactic vaccines against infectious diseases, one immunotherapy for cancer, and one gene therapy delivery of a hormone for a food animal, provides evidence of the efficacy of DNA vaccines in multiple species including horses and pigs. The size of these target animals provides evidence that the somewhat disappointing immunogenicity of DNA vaccines in a number of human clinical trials is not due simply to the larger mass of humans compared with most laboratory animals. The insights gained from the mechanisms of protection in the animal vaccines, the advances in the delivery and expression technologies for increasing the potency of DNA vaccines, and encouragingly potent human immune responses in certain clinical trials, provide insights for future efforts to develop DNA vaccines into a broadly useful vaccine and immunotherapy platform with applications for human and animal health.
Collapse
|
21
|
Lara AR, Knabben I, Regestein L, Sassi J, Caspeta L, Ramírez OT, Büchs J. Comparison of oxygen enriched air vs. pressure cultivations to increase oxygen transfer and to scale-up plasmid DNA production fermentations. Eng Life Sci 2011. [DOI: 10.1002/elsc.201000104] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
|
22
|
Channarong S, Chaicumpa W, Sinchaipanid N, Mitrevej A. Development and evaluation of chitosan-coated liposomes for oral DNA vaccine: the improvement of Peyer's patch targeting using a polyplex-loaded liposomes. AAPS PharmSciTech 2011; 12:192-200. [PMID: 21194014 DOI: 10.1208/s12249-010-9559-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2010] [Accepted: 11/30/2010] [Indexed: 11/30/2022] Open
Abstract
The aim of this study was to develop chitosan-coated and polyplex-loaded liposomes (PLLs) containing DNA vaccine for Peyer's patch targeting. Plain liposomes carrying plasmid pRc/CMV-HBs were prepared by the reverse-phase evaporation method. Chitosan coating was carried out by incubation of the liposomal suspensions with chitosan solution. Main lipid components of liposomes were phosphatidylcholine/cholesterol. Sodium deoxycholate and dicetyl phosphate were used as negative charge inducers. The zeta potentials of plain liposomes were strongly affected by the pH of the medium. Coating with chitosan variably increased the surface charges of the liposomes. To increase the zeta potential and stability of the liposome, chitosan was also used as a DNA condensing agent to form a polyplex. The PLLs were coated with chitosan solution. In vivo study of PLLs was carried out in comparison with chitosan-coated liposomes using plasmid encoding green fluorescence protein as a reporter. A single dose of plasmid equal to 100 μg was intragastrically inoculated into BALB/c mice. The expression of green fluorescence protein (GFP) was detected after 24 h using a confocal laser scanning microscope. The signal of GFP was obtained from positively charged chitosan-coated liposomes but found only at the upper part of duodenum. With chitosan-coated PLL540, the signal of GFP was found throughout the intestine. Chitosan-coated PLL demonstrated a higher potential to deliver the DNA to the distal intestine than the chitosan-coated liposomes due to the increase in permanent positive surface charges and the decreased enzymatic degradation.
Collapse
|
23
|
Clark JR, Bartley K, Jepson CD, Craik V, March JB. Comparison of a bacteriophage-delivered DNA vaccine and a commercially available recombinant protein vaccine against hepatitis B. ACTA ACUST UNITED AC 2011; 61:197-204. [PMID: 21204995 DOI: 10.1111/j.1574-695x.2010.00763.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A bacteriophage lambda DNA vaccine expressing the small surface antigen (HBsAg) of hepatitis B was compared with Engerix B, a commercially available vaccine based on the homologous recombinant protein (r-HBsAg). Rabbits (five per group) were vaccinated intramuscularly at weeks 0, 5 and 10. Antibody responses against r-HBsAg were measured by indirect enzyme-linked immunosorbent assay, by limiting dilutions and by subtyping. Specific lymphocyte proliferation in vitro was also measured. After one vaccination, three of the five phage-vaccinated rabbits showed a strong antibody response, whereas no r-HBsAg-vaccinated animals responded. Following two vaccinations, all phage-vaccinated animals responded and antibody levels remained high throughout the experiment (220 days total). By 2 weeks after the second vaccination, antibody responses were significantly higher (P<0.05) in the phage-vaccinated group in all tests. After three vaccinations, one out of five r-HBsAg-vaccinated rabbit still failed to respond. The recognized correlate of protection against hepatitis B infection is an antibody response against the HBsAg antigen. When combined with the fact that phage vaccines are potentially cheap to produce and stable at a range of temperatures, the results presented here suggest that further studies into the use of phage vaccination against hepatitis B are warranted.
Collapse
Affiliation(s)
- Jason R Clark
- BigDNA Ltd, Wallace Building, Roslin BioCentre, Roslin, UK.
| | | | | | | | | |
Collapse
|
24
|
Guan ZJ, Guo B, Huo YL, Guan ZP, Wei YH. Overview of expression of hepatitis B surface antigen in transgenic plants. Vaccine 2010; 28:7351-62. [PMID: 20850538 DOI: 10.1016/j.vaccine.2010.08.100] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2010] [Revised: 08/20/2010] [Accepted: 08/31/2010] [Indexed: 11/18/2022]
Abstract
Hepatitis B virus (HBV), a pathogen for chronic liver infection, afflicts more than 350 million people world-wide. The effective way to control the virus is to take HBV vaccine. Hepatitis B surface antigen (HBsAg) is an effective protective antigen suitable for vaccine development. At present, "edible" vaccine based on transgenic plants is one of the most promising directions in novel types of vaccines. HBsAg production from transgenic plants has been carried out, and the transgenic plant expression systems have developed from model plants (such as tobacco, potato and tomato) to other various plant platforms. Crude or purified extracts of transformed plants have been found to conduct immunological responses and clinical trials for hepatitis B, which gave the researches of plant-based HBsAg production a big boost. The aim of this review was to summarize the recent data about plant-based HBsAg development including molecular biology of HBsAg gene, selection of expression vector, the expression of HBsAg gene in plants, as well as corresponding immunological responses in animal models or human.
Collapse
Affiliation(s)
- Zheng-jun Guan
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education Northwest University, Xi'an 710069, China.
| | | | | | | | | |
Collapse
|
25
|
Pandey RS, Dixit VK. Evaluation of ISCOM vaccines for mucosal immunization against hepatitis B. J Drug Target 2010; 18:282-91. [PMID: 19958131 DOI: 10.3109/10611860903450015] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Immune stimulating complexes (ISCOMs) incorporating recombinant hepatitis B surface antigen (HBsAg) was prepared for induction of humoral, cellular and mucosal immunity by intranasal administration. Prepared ISCOMs were characterized for its size, shape, incorporation efficiency, zeta potential, and antigen integrity. Designed ISCOMs possessed negative zeta potential (-21.7 mV) and an average size of 44.1 nm with antigen incorporation efficiency approximately 39 %. Serum anti-HBsAg IgG titer after three high nasal doses of ISCOMs was comparable with titer recorded after alum-HBsAg administered subcutaneously. Similarly, modest but higher cellular response (cytokines level in spleen homogenates) and significantly higher secretory sIgA response in mucosal secretions was observed (P < 0.001) in case of HBsAg ISCOM vaccines. Whereas, alum-HBsAg vaccine did not elicit considerable cellular or mucosal response. Thus, ISCOMs produced humoral, mucosal, and cellular immune responses upon nasal administration although high and multidose administrations were required to elicit potent immune responses. These data demonstrate potential of ISCOMs in their use as a carrier adjuvant for nasal subunit vaccines against hepatitis B.
Collapse
Affiliation(s)
- R S Pandey
- Department of Pharmaceutical Sciences, Dr. Hari Singh Gour Vishwavidyalaya, Sagar - 470 003, Madhya Pradesh, India
| | | |
Collapse
|
26
|
Wu Y, Zhang Q, Sales D, Bianco AE, Craig A. Vaccination with peptide mimotopes produces antibodies recognizing bacterial capsular polysaccharides. Vaccine 2010; 28:6425-35. [PMID: 20674874 DOI: 10.1016/j.vaccine.2010.07.049] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2010] [Revised: 06/07/2010] [Accepted: 07/15/2010] [Indexed: 10/19/2022]
Abstract
A phage display peptide library was screened using a panel of antibodies to the capsular polysaccharides of Streptococcus agalactiae and Neisseria meningitidis. Mimotopes NPDHPRVPTFMA (2-8), LIPFHKHPHHRG (3-2) and EQEIFTNITDRV (G3) showing the highest binding capacity and strongest ELISA reaction were selected for immunization experiments. These mimotopes were either synthesised as oligodeoxynucleotides for DNA immunization or MAP (multiple antigen peptide) for peptide immunization. Mimotope-DNA vaccination, particularly for G3, induced antibodies recognizing a number of target bacteria. This response was seen after the second boost injection and was significantly enhanced by the 3rd boost injection with a Th1-associated profile, which was dominated by IgG2a, followed by IgG1. Mimotope-MAP immunization also produced strong humoral immune responses to the bacteria. Antibodies from G3 DNA immunization reacted with the surface molecules of S. agalactiae, N. meningitidis and Escherichia coli K5 shown by indirect immunofluorescence staining, indicating a possible localization to the bacterial capsule. Antibodies produced both from DNA/MAP immunization reacted with purified bacterial capsular polysaccharides by ELISA and were of high avidity. We have further characterized peptide G3 by a 'tiling path' study to examine the effect of changing individual residues in the peptide in raising antibodies, which showed that the EIFTN motif in G3 was important in generating antibodies to several capsulated bacteria. We conclude that mimotope immunization with DNA or MAP potentially induces strong antibody responses against encapsulated bacteria. It is suggested that the antibody targets are polysaccharides, and these antibodies may cross react at least among closely related species of bacteria.
Collapse
Affiliation(s)
- Yang Wu
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK.
| | | | | | | | | |
Collapse
|
27
|
Ingolotti M, Kawalekar O, Shedlock DJ, Muthumani K, Weiner DB. DNA vaccines for targeting bacterial infections. Expert Rev Vaccines 2010; 9:747-63. [PMID: 20624048 PMCID: PMC2962930 DOI: 10.1586/erv.10.57] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
DNA vaccination has been of great interest since its discovery in the 1990s due to its ability to elicit both humoral and cellular immune responses. DNA vaccines consist of a DNA plasmid containing a transgene that encodes the sequence of a target protein from a pathogen under the control of a eukaryotic promoter. This revolutionary technology has proven to be effective in animal models and four DNA vaccine products have recently been approved for veterinary use. Although few DNA vaccines against bacterial infections have been tested, the results are encouraging. Because of their versatility, safety and simplicity a wider range of organisms can be targeted by these vaccines, which shows their potential advantages to public health. This article describes the mechanism of action of DNA vaccines and their potential use for targeting bacterial infections. In addition, it provides an updated summary of the methods used to enhance immunogenicity from codon optimization and adjuvants to delivery techniques including electroporation and use of nanoparticles.
Collapse
Affiliation(s)
- Mariana Ingolotti
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
- Department of Anatomy, School of Biomedical Sciences, Austral University, Pilar, Buenos Aires, Argentina
| | - Omkar Kawalekar
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Devon J Shedlock
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Karuppiah Muthumani
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - David B Weiner
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
28
|
Carvalho JA, Rodgers J, Atouguia J, Prazeres DMF, Monteiro GA. DNA vaccines: a rational design against parasitic diseases. Expert Rev Vaccines 2010; 9:175-91. [PMID: 20109028 DOI: 10.1586/erv.09.158] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Parasitic diseases are one of the most devastating causes of morbidity and mortality worldwide. Although immunization against these infections would be an ideal solution, the development of effective vaccines has been hampered by specific challenges posed by parasitic pathogens. Plasmid-based DNA vaccines may prove to be promising immunization tools in this area because vectors can be designed to integrate several antigens from different stages of the parasite life cycle or different subspecies; vaccines, formulations and immunization protocols can be tuned to match the immune response that offers protective immunity; and DNA vaccination is an affordable platform for developing countries. Partial and full protective immunity have been reported following DNA vaccination against the most significant parasitic diseases in the world.
Collapse
Affiliation(s)
- Joana A Carvalho
- Instituto Superior Técnico, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | | | | | | | | |
Collapse
|
29
|
Xiang SD, Selomulya C, Ho J, Apostolopoulos V, Plebanski M. Delivery of DNA vaccines: an overview on the use of biodegradable polymeric and magnetic nanoparticles. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2010; 2:205-18. [DOI: 10.1002/wnan.88] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
30
|
Li L, Herndon JM, Truscott SM, Hansen TH, Fleming TP, Goedegebuure P, Gillanders WE. Engineering superior DNA vaccines: MHC class I single chain trimers bypass antigen processing and enhance the immune response to low affinity antigens. Vaccine 2010; 28:1911-8. [PMID: 20188246 PMCID: PMC2830906 DOI: 10.1016/j.vaccine.2009.10.096] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
It is commonly believed that delivery of antigen into the class I antigen presentation pathway is a limiting factor in the clinical translation of DNA vaccines. This is of particular concern in the context of cancer vaccine development as many immunodominant peptides derived from self tumor antigens are not processed and presented efficiently. To address this limitation, we have engineered completely assembled peptide/MHC class I complexes whereby all three components (class I heavy chain, beta(2)m, and peptide) are attached by flexible linkers and expressed as a single polypeptide (single chain trimers or SCT). In this study, we tested the efficacy of progressive generations of SCT DNA vaccines engineered to (1) enhance peptide binding, (2) enhance interaction with the CD8 coreceptor, and/or (3) activate CD4(+) helper T cells. Disulfide trap SCT (dtSCT) have been engineered to improve peptide binding, with mutations designed to create a disulfide bond between the class I heavy chain and the peptide linker. dtSCT DNA vaccines dramatically enhance the immune response to model low affinity antigens as measured by ELISPOT analysis and tumor challenge. SCT engineered to enhance interaction with the CD8 coreceptor have a higher affinity for the TCR/CD8 complex, and are associated with more robust CD8(+) T cell responses following vaccination. Finally, SCT constructs that coexpress a universal helper epitope PADRE, dramatically enhance CD8(+) T cell responses. Taken together, our data demonstrate that dtSCT DNA vaccines coexpressing a universal CD4 epitope are highly effective in generating immune responses to poorly processed and presented cancer antigens.
Collapse
Affiliation(s)
- Lijin Li
- Department of Surgery, Washington University School of Medicine
| | - John M. Herndon
- Department of Surgery, Washington University School of Medicine
| | - Steven M. Truscott
- Department of Pathology and Immunology, Washington University School of Medicine
| | - Ted H. Hansen
- Department of Pathology and Immunology, Washington University School of Medicine
- Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine
| | - Timothy P. Fleming
- Department of Surgery, Washington University School of Medicine
- Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine
| | - Peter Goedegebuure
- Department of Surgery, Washington University School of Medicine
- Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine
| | - William E. Gillanders
- Department of Surgery, Washington University School of Medicine
- Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine
| |
Collapse
|
31
|
Eratalay A, Coşkun-Ari FF, Öner F, Özcengīz E. In vitroandin vivoevaluations of PLGA microsphere vaccine formulations containing pDNA coexpressing Hepatitis B surface antigen and Interleukin-2. J Microencapsul 2010; 27:48-56. [DOI: 10.3109/02652040902937666] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
32
|
Abstract
OBJECTIVE To design a vaccine construct containing various but conserved HIV-1-derived epitopes and generating broad CD8 T cell responses. METHODS HLA-B7 transgenic H-2KD KO transgenic mice were used to identify potential new HLA-B07.02-restricted HIV-1-derived epitopes. Immunological recognition of these epitopes was confirmed by IFN-gamma ELISpot assays with PBMCs from HLA-B*0702 HIV-1-infected individuals. For these peptides as well as others previously identified, the capacity to induce cross-reactive responses against their frequent allelic variants was evaluated in the mouse model. A set of epitopes inducing strong T cell responses against various and conserved regions of HIV-1 was selected. A DNA vaccine was designed to express them as a unique antigen with or without a three amino acid ARY extension flanking each epitope. The spectrum of CD8 T responses generated by polyepitope constructs was tested in HLA-B7 transgenic mice. RESULTS Five new epitopes were identified in accessory and regulatory HIV-1 proteins. Twelve HLA-B07.02-restricted epitopes were selected on the basis of their structural conservation and cross-reactive immunogenicity. The ARY N-terminal extension flanking each epitope markedly increases their affinity for TAP and the use of this flanking extension in polyepitope vaccine has a sizable advantage to induce CD8 T cell cytotoxic responses in mice following DNA immunization. CONCLUSION The HLA-B7 mouse model allows to rapidly identify various HIV-1-derived epitopes of vaccine interest. Grouped in a polyepitope construct designed to increase their processing, this vaccine may be suitable for inducing multiple and relevant HIV-1-specific CTL responses in humans.
Collapse
|
33
|
Sällberg M, Frelin L, Weiland O. DNA vaccine therapy for chronic hepatitis C virus (HCV) infection: immune control of a moving target. Expert Opin Biol Ther 2009; 9:805-15. [PMID: 19527105 DOI: 10.1517/14712590902988444] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The use of DNA plasmids for DNA vaccination was first described in the early 1990 s. DNA vaccinations were successful in small animal models but in larger animals and humans problems appeared. One major obstacle, effective delivery, has been partly overcome by new delivery techniques, such as transdermal delivery with the gene gun, and in vivo electroporation. We are entering a new era of DNA vaccination, where such techniques can be tested in humans. DNA vaccination may be a useful therapy for chronic hepatitis C virus (HCV) infections. Patients with these infections have a reduced T cell response to the invading virus. The genetic variability of HCV, its immunomodulatory properties and high replication rate contribute to chronicity. By providing the correct stimulus T cells may be activated to clear the infection. The vaccination is intended to induce a coordinated immune-based attack on the continuously moving HCV target. If effective, this should help in clearing the infection.
Collapse
Affiliation(s)
- Matti Sällberg
- Karolinska Institutet at Karolinska University Hospital Huddinge, Division of Clinical Microbiology, F68, Department of Laboratory Medicine, S-141 86 Stockholm, Sweden.
| | | | | |
Collapse
|
34
|
Sanyal G, Shi L. A review of multiple approaches towards an improved hepatitis B vaccine. Expert Opin Ther Pat 2009; 19:59-72. [PMID: 19441898 DOI: 10.1517/13543770802587226] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Hepatitis B is a DNA virus that can cause liver inflammation, cirrhosis, and cancer in chronically infected and symptomatic carriers. Antiviral treatments are usually limited in their effectiveness in treating the disease states. Vaccination against hepatitis B in pediatric and adolescent populations has proven to be a generally effective means for preventing diseases that could be potentially caused by this virus. Some 5 - 10% of the vaccinees do not develop protective immunity against the virus. Therefore, a significant amount of effort has been made in many research laboratories across the world to increase the potency of the vaccine by various innovative means, e.g., increasing the immunogenicity of the antigen or through introduction of novel adjuvants that elicit strong humoral and cell-mediated immune responses. OBJECTIVES/METHODS The objective of this review is to highlight publications of significant developments that have been made over the past decade and efforts that are continuing towards producing an improved vaccine. A number of patents that protect novel hepatitis B vaccine formulations, including those claiming novel hepatitis B core antigen formulations and combinations of a vaccine with small molecule therapeutics, are discussed. CONCLUSION There have been promising developments in the area of new adjuvants and delivery systems. The practical need for reducing the total number of childhood vaccinations has driven development of, and patent filings on, multivalent and combination vaccine formulations in which the hepatitis B vaccine is included as one component. Efforts and some advances have also been made in the critical area of therapeutic application of the vaccine. The existence of a large population of already infected patients and the inadequacy of most of the current antiviral drugs against hepatitis B diseases have also inspired efforts to produce a vaccine that would be efficacious in clearing an exiting infection.
Collapse
Affiliation(s)
- Gautam Sanyal
- Infection Discovery, AstraZeneca R&D Boston, 35 Gatehouse Drive, Waltham, MA 02451, USA.
| | | |
Collapse
|
35
|
Poh WP, Narasaraju T, Pereira NA, Zhong F, Phoon MC, Macary PA, Wong SH, Lu J, Koh DR, Chow VTK. Characterization of cytotoxic T-lymphocyte epitopes and immune responses to SARS coronavirus spike DNA vaccine expressing the RGD-integrin-binding motif. J Med Virol 2009; 81:1131-9. [PMID: 19475608 PMCID: PMC7166745 DOI: 10.1002/jmv.21571] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Integrins are critical for initiating T‐cell activation events. The integrin‐binding motif Arg‐Gly‐Asp (RGD) was incorporated into the pcDNA 3.1 mammalian expression vector expressing the codon‐optimized extracellular domain of SARS coronavirus (SARS‐CoV) spike protein, and tested by immunizing C57BL/6 mice. Significant cell‐mediated immune responses were characterized by cytotoxic T‐lymphocyte 51Cr release assay and interferon‐gamma secretion ELISPOT assay against RMA‐S target cells presenting predicted MHC class I H2‐Kb epitopes, including those spanning residues 884–891 and 1116–1123 within the S2 subunit of SARS‐CoV spike protein. DNA vaccines incorporating the Spike‐RGD/His motif or the Spike‐His construct generated robust cell‐mediated immune responses. Moreover, the Spike‐His DNA vaccine construct generated a significant antibody response. Immunization with these DNA vaccine constructs elicited significant cellular and humoral immune responses. Additional T‐cell epitopes within the SARS‐CoV spike protein that may contribute to cell‐mediated immunity in vivo were also identified. J. Med. Virol. 81:1131–1139, 2009. © 2009 Wiley‐Liss, Inc.
Collapse
Affiliation(s)
- W P Poh
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore, Singapore
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Diminsky D, Reimann ZJ, Schirmbeck R, Barenholz Y. Structural and Functional Characterization of Liposomal Recombinant Hepatitis B Vaccine. J Liposome Res 2008. [DOI: 10.3109/08982109609031118] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
37
|
Enhanced in vivo transgene expression and immunogenicity from plasmid vectors following electrostimulation in rodents and primates. Vaccine 2008; 26:5202-9. [DOI: 10.1016/j.vaccine.2008.03.058] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
38
|
Abstract
This unit details some of the key methods for setting up and testing DNA vaccines in animal models. The basic procedures are discussed, as well as alternative methods that have been developed over the past several years. The Basic Protocol gives step-by-step instructions for administering the DNA vaccine via intramuscular injection of the quadriceps muscle, while an alternate procedure details injection of the anterior tibialis.
Collapse
|
39
|
Geng LY, Fang M, Yi JM, Jiang F, Moeen-ud-Din M, Yang LG. Effect of overexpression of inhibin α (1–32) fragment on bovine granulosa cell proliferation, apoptosis, steroidogenesis, and development of co-cultured oocytes. Theriogenology 2008; 70:35-43. [DOI: 10.1016/j.theriogenology.2008.02.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2007] [Revised: 01/24/2008] [Accepted: 02/15/2008] [Indexed: 10/22/2022]
|
40
|
Yang K, Whalen BJ, Tirabassi RS, Selin LK, Levchenko TS, Torchilin VP, Kislauskis EH, Guberski DL. A DNA vaccine prime followed by a liposome-encapsulated protein boost confers enhanced mucosal immune responses and protection. THE JOURNAL OF IMMUNOLOGY 2008; 180:6159-67. [PMID: 18424737 DOI: 10.4049/jimmunol.180.9.6159] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
A variety of DNA vaccine prime and recombinant viral boost immunization strategies have been developed to enhance immune responses in humans, but inherent limitations to these strategies exist. There is still an overwhelming need to develop safe and effective approaches that raise broad humoral and T cell-mediated immune responses systemically and on mucosal surfaces. We have developed a novel mucosal immunization regimen that precludes the use of viral vectors yet induces potent T cell responses. Using hepatitis B surface Ag (HBsAg), we observed that vaccination of BALB/c mice with an i.m. HBsAg-DNA vaccine prime followed by an intranasal boost with HBsAg protein encapsulated in biologically inert liposomes enhanced humoral and T cell immune responses, particularly on mucosal surfaces. Intranasal live virus challenge with a recombinant vaccinia virus expressing HBsAg revealed a correlation between T cell immune responses and protection of immunized mice. A shortened immunization protocol was developed that was successful in both adult and neonatal mice. These results support the conclusion that this new approach is capable of generating a Th-type-1-biased, broad spectrum immune response, specifically at mucosal surfaces. The success of this design may provide a safe and effective vaccination alternative for human use.
Collapse
Affiliation(s)
- Kejian Yang
- Oral Vaccine Institute, 10 New Bond Street, Worcester, MA 01606, USA.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Plasmid DNA loaded chitosan nanoparticles for nasal mucosal immunization against hepatitis B. Int J Pharm 2008; 354:235-41. [DOI: 10.1016/j.ijpharm.2007.11.027] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2007] [Revised: 11/01/2007] [Accepted: 11/17/2007] [Indexed: 11/19/2022]
|
42
|
Lu S, Wang S, Grimes-Serrano JM. Current progress of DNA vaccine studies in humans. Expert Rev Vaccines 2008; 7:175-91. [PMID: 18324888 DOI: 10.1586/14760584.7.2.175] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Despite remarkable progress in the field of DNA vaccine research since its discovery in the early 1990 s, the formal acceptance of this novel technology as a new modality of human vaccines depends on the successful demonstration of its safety and efficacy in advanced clinical trials. Although clinical trials conducted so far have provided overwhelming evidence that DNA vaccines are well tolerated and have an excellent safety profile, the early designs of DNA vaccines failed to demonstrate sufficient immunogenicity in humans. However, studies conducted over the last few years have led to promising results, particularly when DNA vaccines were used in combination with other forms of vaccines. Here, we provide a review of the data from reported DNA vaccine clinical studies with an emphasis on the ability of DNA vaccines to elicit antigen-specific, cell-mediated and antibody responses in humans. The majority of these trials are designed to test candidate vaccines against several major human pathogens and the remaining studies tested the immunogenicity of therapeutic vaccines against cancer.
Collapse
Affiliation(s)
- Shan Lu
- Department of Medicine, University of Massachusetts Medical School, 364 Plantation Street, LRB 304, Worcester, MA 01605, USA.
| | | | | |
Collapse
|
43
|
Khatri K, Goyal AK, Gupta PN, Mishra N, Mehta A, Vyas SP. Surface modified liposomes for nasal delivery of DNA vaccine. Vaccine 2008; 26:2225-33. [PMID: 18396362 DOI: 10.1016/j.vaccine.2008.02.058] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2007] [Revised: 02/14/2008] [Accepted: 02/21/2008] [Indexed: 10/22/2022]
Abstract
The aim of the present work was to investigate the potential utility of glycol chitosan coated liposomes as nasal vaccine delivery vehicle for eliciting viral specific humoral mucosal and cellular immune responses. Plasmid pRc/CMV-HBs(S) encapsulated liposomes were prepared by dehydration-rehydration method and subsequently coated with glycol chitosan by simple incubation method. Liposomes were then characterized for their size, surface charge, entrapment efficiency, and ability to protect encapsulated DNA against nuclease digestion and for their mucoadhesiveness. The liposomes were then administered to mice in order to study their feasibility as nasal vaccine carriers. The developed liposomes possessed +9.8 mV zeta potential and an average vesicle size less than 1 microm and entrapment efficiency of approximately 53%. Following intranasal administration, glycol chitosan coated liposomes elicited humoral mucosal and cellular immune responses that were significant as compared to naked DNA justifying the potential advantage of mucosal vaccination in the production of local antibodies at the sites where pathogens enters the body.
Collapse
Affiliation(s)
- Kapil Khatri
- Drug Delivery Research Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar 470003, M.P., India
| | | | | | | | | | | |
Collapse
|
44
|
Phillips S. Section Reviews: Biologicals & Immunologicals: Recent advances in non-viral gene therapy. Expert Opin Investig Drugs 2008. [DOI: 10.1517/13543784.5.9.1101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
45
|
Donnelly JJ, Ulmer JB, Liu MA. Overview: Biologicals & Immunologicals: Recombinant vaccines: technology and applications. Expert Opin Ther Pat 2008. [DOI: 10.1517/13543776.5.3.211] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
46
|
Qu D, Lanier G, Yuan ZH, Wen YM, Howard CR, Ahmed R. Localization of CD8+ cells specific for hepatitis B virus surface protein in the liver of immunized mice. J Med Virol 2008; 80:225-32. [PMID: 18098130 DOI: 10.1002/jmv.21039] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
DNA plasmids are potent inducers of long-lasting antigen-specific CTL responses. Little is known about the distribution of antigen-specific CD8+ T cells in the lymphoid tissue and the non-lymphoid tissue after DNA immunization. HBsAg-specific CD8+ T cells in peripheral blood mononuclear cells, spleen, lymph nodes, and the liver of Balb/c mice have been quantified after injection with a DNA plasmid expressing the major S protein of hepatitis B virus (HBV). The kinetics of CD8+ T-cell responses in the circulation were measured after priming and boosting, showing that antigen-specific CD8+ T cells undergo first expansion and then decline to a sustainable level in the circulation, although the frequencies of HBsAg-specific CD8+ T cells in the circulation were lower than for the spleen. The greater frequencies of HBsAg-specific CD8+ T cells were found in the liver, whereas the largest numbers of antigen-specific CD8+ T cells were found in the spleen. By day 100 after priming, HBsAg-specific CD8+ T cells were still detected in the circulation, the spleen and the liver. After boosting with the same plasmid DNA immunogen, HBsAg-specific CD8+ T cells proliferated quickly and vigorously. By 150 days after boosting, HBsAg-specific memory CD8+ T cells were sustained at higher levels than those recorded after the first, primary injection, both in the spleen and the liver: anti-HBs antibody-secreting plasma cells persisted in the bone marrow and in the spleen, consistent with the detection of anti-HBs antibodies detected in the blood. These findings indicate that DNA immunization has considerable potential for inducing specific T cell responses in the liver and offers a strategy for the development of post-exposure immunotherapy against persistent hepatitis B infections.
Collapse
Affiliation(s)
- Di Qu
- Department of Medical Molecular Virology, Institutes of Bio-medical Sciences, Shanghai Medical College of Fudan University, Shanghai, People's Republic of China
| | | | | | | | | | | |
Collapse
|
47
|
Development and evaluation of a novel DNA vaccine expressing inhibin alpha (1-32) fragment for improving the fertility in rats and sheep. Anim Reprod Sci 2007; 109:251-65. [PMID: 18243599 DOI: 10.1016/j.anireprosci.2007.12.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2007] [Revised: 12/11/2007] [Accepted: 12/12/2007] [Indexed: 11/23/2022]
Abstract
Inhibin is an important protein hormone in regulating folliculogenesis. Immunization against inhibin can improve follicle developments. The objective of present study is to investigate inhibin DNA immunization as a potential tool for improving follicle development and litter sizes of female animals. In our study, the inhibin DNA vaccine was constructed with inhibin alpha (1-32) fragment inserted into the C termination of HBsAg-S. Ninety rats and forty sheep were immunized with inhibin DNA vaccine. In rats, immunization against inhibin resulted in increase of positive sera ratio (P<0.01). The treatment was accompanied by a significant increase in the total number of mature follicles of >0.8mm in diameter on the onset of estrous cycle after twice immunization (31.0+/-3.9 in the test groups versus 27.4+/-5 in control groups) and after third immunization (35.2+/-6.7 in the test groups versus 30.3+/-5.2 in control groups). Litter sizes were significantly (P<0.05) bigger in rats treated with inhibin DNA vaccine (12.7+/-4.5 n=6) than in control (9.8+/-4.5). In sheep, twinning rate in test groups (39.2%) was significantly higher than that in control groups (10%) after immunization (P<0.05). These results indicated that inhibin was an important factor in improvement of fertility in rats and sheep, and demonstrated that DNA immunization against inhibin could induce more mature follicles resulting in increased litter sizes. Our results revealed that inhibin DNA vaccine may be an alternative to the use of exogenous gonadotrophins for increasing ovarian follicular development and improving animal fertility.
Collapse
|
48
|
Haigh O, Guo H, Edgtton K, Mather M, Herd KA, Tindle RW. Multiple copies of a tumor epitope in a recombinant hepatitis B surface antigen (HBsAg) vaccine enhance CTL responses, but not tumor protection. Virology 2007; 368:363-75. [PMID: 17689584 DOI: 10.1016/j.virol.2007.06.041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2007] [Revised: 05/24/2007] [Accepted: 06/04/2007] [Indexed: 01/12/2023]
Abstract
We propose the replacement of endogenous epitopes with foreign epitopes to exploit the highly immunogenic hepatitis B surface antigen (HBsAg) as a vaccine vector to elicit disease-protective cytotoxic T-lymphocyte (CTL) responses. Locations were defined within the HBsAg gene where replacements of DNA encoding HBsAg epitopes may be made to generate functional recombinant (r) HBsAg DNA vaccines. We demonstrate that rHBsAg DNA vaccines encoding multiple copies of a model tumor epitope from human papillomavirus (HPV) elicit enhanced CTL responses compared to rHBsAg DNA vaccines encoding a single copy. We show that rHBsAg DNA vaccines elicit a marked prophylactic and long-lived therapeutic protection against epitope expressing tumor, although protective efficacy was not improved by increasing the number of copies of the tumor epitope DNA. These results demonstrate the efficacy of HBsAg as a vector for the delivery of foreign CTL epitopes using the epitope replacement strategy, and have implications for rHBsAg vaccine design. The results also have implications for the derivation of a therapeutic vaccine for HPV-associated squamous carcinoma.
Collapse
MESH Headings
- Animals
- Carcinoma, Squamous Cell/immunology
- Carcinoma, Squamous Cell/prevention & control
- Cell Line
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/immunology
- Female
- Gene Dosage
- Genetic Vectors
- Hepatitis B Surface Antigens/genetics
- Humans
- Immunization
- Mice
- Mice, Inbred C57BL
- Oncogene Proteins, Viral/genetics
- Oncogene Proteins, Viral/immunology
- Papillomavirus E7 Proteins
- T-Lymphocytes, Cytotoxic/immunology
- Uterine Cervical Neoplasms/immunology
- Uterine Cervical Neoplasms/prevention & control
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/immunology
Collapse
Affiliation(s)
- Oscar Haigh
- Sir Albert Sakzewski Virus Research Centre, Royal Children's Hospital Herston Road, Herston, QLD 4029, Australia
| | | | | | | | | | | |
Collapse
|
49
|
Luxembourg A, Evans CF, Hannaman D. Electroporation-based DNA immunisation: translation to the clinic. Expert Opin Biol Ther 2007; 7:1647-64. [DOI: 10.1517/14712598.7.11.1647] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
50
|
Tang L, Yan Z, Wan Y, Han W, Zhang Y. Myostatin DNA vaccine increases skeletal muscle mass and endurance in mice. Muscle Nerve 2007; 36:342-8. [PMID: 17587222 DOI: 10.1002/mus.20791] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Myostatin is a transforming growth factor-beta family member that acts as a negative regulator of skeletal muscle growth. In mice, genetic disruption of the myostatin gene leads to a marked increase in body weight and muscle mass. Similarly, pharmacological interference with myostatin in vivo in mdx knockout mice results in a functional improvement of the dystrophic phenotype. Consequently, myostatin is an important therapeutic target for treatment of diseases associated with muscle wasting. To construct a therapeutic DNA vaccine against myostatin, we coupled the foreign, immunodominant T-helper epitope of tetanus toxin to the N terminus of myostatin, and BALB/c mice were immunized with the recombinant vector. Sera from vaccinated mice showed the presence of specific antibodies against the recombinant protein. In addition, body weight, muscle mass, and grip endurance of vaccinated mice were significantly increased. Our study provides a novel, pharmacological strategy for treatment of diseases associated with muscle wasting.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antibodies/blood
- Antigen Presentation
- Body Weight
- Epitopes, T-Lymphocyte/chemistry
- Epitopes, T-Lymphocyte/genetics
- Genetic Vectors
- Mice
- Mice, Inbred BALB C
- Molecular Sequence Data
- Muscle, Skeletal/anatomy & histology
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/immunology
- Myostatin
- Organ Size
- Peptide Elongation Factor 1/genetics
- Physical Endurance/drug effects
- Promoter Regions, Genetic
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- Tetanus Toxin/genetics
- Tetanus Toxin/immunology
- Transforming Growth Factor beta/antagonists & inhibitors
- Transforming Growth Factor beta/genetics
- Transforming Growth Factor beta/immunology
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/genetics
- Vaccines, DNA/immunology
Collapse
Affiliation(s)
- Liang Tang
- Biotechnology Center, Fourth Military Medical University, 17 Changle West Road, 710032 Xi'an, China.
| | | | | | | | | |
Collapse
|