1
|
Spunde K, Korotkaja K, Sominskaya I, Zajakina A. Genetic adjuvants: A paradigm shift in vaccine development and immune modulation. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102536. [PMID: 40336572 PMCID: PMC12056970 DOI: 10.1016/j.omtn.2025.102536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
The COVID-19 pandemic underscored the urgency of developing effective vaccines to combat infectious diseases, especially in vulnerable populations such as the elderly and immunocompromised. While recombinant protein vaccines offer safety, their poor immunogenicity highlights the need for advanced vaccination platforms. New genetic/nucleic acid vaccine formulations like plasmid DNA and mRNA showed efficiency and safety in preclinical and clinical studies; however, they demand innovative adjuvants because their mechanism of action differs from traditional protein vaccines. Genetic adjuvants-encoded by nucleic acids within DNA, RNA, or viral vectors-emerge as a promising solution by targeting and modulating specific immune pathways, including antigen presentation, T cell activation, and memory formation. These innovative adjuvants enhance vaccine efficacy by fine-tuning innate and adaptive immune responses, overcoming immune senescence, and addressing the challenges of CD8+ T cell activation in immunocompromised populations. This review explores the potential of genetically encoded adjuvants, including cytokines, chemokines, and other immune modulators. By comparing these adjuvants to traditional formulations, we highlight their capacity to address the limitations of modern vaccines while discussing their integration with emerging technologies like RNA-based vaccines. As genetic adjuvants advance toward clinical application, understanding their mechanisms and optimizing their delivery is pivotal to unlocking next-generation immunization strategies.
Collapse
Affiliation(s)
- Karina Spunde
- Cancer Gene Therapy Group, Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k. 1, LV-1067 Riga, Latvia
| | - Ksenija Korotkaja
- Cancer Gene Therapy Group, Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k. 1, LV-1067 Riga, Latvia
| | - Irina Sominskaya
- Cancer Gene Therapy Group, Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k. 1, LV-1067 Riga, Latvia
| | - Anna Zajakina
- Cancer Gene Therapy Group, Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k. 1, LV-1067 Riga, Latvia
| |
Collapse
|
2
|
Sakai Y, Yamada S, Inoue M, Shiga T, Konagayoshi K, Kasai K, Kimura A, Murakami K. Efficacy of a DNA vaccine encoding the E2 glycoprotein of bovine viral diarrhea virus 1 fused to mouse lysosome-associated membrane protein 1. Vet Microbiol 2024; 298:110283. [PMID: 39488135 DOI: 10.1016/j.vetmic.2024.110283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/17/2024] [Accepted: 10/20/2024] [Indexed: 11/04/2024]
Abstract
The E2 protein of bovine viral diarrhea virus (BVDV) is a known protective antigen and a major target for DNA vaccines. DNA vaccines have various advantages; however, their immunogenicity needs to be enhanced by using adjuvants or drug delivery systems. In this study, we used mouse lysosome-associated membrane protein 1 (mLAMP1) as a molecular adjuvant and developed a DNA vaccine encoding an mLAMP1-BVDV E2 chimeric protein (pVax-mLAMP1-E2). We constructed DNA plasmids in which the E2 gene was inserted within the hinge region (H) or membrane proximal domain (D) of the mLAMP1 gene. Transfection of these plasmids into cultured cells led to high expression of E2 antigen from pVax-mLAMP1-E2 (H). Intradermal immunization of mice with pVax-mLAMP1-E2 (H) induced sufficient neutralizing antibodies and splenocytes with E2 antigen-specific IFN-γ production compared with pVax-mLAMP1-E2 (D). However, the immunogenicity of pVax mLAMP1-E2 (H) in mice did not differ from that of a control plasmid without the LAMP1 molecule (pVax-E2). In cattle, geometric mean serum neutralizing antibody titers after intradermal or intramuscular injection tended to be higher with pVax-mLAMP1-E2 (H) than with pVax that expressed E2 without mLAMP1. In addition, E2 antigen-specific IFN-γ production in peripheral blood mononuclear cells from cattle immunized intradermally with pVax-mLAMP1-E2 (H) was not significantly different from that of pVax-E2. These results suggest that mLAMP1 fusion antigens effectively induce humoral and cellular immunity in mice and cattle, especially when the antigen is inserted in the hinge region of mLAMP1. The LAMP1-E2 fusion antigen may be a useful candidate for a BVDV DNA vaccine in cattle.
Collapse
Affiliation(s)
- Yusuke Sakai
- Graduate School of Veterinary Sciences, Iwate University, Morioka, Iwate 020-8550, Japan; Nippon Zenyaku Kogyo Co., Ltd., Koriyama, Fukushima 963-0196, Japan
| | - Shinji Yamada
- Graduate School of Veterinary Sciences, Iwate University, Morioka, Iwate 020-8550, Japan; Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Iwate University, Morioka, Iwate 020-8550, Japan; Farm Animal Clinic and Disease Control Center, Faculty of Agriculture, Iwate University, Morioka, Iwate 020-8550, Japan
| | - Maho Inoue
- Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Iwate University, Morioka, Iwate 020-8550, Japan
| | - Toshinori Shiga
- Nippon Zenyaku Kogyo Co., Ltd., Koriyama, Fukushima 963-0196, Japan
| | | | - Kei Kasai
- Nippon Zenyaku Kogyo Co., Ltd., Koriyama, Fukushima 963-0196, Japan
| | - Atsushi Kimura
- Graduate School of Veterinary Sciences, Iwate University, Morioka, Iwate 020-8550, Japan; Farm Animal Clinic and Disease Control Center, Faculty of Agriculture, Iwate University, Morioka, Iwate 020-8550, Japan
| | - Kenji Murakami
- Graduate School of Veterinary Sciences, Iwate University, Morioka, Iwate 020-8550, Japan; Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Iwate University, Morioka, Iwate 020-8550, Japan; Farm Animal Clinic and Disease Control Center, Faculty of Agriculture, Iwate University, Morioka, Iwate 020-8550, Japan.
| |
Collapse
|
3
|
Yamaguchi F, Sakane H, Akasaki K. Comparative study of the steady-state subcellular distribution of lysosome-associated membrane glycoprotein-2 (LAMP-2) isoforms with GYXXΦ-type tyrosine-based motifs that interact differently with four adaptor protein (AP) complexes. J Biochem 2024; 175:275-287. [PMID: 37983719 DOI: 10.1093/jb/mvad096] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/23/2023] [Accepted: 11/06/2023] [Indexed: 11/22/2023] Open
Abstract
Lysosome-associated membrane protein-1 and -2 (LAMP-1 and LAMP-2, respectively) are type I transmembrane proteins. LAMP-2 comprises three splice isoforms (LAMP-2A, -B and-C) with different cytoplasmic tails (CTs). These three CTs possess different tyrosine-based motifs (GYXXΦ, where Φ is a bulky hydrophobic amino acid) at their C-termini. Interactions between tyrosine-based motifs and μ-subunits of four tetrameric adaptor protein (AP) complexes are necessary for their vesicular transport to lysosomes. Little is known about how the interaction strengths of these tyrosine motifs with μ-subunits affect the localization of isoforms to lysosomes. The interactions were first investigated using a yeast two-hybrid system to address this question. LAMP-2A-CT interacted with all four μ-subunits (μ1, μ2, μ3A and μ4 of AP-1, AP-2, AP-3 and AP-4, respectively). The interaction with μ3A was more robust than that with other μ-subunits. LAMP-2B-CT interacted exclusively and moderately with μ3A. LAMP-2C-CT did not detectably interact with any of the four μ-subunits. Immunofluorescence microscopy showed that all isoforms were localized in late endosomes and lysosomes. LAMP-2C was present in the plasma membrane and early endosomes; however, LAMP-2A and -2B were barely detectable in these organelles. In cell fractionation, LAMP-2A was the most abundant in the dense lysosomes, whereas LAMP-2C was significantly present in the low-density fraction containing the plasma membrane and early endosomes, in addition to the dense lysosomes. LAMP-2B considerably existed in the low-density late endosomal fraction. These data strongly suggest that the LAMP-2 isoforms are distributed differently in endocytic organelles depending on their interaction strengths with AP-3.
Collapse
Affiliation(s)
- Fumiaki Yamaguchi
- Faculty of Pharmacy and Pharmaceutical Sciences, Fukuyama University, Fukuyama, Hiroshima 729-0292, Japan
| | - Hiroshi Sakane
- Faculty of Pharmacy and Pharmaceutical Sciences, Fukuyama University, Fukuyama, Hiroshima 729-0292, Japan
| | - Kenji Akasaki
- Faculty of Pharmacy and Pharmaceutical Sciences, Fukuyama University, Fukuyama, Hiroshima 729-0292, Japan
| |
Collapse
|
4
|
Teixeira FME, Oliveira LDM, Branco ACCC, Alberca RW, de Sousa ESA, Leite BHDS, Adan WCDS, Duarte AJDS, Lins RD, Sato MN, Viana IFT. Enhanced immunogenicity and protective efficacy in mice following a Zika DNA vaccine designed by modulation of membrane-anchoring regions and its association to adjuvants. Front Immunol 2024; 15:1307546. [PMID: 38361945 PMCID: PMC10867427 DOI: 10.3389/fimmu.2024.1307546] [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: 10/04/2023] [Accepted: 01/04/2024] [Indexed: 02/17/2024] Open
Abstract
Zika virus (ZIKV) is a re-emerging pathogen with high morbidity associated to congenital infection. Despite the scientific advances since the last outbreak in the Americas, there are no approved specific treatment or vaccines. As the development of an effective prophylactic approach remains unaddressed, DNA vaccines surge as a powerful and attractive candidate due to the efficacy of sequence optimization in achieving strong immune response. In this study, we developed four DNA vaccine constructs encoding the ZIKV prM/M (pre-membrane/membrane) and E (envelope) proteins in conjunction with molecular adjuvants. The DNA vaccine candidate (called ZK_ΔSTP), where the entire membrane-anchoring regions were completely removed, was far more immunogenic compared to their counterparts. Furthermore, inclusion of the tPA-SP leader sequence led to high expression and secretion of the target vaccine antigens, therefore contributing to adequate B cell stimulation. The ZK_ΔSTP vaccine induced high cellular and humoral response in C57BL/6 adult mice, which included high neutralizing antibody titers and the generation of germinal center B cells. Administration of ZK-ΔSTP incorporating aluminum hydroxide (Alum) adjuvant led to sustained neutralizing response. In consistency with the high and long-term protective response, ZK_ΔSTP+Alum protected adult mice upon viral challenge. Collectively, the ZK_ΔSTP+Alum vaccine formulation advances the understanding of the requirements for a successful and protective vaccine against flaviviruses and is worthy of further translational studies.
Collapse
Affiliation(s)
- Franciane Mouradian Emidio Teixeira
- Laboratory of Dermatology and Immunodeficiencies, LIM-56, Department of Dermatology, Tropical Medicine Institute of São Paulo, University of São Paulo Medical School, São Paulo, Brazil
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Luana de Mendonça Oliveira
- Laboratory of Dermatology and Immunodeficiencies, LIM-56, Department of Dermatology, Tropical Medicine Institute of São Paulo, University of São Paulo Medical School, São Paulo, Brazil
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Anna Cláudia Calvielli Castelo Branco
- Laboratory of Dermatology and Immunodeficiencies, LIM-56, Department of Dermatology, Tropical Medicine Institute of São Paulo, University of São Paulo Medical School, São Paulo, Brazil
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Ricardo Wesley Alberca
- Laboratory of Dermatology and Immunodeficiencies, LIM-56, Department of Dermatology, Tropical Medicine Institute of São Paulo, University of São Paulo Medical School, São Paulo, Brazil
| | - Emanuella Sarmento Alho de Sousa
- Laboratory of Dermatology and Immunodeficiencies, LIM-56, Department of Dermatology, Tropical Medicine Institute of São Paulo, University of São Paulo Medical School, São Paulo, Brazil
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | | | - Alberto José da Silva Duarte
- Laboratory of Dermatology and Immunodeficiencies, LIM-56, Department of Dermatology, Tropical Medicine Institute of São Paulo, University of São Paulo Medical School, São Paulo, Brazil
| | - Roberto Dias Lins
- Department of Virology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation, Recife, Brazil
| | - Maria Notomi Sato
- Laboratory of Dermatology and Immunodeficiencies, LIM-56, Department of Dermatology, Tropical Medicine Institute of São Paulo, University of São Paulo Medical School, São Paulo, Brazil
| | | |
Collapse
|
5
|
Xu S, Wang Y, Wang Y, Jiang Y, Li H, Han C, Wei B, Qin Q, Wei S. Development and immune evaluation of LAMP1 chimeric DNA vaccine against Singapore grouper iridovirus in orange-spotted grouper, Epinephelus coioides. FISH & SHELLFISH IMMUNOLOGY 2024; 144:109218. [PMID: 37977543 DOI: 10.1016/j.fsi.2023.109218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 10/24/2023] [Accepted: 11/06/2023] [Indexed: 11/19/2023]
Abstract
Grouper is one of the most important and valuable mariculture fish in China, with a high economic value. As the production of grouper has increased, massive outbreaks of epidemic diseases have limited the development of the industry. Singapore grouper iridovirus (SGIV) is one of the most serious infectious viral pathogens and has caused huge economic losses to grouper farming worldwide due to its rapid spread and high lethality. To find new strategies for the effective prevention and control of SGIV, we constructed two chimeric DNA vaccines using Lysosome-associated membrane protein 1 (LAMP1) fused with major capsid proteins (MCP) against SGIV. In addition, we evaluated the immune protective effects of vaccines including pcDNA3.1-3HA, pcDNA3.1-MCP, pcDNA3.1-LAMP1, chimeric DNA vaccine pcDNA3.1-MLAMP and pcDNA3.1-LAMCP by intramuscular injection. Our results showed that compared with groups injected with PBS, pcDNA3.1-3HA, pcDNA3.1-LAMP1 or pcDNA3.1-MCP, the antibody titer significantly increased in the chimeric vaccine groups. Moreover, the mRNA levels of immune-related factors in groupers, including IRF3, MHC-I, TNF-α, and CD8, showed the same trend. However, MHC-II and CD4 were significantly increased only in the chimeric vaccine groups. After 28 days of vaccination, groupers were challenged with SGIV, and mortality was documented for each group within 14 days. The data showed that two chimeric DNA vaccines provided 87 % and 91 % immune protection for groupers which were significantly higher than the 52 % protection rate of pcDNA3.1-MCP group, indicating that both forms of LAMP1 chimeric vaccines possessed higher immune protection against SGIV, providing the theoretical foundation for the creation of novel DNA vaccines for fish.
Collapse
Affiliation(s)
- SuiFeng Xu
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - YueXuan Wang
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - YeWen Wang
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - YunXiang Jiang
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Huang Li
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - ChengZong Han
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - BaoCan Wei
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Qiwei Qin
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China; Nansha-South China Agricultural University Fishery Research Institute, Guangzhou, 511457, China.
| | - Shina Wei
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China; Nansha-South China Agricultural University Fishery Research Institute, Guangzhou, 511457, China.
| |
Collapse
|
6
|
Meng Z, Rodriguez Ehrenfried A, Tan CL, Steffens LK, Kehm H, Zens S, Lauenstein C, Paul A, Schwab M, Förster JD, Salek M, Riemer AB, Wu H, Eckert C, Leonhardt CS, Strobel O, Volkmar M, Poschke I, Offringa R. Transcriptome-based identification of tumor-reactive and bystander CD8 + T cell receptor clonotypes in human pancreatic cancer. Sci Transl Med 2023; 15:eadh9562. [PMID: 37967201 DOI: 10.1126/scitranslmed.adh9562] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 10/16/2023] [Indexed: 11/17/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is generally refractory to immune checkpoint blockade, although patients with genetically unstable tumors can show modest therapeutic benefit. We previously demonstrated the presence of tumor-reactive CD8+ T cells in PDAC samples. Here, we charted the tumor-infiltrating T cell repertoire in PDAC by combining single-cell transcriptomics with functional testing of T cell receptors (TCRs) for reactivity against autologous tumor cells. On the basis of a comprehensive dataset including 93 tumor-reactive and 65 bystander TCR clonotypes, we delineated a gene signature that effectively distinguishes between these T cell subsets in PDAC, as well as in other tumor indications. This revealed a high frequency of tumor-reactive TCR clonotypes in three genetically unstable samples. In contrast, the T cell repertoire in six genetically stable PDAC tumors was largely dominated by bystander T cells. Nevertheless, multiple tumor-reactive TCRs were successfully identified in each of these samples, thereby providing a perspective for personalized immunotherapy in this treatment-resistant indication.
Collapse
Affiliation(s)
- Zibo Meng
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, 69120 Heidelberg, Germany
- Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, China
| | - Aaron Rodriguez Ehrenfried
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, 69120 Heidelberg, Germany
- Helmholtz-Institute for Translational Oncology by DKFZ (HI-TRON), 55131 Mainz, Germany
- Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Chin Leng Tan
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, 69120 Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Laura K Steffens
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, 69120 Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Hannes Kehm
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, 69120 Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Stefan Zens
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, 69120 Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Claudia Lauenstein
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Alina Paul
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, 69120 Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Marius Schwab
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Jonas D Förster
- Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
- Division of Immunotherapy & Immunoprevention, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Molecular Vaccine Design, German Center for Infection Research (DZIF), partner site Heidelberg, 69120 Heidelberg, Germany
| | - Mogjiborahman Salek
- Division of Immunotherapy & Immunoprevention, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Molecular Vaccine Design, German Center for Infection Research (DZIF), partner site Heidelberg, 69120 Heidelberg, Germany
| | - Angelika B Riemer
- Division of Immunotherapy & Immunoprevention, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Molecular Vaccine Design, German Center for Infection Research (DZIF), partner site Heidelberg, 69120 Heidelberg, Germany
| | - Heshui Wu
- Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, China
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, China
| | - Christoph Eckert
- Pathology Institute, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Carl-Stephan Leonhardt
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Oliver Strobel
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Michael Volkmar
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, 69120 Heidelberg, Germany
- Helmholtz-Institute for Translational Oncology by DKFZ (HI-TRON), 55131 Mainz, Germany
| | - Isabel Poschke
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, 69120 Heidelberg, Germany
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, 69120 Heidelberg, Germany
- Immune Monitoring Unit, National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
| | - Rienk Offringa
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, 69120 Heidelberg, Germany
- Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, China
| |
Collapse
|
7
|
Zhu P, Li SY, Ding J, Fei Z, Sun SN, Zheng ZH, Wei D, Jiang J, Miao JL, Li SZ, Luo X, Zhang K, Wang B, Zhang K, Pu S, Wang QT, Zhang XY, Wen GL, Liu JO, August JT, Bian H, Chen ZN, He YW. Combination immunotherapy of glioblastoma with dendritic cell cancer vaccines, anti-PD-1 and poly I:C. J Pharm Anal 2023; 13:616-624. [PMID: 37440907 PMCID: PMC10334272 DOI: 10.1016/j.jpha.2023.04.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 04/13/2023] [Accepted: 04/18/2023] [Indexed: 07/15/2023] Open
Abstract
Glioblastoma (GBM) is a lethal cancer with limited therapeutic options. Dendritic cell (DC)-based cancer vaccines provide a promising approach for GBM treatment. Clinical studies suggest that other immunotherapeutic agents may be combined with DC vaccines to further enhance antitumor activity. Here, we report a GBM case with combination immunotherapy consisting of DC vaccines, anti-programmed death-1 (anti-PD-1) and poly I:C as well as the chemotherapeutic agent cyclophosphamide that was integrated with standard chemoradiation therapy, and the patient remained disease-free for 69 months. The patient received DC vaccines loaded with multiple forms of tumor antigens, including mRNA-tumor associated antigens (TAA), mRNA-neoantigens, and hypochlorous acid (HOCl)-oxidized tumor lysates. Furthermore, mRNA-TAAs were modified with a novel TriVac technology that fuses TAAs with a destabilization domain and inserts TAAs into full-length lysosomal associated membrane protein-1 to enhance major histocompatibility complex (MHC) class I and II antigen presentation. The treatment consisted of 42 DC cancer vaccine infusions, 26 anti-PD-1 antibody nivolumab administrations and 126 poly I:C injections for DC infusions. The patient also received 28 doses of cyclophosphamide for depletion of regulatory T cells. No immunotherapy-related adverse events were observed during the treatment. Robust antitumor CD4+ and CD8+ T-cell responses were detected. The patient remains free of disease progression. This is the first case report on the combination of the above three agents to treat glioblastoma patients. Our results suggest that integrated combination immunotherapy is safe and feasible for long-term treatment in this patient. A large-scale trial to validate these findings is warranted.
Collapse
Affiliation(s)
- Ping Zhu
- Department of Clinical Immunology, Xijing Hospital, Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Shi-You Li
- Beijing Tricision Biotherapeutics Inc., Beijing, 100176, China
| | - Jin Ding
- Department of Clinical Immunology, Xijing Hospital, Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Zhou Fei
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Sheng-Nan Sun
- Beijing Tricision Biotherapeutics Inc., Beijing, 100176, China
| | - Zhao-Hui Zheng
- Department of Clinical Immunology, Xijing Hospital, Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Ding Wei
- Department of Clinical Immunology, Xijing Hospital, Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Jun Jiang
- Zhuhai Tricision Biotherapuetics Inc., Zhuhai, Guangdong, 519040, China
| | - Jin-Lin Miao
- Department of Clinical Immunology, Xijing Hospital, Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - San-Zhong Li
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Xing Luo
- Department of Clinical Immunology, Xijing Hospital, Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Kui Zhang
- Department of Clinical Immunology, Xijing Hospital, Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Bin Wang
- Department of Clinical Immunology, Xijing Hospital, Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Kun Zhang
- Department of Clinical Immunology, Xijing Hospital, Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Su Pu
- Beijing Tricision Biotherapeutics Inc., Beijing, 100176, China
| | - Qian-Ting Wang
- Beijing Tricision Biotherapeutics Inc., Beijing, 100176, China
| | - Xin-Yue Zhang
- Zhuhai Tricision Biotherapuetics Inc., Zhuhai, Guangdong, 519040, China
| | - Gao-Liu Wen
- Zhuhai Tricision Biotherapuetics Inc., Zhuhai, Guangdong, 519040, China
| | - Jun O. Liu
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - John Thomas August
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Huijie Bian
- Department of Clinical Immunology, Xijing Hospital, Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Zhi-Nan Chen
- Department of Clinical Immunology, Xijing Hospital, Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - You-Wen He
- Beijing Tricision Biotherapeutics Inc., Beijing, 100176, China
| |
Collapse
|
8
|
Kubo K, Takeda S, Uchida M, Maeda M, Endo N, Sugahara S, Suzuki H, Fukahori H. Lit-LAMP-DNA-vaccine for shrimp allergy prevents anaphylactic symptoms in a murine model. Int Immunopharmacol 2022; 113:109394. [DOI: 10.1016/j.intimp.2022.109394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/17/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
|
9
|
LAMP-1 Chimeric to HIV-1 p55Gag in the Immunization of Neonate Mice Induces an Early Germinal Center Formation and AID Expression. Vaccines (Basel) 2022; 10:vaccines10081246. [PMID: 36016134 PMCID: PMC9414238 DOI: 10.3390/vaccines10081246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 11/29/2022] Open
Abstract
Neonates have a limited adaptive response of plasma cells, germinal center (GC) B cells, and T follicular helper cells (TFH). As neonatal vaccination can be an important tool for AIDS prevention, these limitations need to be overcome. Chimeric DNA vaccine encoding p55Gag HIV-1 protein conjugated with lysosomal-associated membrane protein 1 (LAMP-1) has been described as immunogenic in the neonate period. Herein, we investigated the immunologic mechanisms involved in neonatal immunization with a LAMP-1/p55Gag (LAMP/Gag) DNA vaccine in a C57BL/6 mouse background. Neonatal LAMP/Gag vaccination induced strong Gag-specific T-cell response until adulthood and elevated levels of anti-Gag IgG antibodies. We also demonstrated for the first time that the immunogenicity of the neonatal period with LAMP/Gag is due to the induction of high-affinity anti-p24 IgG antibodies and long-term plasma cells. Together with that, there is the generation of early TFH cells and the formation of GC sites with the upregulation of activation-induced cytidine deaminase (AID) enzyme mRNA and protein expression in draining lymph nodes after neonatal LAMP/Gag vaccination. These findings underscore that the LAMP-1 strategy in the chimeric vaccine could be useful to enhance antibody production even in the face of neonatal immaturity, and they contribute to the development of new vaccine approaches for other emerging pathogens at an early stage of life.
Collapse
|
10
|
Adhikari AS, Macauley J, Johnson Y, Connolly M, Coleman T, Heiland T. Development and Characterization of an HCMV Multi-Antigen Therapeutic Vaccine for Glioblastoma Using the UNITE Platform. Front Oncol 2022; 12:850546. [PMID: 35651802 PMCID: PMC9149224 DOI: 10.3389/fonc.2022.850546] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 03/18/2022] [Indexed: 01/10/2023] Open
Abstract
Glioblastoma multiforme (GBM) is an aggressive form of brain cancer with a median survival of 15 months that has remained unchanged despite advances in the standard of care. GBM cells express human cytomegalovirus (HCMV) proteins, providing a unique opportunity for targeted therapy. We utilized our UNITE (UNiversal Intracellular Targeted Expression) platform to develop a multi-antigen DNA vaccine (ITI-1001) that codes for the HCMV proteins pp65, gB, and IE-1. The UNITE platform involves lysosomal targeting technology, fusing lysosome-associated membrane protein 1 (LAMP1) with target ntigens. We demonstrate evidence of increased antigen presentation by both MHC-I and -II, delivering a robust antigen-specific CD4 and CD8 T-cell response in addition to a strong humoral response. Using a syngeneic orthotopic GBM mouse model, therapeutic treatment with the ITI-1001 vaccine resulted in ~56% survival of tumor-bearing mice. Investigation of the tumor microenvironment showed significant CD4 infiltration as well as enhanced Th1 and cytotoxic CD8 T activation. Regulatory T cells were also upregulated after ITI-1001 vaccination. In addition, tumor burden negatively correlated with activated interferon (IFN)γ+ CD4 T cells, reiterating the importance of CD4 activation in ITI-1001 efficacy and in identifying treatment responders and non-responders. Further characterization of these two groups showed high infiltration of CD3+, CD4+, and CD8+ T cells in responders compared to non-responders. Thus, we show that vaccination with HCMV antigens using the ITI-1001-UNITE platform generates strong cellular and humoral immune responses, triggering significant antitumor activity, leading to enhanced survival in a mouse model of GBM.
Collapse
Affiliation(s)
| | | | | | - Mike Connolly
- Immunomic Therapeutics, Rockville, MD, United States
| | | | - Teri Heiland
- Immunomic Therapeutics, Rockville, MD, United States
| |
Collapse
|
11
|
Jia X, Shao Q, Chaudhry AR, Kinlock BL, Izban MG, Zhang HY, Villalta F, Hildreth JEK, Liu B. Medroxyprogesterone Acetate (MPA) Enhances HIV-1 Accumulation and Release in Primary Cervical Epithelial Cells by Inhibiting Lysosomal Activity. Pathogens 2021; 10:1192. [PMID: 34578224 PMCID: PMC8465616 DOI: 10.3390/pathogens10091192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/08/2021] [Accepted: 09/10/2021] [Indexed: 12/30/2022] Open
Abstract
Medroxyprogesterone acetate (MPA) is one of the most widely used contraceptives in the world. Epidemiologic studies have uncovered a possible link between the use of MPA and an increased risk of HIV-1 transmission. However, the understanding of the mechanism is still limited. Our previous publication demonstrated that the lysosomal activity in human vaginal epithelial cells attenuated the trafficking of viral particles during HIV-1 transcytosis. In this study, we show that treating human primary cervical epithelial cells with MPA led to a reduction in lysosomal activity. This reduction caused an increase in the intracellular HIV-1 accumulation and, consequently, an increase in viral release. Our study uncovers a novel mechanism by which MPA enhances HIV-1 release in primary cervical epithelial cells, thus providing vital information for HIV intervention and prevention.
Collapse
Affiliation(s)
- Xiangxu Jia
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, TN 37208, USA; (X.J.); (Q.S.); (B.L.K.); (F.V.); (J.E.K.H.)
| | - Qiujia Shao
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, TN 37208, USA; (X.J.); (Q.S.); (B.L.K.); (F.V.); (J.E.K.H.)
| | - Ahsen R. Chaudhry
- Department of Obstetrics and Gynecology, Meharry Medical College, Nashville, TN 37208, USA;
| | - Ballington L. Kinlock
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, TN 37208, USA; (X.J.); (Q.S.); (B.L.K.); (F.V.); (J.E.K.H.)
| | - Michael G. Izban
- Department of Pathology, Meharry Medical College, Nashville, TN 37208, USA;
| | - Hong-Ying Zhang
- Nanjing Municipal Center of Disease Control and Prevention, Nanjing 210003, China;
| | - Fernando Villalta
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, TN 37208, USA; (X.J.); (Q.S.); (B.L.K.); (F.V.); (J.E.K.H.)
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA
| | - James E. K. Hildreth
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, TN 37208, USA; (X.J.); (Q.S.); (B.L.K.); (F.V.); (J.E.K.H.)
| | - Bindong Liu
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, TN 37208, USA; (X.J.); (Q.S.); (B.L.K.); (F.V.); (J.E.K.H.)
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA
| |
Collapse
|
12
|
Petrova SY, Khlgatian SV, Svirshchevskaya EV, Vasilyeva AV, Berzhets VM. DNA vaccines and recombinant allergens with reduced allergenic activity treat allergies. RUSSIAN OPEN MEDICAL JOURNAL 2021. [DOI: 10.15275/rusomj.2021.0102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
This review is intended to familiarize readers with major novel directions of developing allergy vaccines, their structure, as well as the mechanisms of forming a new immunological response in the course of the treating immunoglobulin E (IgE)-mediated allergic diseases. Currently, science offers a huge variety of new experimental forms of recombinant allergens with reduced allergenic activity and increased immunogenicity, or vice-versa, immune tolerance. Often, the mechanisms of their effect on the immune system are not fully understood. Scientific publications, including reviews covering this topic, allowed us identifying top priority areas in the development of allergy vaccines: recombinant hypoallergenic allergen derivatives, T cell epitope-based allergy vaccines, and B cell epitope-based allergy vaccines. In addition, the review discusses use of deoxyribonucleic acid (DNA) vaccines. Immunotherapy with DNA vaccines is the newest and least studied method of treating allergic diseases.
Collapse
Affiliation(s)
| | | | - Elena V. Svirshchevskaya
- M.M. Shemyakin – Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences
| | | | | |
Collapse
|
13
|
Lu L, Jiang J, Zhan M, Zhang H, Wang QT, Sun SN, Guo XK, Yin H, Wei Y, Li SY, Liu JO, Li Y, He YW. Targeting Tumor-Associated Antigens in Hepatocellular Carcinoma for Immunotherapy: Past Pitfalls and Future Strategies. Hepatology 2021; 73:821-832. [PMID: 32767586 DOI: 10.1002/hep.31502] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/23/2020] [Accepted: 07/02/2020] [Indexed: 12/15/2022]
Affiliation(s)
- Ligong Lu
- Zhuhai Interventional Medical CenterZhuhai Precision Medical CenterZhuhai People's HospitalZhuhai Hospital Affiliated with Jinan UniversityZhuhaiGuangdong ProvinceP.R. China
| | - Jun Jiang
- Tricision Biotherapeutic Inc. Jinwan DistrictZhuhaiChina
| | - Meixiao Zhan
- Zhuhai Interventional Medical CenterZhuhai Precision Medical CenterZhuhai People's HospitalZhuhai Hospital Affiliated with Jinan UniversityZhuhaiGuangdong ProvinceP.R. China
| | - Hui Zhang
- First Affiliated HospitalChina Medical UniversityShenyangChina
| | - Qian-Ting Wang
- Tricision Biotherapeutic Inc. Jinwan DistrictZhuhaiChina
| | - Sheng-Nan Sun
- Tricision Biotherapeutic Inc. Jinwan DistrictZhuhaiChina
| | - Xiao-Kai Guo
- Tricision Biotherapeutic Inc. Jinwan DistrictZhuhaiChina
| | - Hua Yin
- Zhuhai Interventional Medical CenterZhuhai Precision Medical CenterZhuhai People's HospitalZhuhai Hospital Affiliated with Jinan UniversityZhuhaiGuangdong ProvinceP.R. China
| | - Yadong Wei
- Department of Pharmacology and Molecular SciencesJohns Hopkins University School of MedicineBaltimoreMD
| | - Shi-You Li
- Tricision Biotherapeutic Inc. Jinwan DistrictZhuhaiChina
| | - Jun O Liu
- Department of Pharmacology and Molecular SciencesJohns Hopkins University School of MedicineBaltimoreMD
| | - Yong Li
- Zhuhai Interventional Medical CenterZhuhai Precision Medical CenterZhuhai People's HospitalZhuhai Hospital Affiliated with Jinan UniversityZhuhaiGuangdong ProvinceP.R. China
| | - You-Wen He
- Department of ImmunologyDuke University Medical University Medical CenterDurhamNC
| |
Collapse
|
14
|
Castanha PMS, Marques ETA. A Glimmer of Hope: Recent Updates and Future Challenges in Zika Vaccine Development. Viruses 2020; 12:E1371. [PMID: 33266129 PMCID: PMC7761420 DOI: 10.3390/v12121371] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/19/2020] [Accepted: 11/26/2020] [Indexed: 02/07/2023] Open
Abstract
The emergence and rapid spread of Zika virus (ZIKV) on a global scale as well as the establishment of a causal link between Zika infection and congenital syndrome and neurological disorders triggered unprecedented efforts towards the development of a safe and effective Zika vaccine. Multiple vaccine platforms, including purified inactivated virus, nucleic acid vaccines, live-attenuated vaccines, and viral-vectored vaccines, have advanced to human clinical trials. In this review, we discuss the recent advances in the field of Zika vaccine development and the challenges for future clinical efficacy trials. We provide a brief overview on Zika vaccine platforms in the pipeline before summarizing the vaccine candidates in clinical trials, with a focus on recent, promising results from vaccine candidates that completed phase I trials. Despite low levels of transmission during recent years, ZIKV has become endemic in the Americas and the potential of large Zika outbreaks remains real. It is important for vaccine developers to continue developing their Zika vaccines, so that a potential vaccine is ready for deployment and clinical efficacy trials when the next ZIKV outbreak occurs.
Collapse
Affiliation(s)
| | - Ernesto T. A. Marques
- Graduate School of Public Health, Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, PA 15261, USA;
| |
Collapse
|
15
|
Chen AC, Xu R, Wang T, Wei J, Yang XY, Liu CX, Lei G, Lyerly HK, Heiland T, Hartman ZC. HER2-LAMP vaccines effectively traffic to endolysosomal compartments and generate enhanced polyfunctional T cell responses that induce complete tumor regression. J Immunother Cancer 2020; 8:jitc-2019-000258. [PMID: 32532838 PMCID: PMC7295440 DOI: 10.1136/jitc-2019-000258] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/01/2020] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND The advent of immune checkpoint blockade antibodies has demonstrated that effective mobilization of T cell responses can cause tumor regression of metastatic cancers, although these responses are heterogeneous and restricted to certain histologic types of cancer. To enhance these responses, there has been renewed emphasis in developing effective cancer-specific vaccines to stimulate and direct T cell immunity to important oncologic targets, such as the oncogene human epidermal growth factor receptor 2 (HER2), expressed in ~20% of breast cancers (BCs). METHODS In our study, we explored the use of alternative antigen trafficking through use of a lysosome-associated membrane protein 1 (LAMP) domain to enhance vaccine efficacy against HER2 and other model antigens in both in vitro and in vivo studies. RESULTS We found that inclusion of this domain in plasmid vaccines effectively trafficked antigens to endolysosomal compartments, resulting in enhanced major histocompatibility complex (MHC) class I and II presentation. Additionally, this augmented the expansion/activation of antigen-specific CD4+ and CD8+ T cells and also led to elevated levels of antigen-specific polyfunctional CD8+ T cells. Significantly, vaccination with HER2-LAMP produced tumor regression in ~30% of vaccinated mice with established tumors in an endogenous model of metastatic HER2+ BC, compared with 0% of HER2-WT vaccinated mice. This therapeutic benefit is associated with enhanced tumor infiltration of activated CD4+ and CD8+ T cells. CONCLUSIONS These data demonstrate the potential of using LAMP-based endolysosomal trafficking as a means to augment the generation of polyfunctional, antigen-specific T cells in order to improve antitumor therapeutic responses using cancer antigen vaccines.
Collapse
Affiliation(s)
- Alan Chen Chen
- Department of Surgery, Duke University, Duke University, Durham, North Carolina, USA
| | - Renhuan Xu
- Department of R&D, Immunomic Therapeutics, Rockville, Maryland, USA
| | - Tao Wang
- Department of Surgery, Duke University, Duke University, Durham, North Carolina, USA
| | - Junping Wei
- Department of Surgery, Duke University, Duke University, Durham, North Carolina, USA
| | - Xiao-Yi Yang
- Department of Surgery, Duke University, Duke University, Durham, North Carolina, USA
| | - Cong-Xiao Liu
- Department of Surgery, Duke University, Duke University, Durham, North Carolina, USA
| | - Gangjun Lei
- Department of Surgery, Duke University, Duke University, Durham, North Carolina, USA
| | - Herbert Kim Lyerly
- Department of Surgery, Duke University, Duke University, Durham, North Carolina, USA.,Department of Pathology, Duke University, Durham, North Carolina, USA.,Department of Immunology, Duke University, Durham, NC, USA
| | - Teri Heiland
- Department of R&D, Immunomic Therapeutics, Rockville, Maryland, USA
| | - Zachary Conrad Hartman
- Department of Surgery, Duke University, Duke University, Durham, North Carolina, USA .,Department of Pathology, Duke University, Durham, North Carolina, USA
| |
Collapse
|
16
|
Krotova K, Day A, Aslanidi G. An Engineered AAV6-Based Vaccine Induces High Cytolytic Anti-Tumor Activity by Directly Targeting DCs and Improves Ag Presentation. Mol Ther Oncolytics 2019; 15:166-177. [PMID: 31720373 PMCID: PMC6838889 DOI: 10.1016/j.omto.2019.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 10/01/2019] [Indexed: 12/30/2022] Open
Abstract
We have previously shown that an AAV6-based vaccine generates high levels of antigen-specific CD8+ T cells. Further modifications described here led to significantly increased levels of antigen-specific CD8+ and CD4+ T cells, enhanced formation of memory cells, and superior antigen-specific killing capacity in a murine model. By tracking reporter-gene-positive dendritic cells, we showed that they were directly targeted with modified AAV6 in vivo. Our vaccine's anti-cancer potential was evaluated with the antigen ovalbumin against a B16F10 melanoma cell line stably expressing ovalbumin. The vaccination showed superior protection in a murine model of metastatic melanoma. The vaccination significantly delayed solid tumor growth but did not completely prevent tumor development. We show that tumors in immunized mice escaped vaccine-induced killing by losing ovalbumin expression. The vaccine induced massive tumor infiltration with NK and CD8+ T cells with upregulated PD-1 expression. Thus, a vaccination of a combination of anti-PD-1 antibodies demonstrated significant improvement in the treatment efficacy. To summarize, we showed that a bioengineered AAV6-based vaccine elicits strong and long-lasting cellular and humoral responses against an encoded antigen. To increase AAV vaccine efficiency and mitigate tumor escape through antigen loss, we intended to target several antigens in combination with treatments targeting the tumor microenvironment.
Collapse
Affiliation(s)
- Karina Krotova
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Andrew Day
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - George Aslanidi
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| |
Collapse
|
17
|
Jiang DB, Zhang JP, Cheng LF, Zhang GW, Li Y, Li ZC, Lu ZH, Zhang ZX, Lu YC, Zheng LH, Zhang FL, Yang K. Hantavirus Gc induces long-term immune protection via LAMP-targeting DNA vaccine strategy. Antiviral Res 2018; 150:174-182. [PMID: 29273568 DOI: 10.1016/j.antiviral.2017.12.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 12/14/2017] [Accepted: 12/16/2017] [Indexed: 01/22/2023]
Abstract
Hemorrhagic fever with renal syndrome (HFRS) occurs widely throughout Eurasia. Unfortunately, there is no effective treatment, and prophylaxis remains the best option against the major pathogenic agent, hantaan virus (HTNV), which is an Old World hantavirus. However, the absence of cellular immune responses and immunological memory hampers acceptance of the current inactivated HFRS vaccine. Previous studies revealed that a lysosome-associated membrane protein 1 (LAMP1)-targeting strategy involving a DNA vaccine based on the HTNV glycoprotein Gn successfully conferred long-term immunity, and indicated that further research on Gc, another HTNV antigen, was warranted. Plasmids encoding Gc and lysosome-targeted Gc, designated pVAX-Gc and pVAX-LAMP/Gc, respectively, were constructed. Proteins of interest were identified by fluorescence microscopy following cell line transfection. Five groups of 20 female BALB/c mice were subjected to the following inoculations: inactivated HTNV vaccine, pVAX-LAMP/Gc, pVAX-Gc, and, as the negative controls, pVAX-LAMP or the blank vector pVAX1. Humoral and cellular immunity were assessed by enzyme-linked immunosorbent assays (ELISAs) and 15-mer peptide enzyme-linked immunospot (ELISpot) epitope mapping assays. Repeated immunization with pVAX-LAMP/Gc enhanced adaptive immune responses, as demonstrated by the specific and neutralizing antibody titers and increased IFN-γ production. The inactivated vaccine induced a comparable humoral reaction, but the negative controls only elicited insignificant responses. Using a mouse model of HTNV challenge, the in vivo protection conferred by the inactivated vaccine and Gc-based constructs (with/without LAMP recombination) was confirmed. Evidence of pan-epitope reactions highlighted the long-term cellular response to the LAMP-targeting strategy, and histological observations indicated the safety of the LAMP-targeting vaccines. The long-term protective immune responses induced by pVAX-LAMP/Gc may be due to the advantage afforded by lysosomal targeting after exogenous antigen processing initiation and major histocompatibility complex (MHC) class II antigen presentation trafficking. MHC II-restricted antigen recognition effectively primes HTNV-specific CD4+ T-cells, leading to the promotion of significant immune responses and immunological memory. An epitope-spreading phenomenon was observed, which mirrors the previous result from the Gn study, in which the dominant IFN-γ-responsive hot-spot epitopes were shared between HLA-II and H2d. Importantly, the pan-epitope reaction to Gc indicated that Gc should be with potential for use in further hantavirus DNA vaccine investigations.
Collapse
Affiliation(s)
- Dong-Bo Jiang
- Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Jin-Peng Zhang
- Department of Immunology, Fourth Military Medical University, Xi'an, China; Brigade of Cadet, Fourth Military Medical University, Xi'an, China
| | - Lin-Feng Cheng
- Department of Microbiology, Fourth Military Medical University, Xi'an, China
| | - Guan-Wen Zhang
- Department of Immunology, Fourth Military Medical University, Xi'an, China; Brigade of Cadet, Fourth Military Medical University, Xi'an, China
| | - Yun Li
- Department of Immunology, Fourth Military Medical University, Xi'an, China; Brigade of Cadet, Fourth Military Medical University, Xi'an, China
| | - Zi-Chao Li
- Department of Immunology, Fourth Military Medical University, Xi'an, China; Brigade of Cadet, Fourth Military Medical University, Xi'an, China
| | - Zhen-Hua Lu
- Department of Immunology, Fourth Military Medical University, Xi'an, China; Brigade of Cadet, Fourth Military Medical University, Xi'an, China
| | - Zi-Xin Zhang
- Department of Immunology, Fourth Military Medical University, Xi'an, China; Brigade of Cadet, Fourth Military Medical University, Xi'an, China
| | - Yu-Chen Lu
- Department of Immunology, Fourth Military Medical University, Xi'an, China; Brigade of Cadet, Fourth Military Medical University, Xi'an, China
| | - Lian-He Zheng
- Department of Orthopedics, Tangdu Hospital, Xi'an, China.
| | - Fang-Lin Zhang
- Department of Microbiology, Fourth Military Medical University, Xi'an, China.
| | - Kun Yang
- Department of Immunology, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
18
|
Su Y, Romeu-Bonilla E, Anagnostou A, Fitz-Patrick D, Hearl W, Heiland T. Safety and long-term immunological effects of CryJ2-LAMP plasmid vaccine in Japanese red cedar atopic subjects: A phase I study. Hum Vaccin Immunother 2017; 13:2804-2813. [PMID: 28605294 PMCID: PMC5718801 DOI: 10.1080/21645515.2017.1329070] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Japanese Red Cedar (JRC) pollen induced allergy affects one third of Japanese and the development of effective therapies remains an unachieved challenge. We designed a DNA vaccine encoding CryJ2 allergen from the JRC pollen and Lysosomal Associated Membrane Protein 1 (LAMP-1) to treat JRC allergy. These Phase IA and IB trials assessed safety and immunological effects of the investigational CryJ2-LAMP DNA vaccine in both non-sensitive and sensitive Japanese expatriates living in Honolulu, Hawaii. In the Phase IA trial, 6 JRC non-sensitive subjects and 9 JRC and/or Mountain Cedar (MC) sensitive subjects were given 4 vaccine doses (each 4mg/1ml) intramuscularly (IM) at 14-day intervals. Nine JRC and/or MC sensitive subjects were given 4 doses (2 mg/0.5 ml) IM at 14-day intervals. The safety and functional biomarkers were followed for 132 d. Following this, 17 of 24 subjects were recruited into the IB trial and received one booster dose (2 mg/0.5 ml) IM approximately 300 d after the first vaccination dose to which they were randomized in the first phase of the trial. All safety endpoints were met and all subjects tolerated CryJ2-LAMP vaccinations well. At the end of the IA trial, 10 out of 12 JRC sensitive and 6 out of 11 MC sensitive subjects experienced skin test negative conversion, possibly related to the CryJ2-LAMP vaccinations. Collectively, these data suggested that the CryJ2-LAMP DNA vaccine is safe and may be immunologically effective in treating JRC induced allergy.
Collapse
Affiliation(s)
- Yan Su
- a Immunomic Therapeutics, Inc. (ITI) , Rockville , MD , USA
| | | | | | | | - William Hearl
- a Immunomic Therapeutics, Inc. (ITI) , Rockville , MD , USA
| | - Teri Heiland
- a Immunomic Therapeutics, Inc. (ITI) , Rockville , MD , USA
| |
Collapse
|
19
|
Hou J, Zhang Q, Liu Z, Wang S, Li D, Liu C, Liu Y, Shao Y. Cyclophilin A as a potential genetic adjuvant to improve HIV-1 Gag DNA vaccine immunogenicity by eliciting broad and long-term Gag-specific cellular immunity in mice. Hum Vaccin Immunother 2016; 12:545-53. [PMID: 26305669 DOI: 10.1080/21645515.2015.1082692] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Previous research has shown that host Cyclophilin A (CyPA) can promote dendritic cell maturation and the subsequent innate immune response when incorporated into an HIV-1 Gag protein to circumvent the resistance of dendritic cells to HIV-1 infection. This led us to hypothesize that CyPA may improve HIV-1 Gag-specific vaccine immunogenicity via binding with Gag antigen. The adjuvant effect of CyPA was evaluated using a DNA vaccine with single or dual expression cassettes. Mouse studies indicated that CyPA specifically and markedly promoted HIV-1 Gag-specific cellular immunity but not an HIV-1 Env-specific cellular response. The Gag/CyPA dual expression cassettes stimulated a greater Gag-specific cellular immune response, than Gag immunization alone. Furthermore, CyPA induced a broad Gag-specific T cell response and strong cellular immunity that lasted up to 5 months. In addition, CyPA skewed to cellular rather than humoral immunity. To investigate the mechanisms of the adjuvant effect, site-directed mutagenesis in CyPA, including active site residues H54Q and F60A resulted in mutants that were co-expressed with Gag in dual cassettes. The immune response to this vaccine was analyzed in vivo. Interestingly, the wild type CyPA markedly increased Gag cellular immunity, but the H54Q and F60A mutants drastically reduced CyPA adjuvant activation. Therefore, we suggest that the adjuvant effect of CyPA was based on Gag-CyPA-specific interactions. Herein, we report that Cyclophilin A can augment HIV-1 Gag-specific cellular immunity as a genetic adjuvant in multiplex DNA immunization strategies, and that activity of this adjuvant is specific, broad, long-term, and based on Gag-CyPA interaction.
Collapse
Affiliation(s)
- Jue Hou
- a State Key Laboratory for Infectious Disease Prevention and Control; National Center for AIDS/STD Control and Prevention; Chinese Center for Disease Control and Prevention ; Beijing , China
| | - Qicheng Zhang
- a State Key Laboratory for Infectious Disease Prevention and Control; National Center for AIDS/STD Control and Prevention; Chinese Center for Disease Control and Prevention ; Beijing , China
| | - Zheng Liu
- a State Key Laboratory for Infectious Disease Prevention and Control; National Center for AIDS/STD Control and Prevention; Chinese Center for Disease Control and Prevention ; Beijing , China
| | - Shuhui Wang
- a State Key Laboratory for Infectious Disease Prevention and Control; National Center for AIDS/STD Control and Prevention; Chinese Center for Disease Control and Prevention ; Beijing , China
| | - Dan Li
- a State Key Laboratory for Infectious Disease Prevention and Control; National Center for AIDS/STD Control and Prevention; Chinese Center for Disease Control and Prevention ; Beijing , China
| | - Chang Liu
- a State Key Laboratory for Infectious Disease Prevention and Control; National Center for AIDS/STD Control and Prevention; Chinese Center for Disease Control and Prevention ; Beijing , China
| | - Ying Liu
- a State Key Laboratory for Infectious Disease Prevention and Control; National Center for AIDS/STD Control and Prevention; Chinese Center for Disease Control and Prevention ; Beijing , China
| | - Yiming Shao
- a State Key Laboratory for Infectious Disease Prevention and Control; National Center for AIDS/STD Control and Prevention; Chinese Center for Disease Control and Prevention ; Beijing , China
| |
Collapse
|
20
|
Lucas CGDO, Matassoli FL, Peçanha LMT, Santillo BT, Oliveira LMDS, Oshiro TM, Marques ETDA, Oxenius A, de Arruda LB. Dendritic cells primed with a chimeric plasmid containing HIV-1-gag associated with lysosomal-associated protein-1 (LAMP/gag) is a potential therapeutic vaccine against HIV. FASEB J 2016; 30:2970-84. [PMID: 27199296 DOI: 10.1096/fj.201500059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 05/02/2016] [Indexed: 11/11/2022]
Abstract
The decline in number and function of T cells is a hallmark of HIV infection, and preservation or restoration of HIV-specific cellular immune response is a major goal of AIDS treatment. Dendritic cells (DCs) play a key role in the initiation and maintenance of the immune response, and their use as a vaccine vehicle is a promising strategy for enhancing vaccine efficacy. We evaluated the potential of DC-mediated immunization with a DNA vaccine consisting of HIV-1-p55gag (gag, group-specific antigen) associated to lysosomal associated protein (LAMP) sequence (LAMP/gag vaccine). Immunization of mice with mouse DCs transfected with LAMP/gag (Lg-mDCs) stimulated more potent B- and T-cell responses than naked DNA or DCs pulsed with inactivated HIV. Anti-Gag antibody levels were sustained for at least 3 mo after immunization, and recall T-cell responses were also strongly detected at this time point. Human DCs transfected with LAMP/gag (Lg-hDCs) were also activated and able to stimulate greater T-cell response than native gag-transfected DCs. Coculture between Lg-hDCs and T lymphocytes obtained from patients with HIV resulted in upregulation of CD38, CD69, HLA-DR, and granzyme B by CD4(+) and CD8(+) T cells, and increased IFN-γ and TNF-α production. These results indicate that the use of LAMP/gag-DC may be an efficient strategy for enhancing immune function in patients with HIV.-Lucas, C. G. D. O., Matassoli, F. L., Peçanha, L. M. T., Santillo, B. T., Oliveira, L. M. D. S., Oshiro, T. M., Marques, E. T. D. A., Jr., Oxenius, A., de Arruda, L. B. Dendritic cells primed with a chimeric plasmid containing HIV-1-gag associated with lysosomal-associated protein-1 (LAMP/gag) is a potential therapeutic vaccine against HIV.
Collapse
Affiliation(s)
- Carolina G D O Lucas
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Flavio L Matassoli
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ligia M T Peçanha
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Bruna Tereso Santillo
- Laboratório de Dermatologia e Imunodeficiências (LIM-56), Departamento de Dermatologia, Escola de Medicina, Universidade de São Paulo, São Paulo, Brazil; Institute of Microbiology, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Luanda Mara da Silva Oliveira
- Laboratório de Dermatologia e Imunodeficiências (LIM-56), Departamento de Dermatologia, Escola de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Telma Miyuki Oshiro
- Laboratório de Dermatologia e Imunodeficiências (LIM-56), Departamento de Dermatologia, Escola de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Ernesto T D A Marques
- Department of Infectious Diseases and Microbiology, Center for Vaccine Research, Pittsburgh, Pennsylvania, USA; and Department of Virology, Centro de Pesquisas Aggeu Magalhães (CPqAM), Fundação Oswaldo Cruz (Fiocruz)-Pernambuco, Recife, Brazil
| | - Annette Oxenius
- Institute of Microbiology, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Luciana B de Arruda
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil;
| |
Collapse
|
21
|
Su Y, Connolly M, Marketon A, Heiland T. CryJ-LAMP DNA Vaccines for Japanese Red Cedar Allergy Induce Robust Th1-Type Immune Responses in Murine Model. J Immunol Res 2016; 2016:4857869. [PMID: 27239481 PMCID: PMC4867073 DOI: 10.1155/2016/4857869] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 03/23/2016] [Accepted: 03/27/2016] [Indexed: 12/29/2022] Open
Abstract
Allergies caused by Japanese Red Cedar (JRC) pollen affect up to a third of Japanese people, necessitating development of an effective therapeutic. We utilized the lysosomal targeting property of lysosomal-associated membrane protein-1 (LAMP-1) to make DNA vaccines that encode LAMP-1 and the sequences of immunodominant allergen CryJ1 or CryJ2 from the JRC pollen. This novel strategy is designed to skew the CD4 T cell responses to the target allergens towards a nonallergenic Th1 response. CryJ1-LAMP and CryJ2-LAMP were administrated to BALB/c mice and antigen-specific Th1-type IgG2a and Th2-type IgG1 antibodies, as well as IgE antibodies, were assayed longitudinally. We also isolated different T cell populations from immunized mice and adoptively transferred them into naïve mice followed by CryJ1/CryJ2 protein boosts. We demonstrated that CryJ-LAMP immunized mice produce high levels of IFN-γ and anti-CryJ1 or anti-CryJ2 IgG2a antibodies and low levels of IgE antibodies, suggesting that a Th1 response was induced. In addition, we found that CD4(+) T cells are the immunological effectors of DNA vaccination in this allergy model. Together, our results suggest the CryJ-LAMP Vaccine has a potential as an effective therapeutic for JRC induced allergy by skewing Th1/Th2 responses.
Collapse
Affiliation(s)
- Yan Su
- Department of R&D, Immunomic Therapeutics, Inc. (ITI), Rockville, MD 20850, USA
| | - Michael Connolly
- Department of R&D, Immunomic Therapeutics, Inc. (ITI), Rockville, MD 20850, USA
| | - Anthony Marketon
- Department of R&D, Immunomic Therapeutics, Inc. (ITI), Rockville, MD 20850, USA
| | - Teri Heiland
- Department of R&D, Immunomic Therapeutics, Inc. (ITI), Rockville, MD 20850, USA
| |
Collapse
|
22
|
Gandhi RT, Kwon DS, Macklin EA, Shopis JR, McLean AP, McBrine N, Flynn T, Peter L, Sbrolla A, Kaufmann DE, Porichis F, Walker BD, Bhardwaj N, Barouch DH, Kavanagh DG. Immunization of HIV-1-Infected Persons With Autologous Dendritic Cells Transfected With mRNA Encoding HIV-1 Gag and Nef: Results of a Randomized, Placebo-Controlled Clinical Trial. J Acquir Immune Defic Syndr 2016; 71:246-53. [PMID: 26379068 PMCID: PMC4752409 DOI: 10.1097/qai.0000000000000852] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Accepted: 08/31/2015] [Indexed: 11/26/2022]
Abstract
BACKGROUND HIV-1 eradication may require reactivation of latent virus along with stimulation of HIV-1-specific immune responses to clear infected cells. Immunization with autologous dendritic cells (DCs) transfected with viral mRNA is a promising strategy for eliciting HIV-1-specific immune responses. We performed a randomized controlled clinical trial to evaluate the immunogenicity of this approach in HIV-1-infected persons on antiretroviral therapy. METHODS Fifteen participants were randomized 2:1 to receive intradermal immunization with HIV-1 Gag- and Nef-transfected DCs (vaccine) or mock-transfected DCs (placebo) at weeks 0, 2, 6, and 10. All participants also received DCs pulsed with keyhole limpet hemocyanin (KLH) to assess whether responses to a neo-antigen could be induced. RESULTS After immunization, there were no differences in interferon-gamma enzyme-linked immunospot responses to HIV-1 Gag or Nef in the vaccine or placebo group. CD4 proliferative responses to KLH increased 2.4-fold (P = 0.026) and CD8 proliferative responses to KLH increased 2.5-fold (P = 0.053) after vaccination. There were increases in CD4 proliferative responses to HIV-1 Gag (2.5-fold vs. baseline, 3.4-fold vs. placebo, P = 0.054) and HIV-1 Nef (2.3-fold vs. baseline, 6.3-fold vs. placebo, P = 0.009) among vaccine recipients, but these responses were short-lived. CONCLUSION Immunization with DCs transfected with mRNA encoding HIV-1 Gag and Nef did not induce significant interferon-gamma enzyme-linked immunospot responses. There were increases in proliferative responses to HIV-1 antigens and to a neo-antigen, KLH, but the effects were transient. Dendritic cell vaccination should be optimized to elicit stronger and long-lasting immune responses for this strategy to be effective as an HIV-1 therapeutic vaccine.
Collapse
Affiliation(s)
- Rajesh T. Gandhi
- Massachusetts General Hospital, Division of Infectious Diseases, Boston, MA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge, MA
| | - Douglas S. Kwon
- Massachusetts General Hospital, Division of Infectious Diseases, Boston, MA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge, MA
| | - Eric A. Macklin
- Biostatistics Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Janet R. Shopis
- Massachusetts General Hospital, Division of Infectious Diseases, Boston, MA
| | - Anna P. McLean
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge, MA
| | - Nicole McBrine
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge, MA
| | - Theresa Flynn
- Massachusetts General Hospital, Division of Infectious Diseases, Boston, MA
| | - Lauren Peter
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Amy Sbrolla
- Massachusetts General Hospital, Division of Infectious Diseases, Boston, MA
| | - Daniel E. Kaufmann
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge, MA
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and University of Montreal, Montréal, QC, Canada
| | - Filippos Porichis
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge, MA
| | - Bruce D. Walker
- Massachusetts General Hospital, Division of Infectious Diseases, Boston, MA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge, MA
- Howard Hughes Medical Institute, Chevy Chase, MD
| | - Nina Bhardwaj
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY; and
| | - Dan H. Barouch
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge, MA
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Daniel G. Kavanagh
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge, MA
| |
Collapse
|
23
|
Genomic Analysis, Phenotype, and Virulence of the Historical Brazilian Smallpox Vaccine Strain IOC: Implications for the Origins and Evolutionary Relationships of Vaccinia Virus. J Virol 2015; 89:11909-25. [PMID: 26378174 DOI: 10.1128/jvi.01833-15] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Accepted: 09/08/2015] [Indexed: 02/07/2023] Open
Abstract
UNLABELLED Smallpox was declared eradicated in 1980 after an intensive vaccination program using different strains of vaccinia virus (VACV; Poxviridae). VACV strain IOC (VACV-IOC) was the seed strain of the smallpox vaccine manufactured by the major vaccine producer in Brazil during the smallpox eradication program. However, little is known about the biological and immunological features as well as the phylogenetic relationships of this first-generation vaccine. In this work, we present a comprehensive characterization of two clones of VACV-IOC. Both clones had low virulence in infected mice and induced a protective immune response against a lethal infection comparable to the response of the licensed vaccine ACAM2000 and the parental strain VACV-IOC. Full-genome sequencing revealed the presence of several fragmented virulence genes that probably are nonfunctional, e.g., F1L, B13R, C10L, K3L, and C3L. Most notably, phylogenetic inference supported by the structural analysis of the genome ends provides evidence of a novel, independent cluster in VACV phylogeny formed by VACV-IOC, the Brazilian field strains Cantagalo (CTGV) and Serro 2 viruses, and horsepox virus, a VACV-like virus supposedly related to an ancestor of the VACV lineage. Our data strongly support the hypothesis that CTGV-like viruses represent feral VACV that evolved in parallel with VACV-IOC after splitting from a most recent common ancestor, probably an ancient smallpox vaccine strain related to horsepox virus. Our data, together with an interesting historical investigation, revisit the origins of VACV and propose new evolutionary relationships between ancient and extant VACV strains, mainly horsepox virus, VACV-IOC/CTGV-like viruses, and Dryvax strain. IMPORTANCE First-generation vaccines used to eradicate smallpox had rates of adverse effects that are not acceptable by current health care standards. Moreover, these vaccines are genetically heterogeneous and consist of a pool of quasispecies of VACV. Therefore, the search for new-generation smallpox vaccines that combine low pathogenicity, immune protection, and genetic homogeneity is extremely important. In addition, the phylogenetic relationships and origins of VACV strains are quite nebulous. We show the characterization of two clones of VACV-IOC, a unique smallpox vaccine strain that contributed to smallpox eradication in Brazil. The immunogenicity and reduced virulence make the IOC clones good options for alternative second-generation smallpox vaccines. More importantly, this study reveals the phylogenetic relationship between VACV-IOC, feral VACV established in nature, and the ancestor-like horsepox virus. Our data expand the discussion on the origins and evolutionary connections of VACV lineages.
Collapse
|
24
|
Maciel M, Cruz FDSP, Cordeiro MT, da Motta MA, Cassemiro KMSDM, Maia RDCC, de Figueiredo RCBQ, Galler R, Freire MDS, August JT, Marques ETA, Dhalia R. A DNA vaccine against yellow fever virus: development and evaluation. PLoS Negl Trop Dis 2015; 9:e0003693. [PMID: 25875109 PMCID: PMC4395287 DOI: 10.1371/journal.pntd.0003693] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 03/10/2015] [Indexed: 11/30/2022] Open
Abstract
Attenuated yellow fever (YF) virus 17D/17DD vaccines are the only available protection from YF infection, which remains a significant source of morbidity and mortality in the tropical areas of the world. The attenuated YF virus vaccine, which is used worldwide, generates both long-lasting neutralizing antibodies and strong T-cell responses. However, on rare occasions, this vaccine has toxic side effects that can be fatal. This study presents the design of two non-viral DNA-based antigen formulations and the characterization of their expression and immunological properties. The two antigen formulations consist of DNA encoding the full-length envelope protein (p/YFE) or the full-length envelope protein fused to the lysosomal-associated membrane protein signal, LAMP-1 (pL/YFE), aimed at diverting antigen processing/presentation through the major histocompatibility complex II precursor compartments. The immune responses triggered by these formulations were evaluated in H2b and H2d backgrounds, corresponding to the C57Bl/6 and BALB/c mice strains, respectively. Both DNA constructs were able to induce very strong T-cell responses of similar magnitude against almost all epitopes that are also generated by the YF 17DD vaccine. The pL/YFE formulation performed best overall. In addition to the T-cell response, it was also able to stimulate high titers of anti-YF neutralizing antibodies comparable to the levels elicited by the 17DD vaccine. More importantly, the pL/YFE vaccine conferred 100% protection against the YF virus in intracerebrally challenged mice. These results indicate that pL/YFE DNA is an excellent vaccine candidate and should be considered for further developmental studies.
Collapse
Affiliation(s)
- Milton Maciel
- Johns Hopkins University, School of Medicine, Department of Pharmacology & Molecular Sciences, Baltimore, Maryland, United States of America
| | - Fábia da Silva Pereira Cruz
- Oswaldo Cruz Foundation (FIOCRUZ), Aggeu Magalhães Research Centre, Department of Virology, Laboratório de Virologia e Terapia Experimental (LAVITE), Universidade Federal de Pernambuco (UFPE), University City, Recife, Pernambuco, Brazil
| | - Marli Tenório Cordeiro
- Oswaldo Cruz Foundation (FIOCRUZ), Aggeu Magalhães Research Centre, Department of Virology, Laboratório de Virologia e Terapia Experimental (LAVITE), Universidade Federal de Pernambuco (UFPE), University City, Recife, Pernambuco, Brazil
- Health Secretariat of the State of Pernambuco, Central Public Health Laboratory-LACEN, Boa Vista, Recife, Pernambuco, Brazil
| | - Márcia Archer da Motta
- Oswaldo Cruz Foundation (FIOCRUZ), Oswaldo Cruz Institute, Bio-Manguinhos, Laboratório de Tecnologia Virológica (LATEV), Manguinhos, Rio de Janeiro, Brazil
| | - Klécia Marília Soares de Melo Cassemiro
- Oswaldo Cruz Foundation (FIOCRUZ), Aggeu Magalhães Research Centre, Department of Virology, Laboratório de Virologia e Terapia Experimental (LAVITE), Universidade Federal de Pernambuco (UFPE), University City, Recife, Pernambuco, Brazil
| | - Rita de Cássia Carvalho Maia
- Federal Rural University of Pernambuco, Department of Veterinary Medicine, Dois Irmãos, Recife, Pernambuco, Brazil
| | - Regina Célia Bressan Queiroz de Figueiredo
- Oswaldo Cruz Foundation (FIOCRUZ), Aggeu Magalhães Research Centre, Department of Virology, Laboratório de Virologia e Terapia Experimental (LAVITE), Universidade Federal de Pernambuco (UFPE), University City, Recife, Pernambuco, Brazil
| | - Ricardo Galler
- Oswaldo Cruz Foundation (FIOCRUZ), Oswaldo Cruz Institute, Bio-Manguinhos, Laboratório de Tecnologia Virológica (LATEV), Manguinhos, Rio de Janeiro, Brazil
| | - Marcos da Silva Freire
- Oswaldo Cruz Foundation (FIOCRUZ), Oswaldo Cruz Institute, Bio-Manguinhos, Laboratório de Tecnologia Virológica (LATEV), Manguinhos, Rio de Janeiro, Brazil
| | - Joseph Thomas August
- Johns Hopkins University, School of Medicine, Department of Pharmacology & Molecular Sciences, Baltimore, Maryland, United States of America
| | - Ernesto T. A. Marques
- Johns Hopkins University, School of Medicine, Department of Pharmacology & Molecular Sciences, Baltimore, Maryland, United States of America
- Oswaldo Cruz Foundation (FIOCRUZ), Aggeu Magalhães Research Centre, Department of Virology, Laboratório de Virologia e Terapia Experimental (LAVITE), Universidade Federal de Pernambuco (UFPE), University City, Recife, Pernambuco, Brazil
- University of Pittsburgh, Center for Vaccine Research, Pittsburgh, Pennsylvania, United States of America
| | - Rafael Dhalia
- Oswaldo Cruz Foundation (FIOCRUZ), Aggeu Magalhães Research Centre, Department of Virology, Laboratório de Virologia e Terapia Experimental (LAVITE), Universidade Federal de Pernambuco (UFPE), University City, Recife, Pernambuco, Brazil
| |
Collapse
|
25
|
Abstract
DNA vaccines represent a novel approach for protective and therapeutic intervention against type I allergies. In contrast to classical subcutaneous immunotherapy, which relies on the injection of alum-adsorbed protein extracts, DNA vaccines do not suffer from side effects such as anaphylaxis or therapy-induced IgE antibodies. In animal models, DNA vaccines have been demonstrated to prevent TH2 sensitization or balance an existing TH2-mediated allergic immune response by induction of TH1 or regulatory T cells, rendering them promising candidates for prophylactic vaccination as well as therapy. In this chapter, we discuss methods relevant for evaluation of DNA expression vectors for targeting antigen to different cellular compartments for use as a vaccine in an asthma mouse model. Attaching signal sequences has proven to be a successful way to manipulate and boost the immune responses following DNA immunization and also creating hypoallergenic DNA vaccines.
Collapse
|
26
|
Akasaki K, Shiotsu K, Michihara A, Ide N, Wada I. Constitutive expression of a COOH-terminal leucine mutant of lysosome-associated membrane protein-1 causes its exclusive localization in low density intracellular vesicles. J Biochem 2014; 156:39-49. [PMID: 24695761 DOI: 10.1093/jb/mvu020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Lysosome-associated membrane protein-1 (LAMP-1) is a type I transmembrane protein with a short cytoplasmic tail that possesses a lysosome-targeting signal of GYQTI(382)-COOH. Wild-type (WT)-LAMP-1 was exclusively localized in high density lysosomes, and efficiency of LAMP-1's transport to lysosomes depends on its COOH-terminal amino acid residue. Among many different COOH-terminal amino acid substitution mutants of LAMP-1, a leucine-substituted mutant (I382L) displays the most efficient targeting to late endosomes and lysosomes [Akasaki et al. (2010) J. Biochem. 148: , 669-679]. In this study, we generated two human hepatoma cell lines (HepG2 cell lines) that stably express WT-LAMP-1 and I382L, and compared their intracellular distributions. The subcellular fractionation study using Percoll density gradient centrifugation revealed that WT-LAMP-1 had preferential localization in the high density secondary lysosomes where endogenous human LAMP-1 was enriched. In contrast, a major portion of I382L was located in a low density fraction. The low density fraction also contained approximately 80% of endogenous human LAMP-1 and significant amounts of endogenous β-glucuronidase and LAMP-2, which probably represents occurrence of low density lysosomes in the I382L-expressing cells. Double immunofluorescence microscopic analyses distinguished I382L-containing intracellular vesicles from endogenous LAMP-1-containing lysosomes and early endosomes. Altogether, constitutive expression of I382L causes its aberrant intracellular localization and generation of low density lysosomes, indicating that the COOH-terminal isoleucine is critical for normal localization of LAMP-1 in the dense lysosomes.
Collapse
Affiliation(s)
- Kenji Akasaki
- Faculty of Pharmacy and Pharmaceutical Sciences, Fukuyama University, Fukuyama, Hiroshima 729-0292; and Department of Cell Science, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Keiko Shiotsu
- Faculty of Pharmacy and Pharmaceutical Sciences, Fukuyama University, Fukuyama, Hiroshima 729-0292; and Department of Cell Science, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Akihiro Michihara
- Faculty of Pharmacy and Pharmaceutical Sciences, Fukuyama University, Fukuyama, Hiroshima 729-0292; and Department of Cell Science, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Norie Ide
- Faculty of Pharmacy and Pharmaceutical Sciences, Fukuyama University, Fukuyama, Hiroshima 729-0292; and Department of Cell Science, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Ikuo Wada
- Faculty of Pharmacy and Pharmaceutical Sciences, Fukuyama University, Fukuyama, Hiroshima 729-0292; and Department of Cell Science, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| |
Collapse
|
27
|
Godinho RMDC, Matassoli FL, Lucas CGDO, Rigato PO, Gonçalves JLS, Sato MN, Maciel M, Peçanha LMT, August JT, Marques ETDA, de Arruda LB. Regulation of HIV-Gag expression and targeting to the endolysosomal/secretory pathway by the luminal domain of lysosomal-associated membrane protein (LAMP-1) enhance Gag-specific immune response. PLoS One 2014; 9:e99887. [PMID: 24932692 PMCID: PMC4059647 DOI: 10.1371/journal.pone.0099887] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Accepted: 05/19/2014] [Indexed: 12/17/2022] Open
Abstract
We have previously demonstrated that a DNA vaccine encoding HIV-p55gag in association with the lysosomal associated membrane protein-1 (LAMP-1) elicited a greater Gag-specific immune response, in comparison to a DNA encoding the native gag. In vitro studies have also demonstrated that LAMP/Gag was highly expressed and was present in MHCII containing compartments in transfected cells. In this study, the mechanisms involved in these processes and the relative contributions of the increased expression and altered traffic for the enhanced immune response were addressed. Cells transfected with plasmid DNA constructs containing p55gag attached to truncated sequences of LAMP-1 showed that the increased expression of gag mRNA required p55gag in frame with at least 741 bp of the LAMP-1 luminal domain. LAMP luminal domain also showed to be essential for Gag traffic through lysosomes and, in this case, the whole sequence was required. Further analysis of the trafficking pathway of the intact LAMP/Gag chimera demonstrated that it was secreted, at least in part, associated with exosome-like vesicles. Immunization of mice with LAMP/gag chimeric plasmids demonstrated that high expression level alone can induce a substantial transient antibody response, but targeting of the antigen to the endolysosomal/secretory pathways was required for establishment of cellular and memory response. The intact LAMP/gag construct induced polyfunctional CD4+ T cell response, which presence at the time of immunization was required for CD8+ T cell priming. LAMP-mediated targeting to endolysosomal/secretory pathway is an important new mechanistic element in LAMP-mediated enhanced immunity with applications to the development of novel anti-HIV vaccines and to general vaccinology field.
Collapse
Affiliation(s)
- Rodrigo Maciel da Costa Godinho
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Flavio Lemos Matassoli
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Paula Ordonhez Rigato
- Laboratorio de Dermatologia e Imunodeficiencias, LIM-56, Departamento de Dermatologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Jorge Luiz Santos Gonçalves
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Maria Notomi Sato
- Laboratorio de Dermatologia e Imunodeficiencias, LIM-56, Departamento de Dermatologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Milton Maciel
- Enteric Diseases Department, Infectious Diseases Directorate, Naval Medical Research Center, Silver Spring, Maryland, United States of America; Department of Pharmacology and Molecular Sciences, The Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
| | - Ligia Maria Torres Peçanha
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - J Thomas August
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
| | - Ernesto Torres de Azevedo Marques
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America; Department of Infectious Diseases and Microbiology, Center for Vaccine Research, Pittsburgh, Pennsylvania, United States of America; Departamento de Virologia, Fiocruz - Pernambuco, Recife, Brazil
| | - Luciana Barros de Arruda
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
28
|
Garrod TJ, Grubor-Bauk B, Gargett T, Li Y, Miller DS, Yu W, Major L, Burrell CJ, Wesselingh S, Suhrbier A, Gowans EJ. DNA vaccines encoding membrane-bound or secreted forms of heat shock protein 70 exhibit improved potency. Eur J Immunol 2014; 44:1992-2002. [PMID: 24723366 DOI: 10.1002/eji.201343983] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 02/17/2014] [Accepted: 03/31/2014] [Indexed: 11/07/2022]
Abstract
Traditional vaccine strategies are inefficient against challenge with complex pathogens including HIV; therefore, novel vaccine technologies are required. DNA vaccines are attractive as they are relatively cheap and easy to manufacture, but a major limitation has been their lack of immunogenicity in humans, which may be overcome with the incorporation of an adjuvant. HSP70 is a recognised damage-associated molecular pattern, which is a potential adjuvant. We investigated the immunogenicity of a DNA vaccine encoding HIV gag and HSP70; the latter was genetically modified to produce cytoplasmic, secreted or membrane-bound HSP70, the expression of which was controlled by an independent promoter. The DNA was administered to C57BL/6 mice to evaluate gag-specific T-cell responses. Our results demonstrated the ability of membrane-bound and secreted HSP70 to significantly enhance gag-specific T-cell responses and increase the breadth of T-cell responses to include subdominant epitopes. Membrane-bound or secreted HSP70 also significantly improved the multifunctionality of HIV-specific T cells and T-cell proliferation, which is important for maintaining T-cell integrity. Most importantly, the inclusion of membrane-bound HSP70, secreted HSP70 or a combination significantly increased protection in mice challenged with EcoHIV, a chimeric virus that replicates in mouse leukocytes in vivo.
Collapse
Affiliation(s)
- Tamsin J Garrod
- Department of Surgery, Virology Laboratory, Basil Hetzel Institute, University of Adelaide, Adelaide, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Kulkarni V, Rosati M, Valentin A, Ganneru B, Singh AK, Yan J, Rolland M, Alicea C, Beach RK, Zhang GM, Le Gall S, Broderick KE, Sardesai NY, Heckerman D, Mothe B, Brander C, Weiner DB, Mullins JI, Pavlakis GN, Felber BK. HIV-1 p24(gag) derived conserved element DNA vaccine increases the breadth of immune response in mice. PLoS One 2013; 8:e60245. [PMID: 23555935 PMCID: PMC3610668 DOI: 10.1371/journal.pone.0060245] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 02/24/2013] [Indexed: 11/18/2022] Open
Abstract
Viral diversity is considered a major impediment to the development of an effective HIV-1 vaccine. Despite this diversity, certain protein segments are nearly invariant across the known HIV-1 Group M sequences. We developed immunogens based on the highly conserved elements from the p24gag region according to two principles: the immunogen must (i) include strictly conserved elements of the virus that cannot mutate readily, and (ii) exclude both HIV regions capable of mutating without limiting virus viability, and also immunodominant epitopes located in variable regions. We engineered two HIV-1 p24gag DNA immunogens that express 7 highly Conserved Elements (CE) of 12–24 amino acids in length and differ by only 1 amino acid in each CE (‘toggle site’), together covering >99% of the HIV-1 Group M sequences. Altering intracellular trafficking of the immunogens changed protein localization, stability, and also the nature of elicited immune responses. Immunization of C57BL/6 mice with p55gag DNA induced poor, CD4+ mediated cellular responses, to only 2 of the 7 CE; in contrast, vaccination with p24CE DNA induced cross-clade reactive, robust T cell responses to 4 of the 7 CE. The responses were multifunctional and composed of both CD4+ and CD8+ T cells with mature cytotoxic phenotype. These findings provide a method to increase immune response to universally conserved Gag epitopes, using the p24CE immunogen. p24CE DNA vaccination induced humoral immune responses similar in magnitude to those induced by p55gag, which recognize the virus encoded p24gag protein. The inclusion of DNA immunogens composed of conserved elements is a promising vaccine strategy to induce broader immunity by CD4+ and CD8+ T cells to additional regions of Gag compared to vaccination with p55gag DNA, achieving maximal cross-clade reactive cellular and humoral responses.
Collapse
Affiliation(s)
- Viraj Kulkarni
- Human Retrovirus Pathogenesis Section, Center for Cancer Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Margherita Rosati
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Antonio Valentin
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Brunda Ganneru
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Ashish K. Singh
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Jian Yan
- University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Morgane Rolland
- Departments of Microbiology Medicine and Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
| | - Candido Alicea
- Human Retrovirus Pathogenesis Section, Center for Cancer Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Rachel Kelly Beach
- Human Retrovirus Pathogenesis Section, Center for Cancer Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Gen-Mu Zhang
- Human Retrovirus Pathogenesis Section, Center for Cancer Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Sylvie Le Gall
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, United States of America
| | - Kate E. Broderick
- Inovio Pharmaceuticals, Inc., Blue Bell, Pennsylvania, United States of America
| | | | - David Heckerman
- Microsoft Research, Redmond, Washington, United States of America
| | - Beatriz Mothe
- IrsiCaixa AIDS Research Institute-HIVACAT, Autonomous University of Barcelona, Barcelona, Spain
| | - Christian Brander
- IrsiCaixa AIDS Research Institute-HIVACAT, Autonomous University of Barcelona, Barcelona, Spain
- Institucio Catalana de Recerca i Estudis Avancats (ICREA), Barcelona, Spain
| | - David B. Weiner
- University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - James I. Mullins
- Departments of Microbiology Medicine and Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
| | - George N. Pavlakis
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
- * E-mail: (BKF); (GNP)
| | - Barbara K. Felber
- Human Retrovirus Pathogenesis Section, Center for Cancer Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
- * E-mail: (BKF); (GNP)
| |
Collapse
|
30
|
Li L, Saade F, Petrovsky N. The future of human DNA vaccines. J Biotechnol 2012; 162:171-82. [PMID: 22981627 DOI: 10.1016/j.jbiotec.2012.08.012] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Revised: 08/01/2012] [Accepted: 08/06/2012] [Indexed: 01/03/2023]
Abstract
DNA vaccines have evolved greatly over the last 20 years since their invention, but have yet to become a competitive alternative to conventional protein or carbohydrate based human vaccines. Whilst safety concerns were an initial barrier, the Achilles heel of DNA vaccines remains their poor immunogenicity when compared to protein vaccines. A wide variety of strategies have been developed to optimize DNA vaccine immunogenicity, including codon optimization, genetic adjuvants, electroporation and sophisticated prime-boost regimens, with each of these methods having its advantages and limitations. Whilst each of these methods has contributed to incremental improvements in DNA vaccine efficacy, more is still needed if human DNA vaccines are to succeed commercially. This review foresees a final breakthrough in human DNA vaccines will come from application of the latest cutting-edge technologies, including "epigenetics" and "omics" approaches, alongside traditional techniques to improve immunogenicity such as adjuvants and electroporation, thereby overcoming the current limitations of DNA vaccines in humans.
Collapse
Affiliation(s)
- Lei Li
- Vaxine Pty Ltd, Bedford Park, Adelaide 5042, Australia
| | | | | |
Collapse
|
31
|
Jalah R, Patel V, Kulkarni V, Rosati M, Alicea C, Ganneru B, von Gegerfelt A, Huang W, Guan Y, Broderick KE, Sardesai NY, LaBranche C, Montefiori DC, Pavlakis GN, Felber BK. IL-12 DNA as molecular vaccine adjuvant increases the cytotoxic T cell responses and breadth of humoral immune responses in SIV DNA vaccinated macaques. Hum Vaccin Immunother 2012; 8:1620-9. [PMID: 22894956 DOI: 10.4161/hv.21407] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Intramuscular injection of macaques with an IL-12 expression plasmid (0.1 or 0.4 mg DNA/animal) optimized for high level of expression and delivered using in vivo electroporation, resulted in the detection of systemic IL-12 cytokine in the plasma. Peak levels obtained by day 4-5 post injection were paralleled by a rapid increase of IFN-γ, indicating bioactivity of the IL-12 cytokine. Both plasma IL-12 and IFN-γ levels were reduced to basal levels by day 14, indicating a short presence of elevated levels of the bioactive IL-12. The effect of IL-12 as adjuvant together with an SIVmac239 DNA vaccine was further examined comparing two groups of rhesus macaques vaccinated in the presence or absence of IL-12 DNA. The IL-12 DNA-adjuvanted group developed significantly higher SIV-specific cellular immune responses, including IFN-γ (+) Granzyme B (+) T cells, demonstrating increased levels of vaccine-induced T cells with cytotoxic potential, and this difference persisted for 6 mo after the last vaccination. Coinjection of IL-12 DNA led to increases in Gag-specific CD4 (+) and CD4 (+) CD8 (+) double-positive memory T cell subsets, whereas the Env-specific increases were mainly mediated by the CD8 (+) and CD4 (+) CD8 (+) double-positive memory T cell subsets. The IL-12 DNA-adjuvanted vaccine group developed higher binding antibody titers to Gag and mac251 Env, and showed higher and more durable neutralizing antibodies to heterologous SIVsmE660. Therefore, co-delivery of IL-12 DNA with the SIV DNA vaccine enhanced the magnitude and breadth of immune responses in immunized rhesus macaques, and supports the inclusion of IL-12 DNA as vaccine adjuvant.
Collapse
Affiliation(s)
- Rashmi Jalah
- Human Retrovirus Pathogenesis Section; Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Sato MN. Maternal antibodies as an immunotherapeutic strategy in the newborn. Immunotherapy 2012; 4:659-62. [PMID: 22853749 DOI: 10.2217/imt.12.57] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
33
|
Wei Y, Sun Y, Song C, Li H, Li Y, Zhang K, Gong J, Liu F, Liu Z, August JT, Jin B, Yang K. Enhancement of DNA vaccine efficacy by targeting the xenogeneic human chorionic gonadotropin, survivin and vascular endothelial growth factor receptor 2 combined tumor antigen to the major histocompatibility complex class II pathway. J Gene Med 2012; 14:353-62. [PMID: 22438278 DOI: 10.1002/jgm.2624] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND A number of strategies have been used to improve the efficacy of the DNA vaccine for the treatment of tumors. These strategies, ranging from activating CD4+ T cell, manipulating antigen presentation and/or processing to anti-angiogenesis, focus on one certain aspect in the functioning of the vaccine. Therefore, their combination is necessary for rational DNA vaccines design by synergizing different regimens and overcoming the limitations of each strategy. METHODS A DNA fragment (HSV) encoding the C terminal 37 amino acids of human chorionic gonadotropin β chain (hCGβ), 5 different HLA-restricted cytotoxic T lymphocyte epitopes from human survivin and the third and fourth extracellular domains of vascular endothelial growth factor receptor 2 (VEGFR2) was inserted into the sequence between the luminal and transmembrane domain of human lysosome-associated membrane protein-1 cDNA for the construction of a novel DNA vaccine. RESULTS This novel vaccine, named p-L/HSV, has a potent antitumor effect on the LL/2 lung carcinoma model in syngeneic C57BL/6 mice. The immunologic mechanism involved in the antitumor effect referred to the activation of both cellular and humoral immune response. In addition, the tumor vasculature was abrogated as observed by immunohistochemistry in p-L/HSV immunized mice. Furthermore, the immunized mice received an additional boost with p-L/HSV 6 months later and showed a strong immune recall response. CONCLUSIONS The present study indicates that the strategies of combining antitumor with antiangiogenesis and targeting the tumor antigen to the major histocompatibility complex class II pathway cooperate well. Such a study may shed new light on designing vaccine for cancer in the future.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/genetics
- CD8-Positive T-Lymphocytes
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/genetics
- Carcinoma, Lewis Lung/genetics
- Carcinoma, Lewis Lung/therapy
- Chorionic Gonadotropin, beta Subunit, Human/genetics
- Epitopes/genetics
- Female
- Genetic Vectors/administration & dosage
- HEK293 Cells
- Humans
- Immunity, Active/genetics
- Inhibitor of Apoptosis Proteins/genetics
- Inhibitor of Apoptosis Proteins/immunology
- Lysosomal Membrane Proteins/genetics
- Lysosomal Membrane Proteins/immunology
- Major Histocompatibility Complex/genetics
- Major Histocompatibility Complex/immunology
- Mice
- Mice, Inbred C57BL
- Survivin
- T-Lymphocytes, Cytotoxic/immunology
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/genetics
- Vascular Endothelial Growth Factor Receptor-2/genetics
- Vascular Endothelial Growth Factor Receptor-2/immunology
Collapse
Affiliation(s)
- Yuying Wei
- Department of Immunology, The Fourth Military Medical University, Xi'an, Shaanxi Province, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Rigato PO, Maciel M, Goldoni AL, Piubelli OG, Orii NM, Marques ET, August JT, Duarte AJDS, Sato MN. Maternal LAMP/p55gagHIV-1 DNA immunization induces in utero priming and a long-lasting immune response in vaccinated neonates. PLoS One 2012; 7:e31608. [PMID: 22355381 PMCID: PMC3280311 DOI: 10.1371/journal.pone.0031608] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Accepted: 01/16/2012] [Indexed: 12/21/2022] Open
Abstract
Infants born to HIV-infected mothers are at high risk of becoming infected during gestation or the breastfeeding period. A search is thus warranted for vaccine formulations that will prevent mother-to-child HIV transmission. The LAMP/gag DNA chimeric vaccine encodes the HIV-1 p55gag fused to the lysosome-associated membrane protein-1 (LAMP-1) and has been shown to enhance anti-Gag antibody (Ab) and cellular immune responses in adult and neonatal mice; such a vaccine represents a new concept in antigen presentation. In this study, we evaluated the effect of LAMP/gag DNA immunization on neonates either before conception or during pregnancy. LAMP/gag immunization of BALB/c mice before conception by the intradermal route led to the transfer of anti-Gag IgG1 Ab through the placenta and via breastfeeding. Furthermore, there were an increased percentage of CD4+CD25+Foxp3+T cells in the spleens of neonates. When offspring were immunized with LAMP/gag DNA, the anti-Gag Ab response and the Gag-specific IFN-γ-secreting cells were decreased. Inhibition of anti-Gag Ab production and cellular responses were not observed six months after immunization, indicating that maternal immunization did not interfere with the long-lasting memory response in offspring. Injection of purified IgG in conjunction with LAMP/gag DNA immunization decreased humoral and cytotoxic T-cell responses. LAMP/gag DNA immunization by intradermal injection prior to conception promoted the transfer of Ab, leading to a diminished response to Gag without interfering with the development of anti-Gag T- and B-cell memory. Finally, we assessed responses after one intravenous injection of LAMP/gag DNA during the last five days of pregnancy. The intravenous injection led to in utero immunization. In conclusion, DNA vaccine enconding LAMP-1 with Gag and other HIV-1 antigens should be considered in the development of a protective vaccine for the maternal/fetal and newborn periods.
Collapse
Affiliation(s)
- Paula Ordonhez Rigato
- Laboratory of Dermatology and Immunodeficiencies, LIM-56, Department of Dermatology, Medical School, University of São Paulo, São Paulo, Brazil
| | - Milton Maciel
- Enteric Diseases Department, Infectious Diseases Directorate, Naval Medical Research Center, Silver Spring, Maryland, United States of America
| | - Adriana Letícia Goldoni
- Laboratory of Dermatology and Immunodeficiencies, LIM-56, Department of Dermatology, Medical School, University of São Paulo, São Paulo, Brazil
| | - Orlando Guerra Piubelli
- Laboratory of Dermatology and Immunodeficiencies, LIM-56, Department of Dermatology, Medical School, University of São Paulo, São Paulo, Brazil
| | - Noemia Mie Orii
- Laboratory of Dermatology and Immunodeficiencies, LIM-56, Department of Dermatology, Medical School, University of São Paulo, São Paulo, Brazil
| | - Ernesto Torres Marques
- Department of Infectious Diseases and Microbiology, Center for Vaccine Research, Pittsburgh, Pennsylvania, United States of America
| | - Joseph Thomas August
- Department of Pharmacology and Molecular Sciences, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Alberto José da Silva Duarte
- Laboratory of Dermatology and Immunodeficiencies, LIM-56, Department of Dermatology, Medical School, University of São Paulo, São Paulo, Brazil
| | - Maria Notomi Sato
- Laboratory of Dermatology and Immunodeficiencies, LIM-56, Department of Dermatology, Medical School, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
35
|
Heterologous plasmid DNA prime-recombinant human adenovirus 5 boost vaccination generates a stable pool of protective long-lived CD8(+) T effector memory cells specific for a human parasite, Trypanosoma cruzi. Infect Immun 2011; 79:2120-30. [PMID: 21357719 DOI: 10.1128/iai.01190-10] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Recently, we described a heterologous prime-boost strategy using plasmid DNA followed by replication-defective human recombinant adenovirus type 5 as a powerful strategy to elicit long-lived CD8(+) T-cell-mediated protective immunity against experimental systemic infection of mice with a human intracellular protozoan parasite, Trypanosoma cruzi. In the present study, we further characterized the protective long-lived CD8(+) T cells. We compared several functional and phenotypic aspects of specific CD8(+) T cells present 14 or 98 days after the last immunizing dose and found the following: (i) the numbers of specific cells were similar, as determined by multimer staining or by determining the number of gamma interferon (IFN-γ)-secreting cells by enzyme-linked immunospot (ELISPOT) assay; (ii) these cells were equally cytotoxic in vivo; (iii) following in vitro stimulation, a slight decline in the frequency of multifunctional cells (CD107a(+) IFN-γ(+) or CD107a(+) IFN-γ(+) tumor necrosis factor alpha positive [TNF-α(+)]) was paralleled by a significant increase of CD107a singly positive cells after 98 days; (iv) the expression of several surface markers was identical, except for the reexpression of CD127 after 98 days; (v) the use of genetically deficient mice revealed a role for interleukin-12 (IL-12)/IL-23, but not IFN-γ, in the maintenance of these memory cells; and (vi) subsequent immunizations with an unrelated virus or a plasmid vaccine or the depletion of CD4(+) T cells did not significantly erode the number or function of these CD8(+) T cells during the 15-week period. From these results, we concluded that heterologous plasmid DNA prime-adenovirus boost vaccination generated a stable pool of functional protective long-lived CD8(+) T cells with an effector memory phenotype.
Collapse
|
36
|
Rigato PO, Fusaro AE, Victor JR, Sato MN. Maternal immunization to modulate the development of allergic response and pathogen infections. Immunotherapy 2011; 1:141-56. [PMID: 20635979 DOI: 10.2217/1750743x.1.1.141] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
This article reviews recent experimental approaches of preventive strategies regarding allergy and infections by pathogens, particularly in early childhood, by targeting maternal immunomodulation. Basic research is essential to understand maternal vaccination as a strategy to control allergic disease and bacterial and viral infections; thus, providing support for future translational research. The environmental stimuli and host genetic factors, along with maternal influences in early life when immune systems are developing and during postnatal life, are essential for the decision between tolerance induction or allergen sensitization. Maternal immunomodulation strategies should serve as a challenge when attempting to halt the spread of allergy responses and viral infections, until the innate and adaptive arms of the immune system of the neonates are competent.
Collapse
Affiliation(s)
- Paula Ordonhez Rigato
- Laboratório de Dermatologia e Imunodeficiências - LIM56, Faculdade de Medicina da Universidade de São Paulo, Instituto de Medicina Tropical - Prédio II, Av Dr Enéas de Carvalho Aguiar, 500, 3 masculine andar, 05403-05000, São Paulo, Brazil
| | | | | | | |
Collapse
|
37
|
Rigato PO, Maciel M, Goldoni AL, Piubelli O, de Brito CA, Fusaro AE, Eurico de Alencar LX, August T, Azevedo Marques ET, da Silva Duarte AJ, Sato MN. Immunization of neonatal mice with LAMP/p55 HIV gag DNA elicits robust immune responses that last to adulthood. Virology 2010; 406:37-47. [PMID: 20667577 DOI: 10.1016/j.virol.2010.06.050] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2010] [Revised: 06/01/2010] [Accepted: 06/27/2010] [Indexed: 10/19/2022]
Abstract
Successful T cell priming in early postnatal life that can generate effective long-lasting responses until adulthood is critical in HIV vaccination strategies because it prevents early sexual initiation and breastfeeding transmission of HIV. A chimeric DNA vaccine encoding p55 HIV gag associated with lysosome-associated membrane protein 1 (LAMP-1; which drives the antigen to the MIIC compartment), has been used to enhance cellular and humoral antigen-specific responses in adult mice and macaques. Herein, we investigated LAMP-1/gag vaccine immunogenicity in the neonatal period in mice and its ability to generate long-lasting effects. Neonatal vaccination with chimeric LAMP/gag generated stronger Gag-specific immune responses, as measured by the breadth of the Gag peptide-specific IFN-gamma, proliferative responsiveness, cytokine production and antibody production, all of which revealed activation of CD4+ T cells as well as the generation of a more robust CTL response compared to gag vaccine alone. To induce long-lived T and B cell memory responses, it was necessary to immunize neonates with the chimeric LAMP/gag DNA vaccine. The LAMP/gag DNA vaccine strategy could be particularly useful for generating an anti-HIV immune response in the early postnatal period capable of inducing long-term immunological memory.
Collapse
Affiliation(s)
- Paula Ordonhez Rigato
- Laboratory of Dermatology and Immunodeficiency, LIM-56, School of Medicine, University of São Paulo, Brazil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Qu BX, Lambracht-Washington D, Fu M, Eagar TN, Stüve O, Rosenberg RN. Analysis of three plasmid systems for use in DNA A beta 42 immunization as therapy for Alzheimer's disease. Vaccine 2010; 28:5280-7. [PMID: 20562015 DOI: 10.1016/j.vaccine.2010.05.054] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2010] [Revised: 04/26/2010] [Accepted: 05/20/2010] [Indexed: 01/01/2023]
Abstract
In an effort to optimize DNA immunization-elicited antibody production responses against A beta 1-42 (A beta 42) as a therapy for Alzheimer's disease (AD), comparisons were made between three distinct plasmid systems using gene gun delivery. Plasmids encoding A beta 42 monomer and a novel A beta 42 trimeric fusion protein were evaluated in conjunction with CMV or Gal4/UAS promoter elements. It was found that vaccination A beta 42 trimer under the Gal4/UAS promoter elicited high levels of anti-A beta 42 antibody production. Serum antibody levels from Gal4/UAS-A beta 42 trimer immunized mice were found to be 16.6+/-5.5 microg/ml compared to 6.5+/-2.5 microg/ml with Gal4/UAS-A beta 42 monomer or even less with CMV-A beta 42 trimer. As compared to monomeric A beta 42 or A beta 42 trimer expressed under the CMV promoter, injection of the Gal4/UAS-A beta 42 trimer induced high levels of A beta 42 antigen expression in tissue suggesting a mechanism for the increase in anti-A beta 42 antibody. Antibodies were found to be primarily IgG1 suggesting a predominant Th2 response (IgG1/IgG2a ratio of 9). Serum from A beta 42 trimer-vaccinated mice was also found to identify amyloid plaques in the brains of APP/PS1 transgenic mice. These results demonstrate the potential therapeutic use of Gal4/UAS DNA A beta 42 trimer immunization in preventing Alzheimer's disease.
Collapse
Affiliation(s)
- Bao-Xi Qu
- Alzheimer's Disease Center, Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9036, USA
| | | | | | | | | | | |
Collapse
|
39
|
Repeated DNA therapeutic vaccination of chronically SIV-infected macaques provides additional virological benefit. Vaccine 2010; 28:1962-74. [PMID: 20188252 DOI: 10.1016/j.vaccine.2009.10.099] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We have previously reported that therapeutic immunization by intramuscular injection of optimized plasmid DNAs encoding SIV antigens effectively induces immune responses able to reduce viremia in antiretroviral therapy (ART)-treated SIVmac251-infected Indian rhesus macaques. We subjected such therapeutically immunized macaques to a second round of therapeutic vaccination using a combination of plasmids expressing SIV genes and the IL-15/IL-15 receptor alpha as molecular adjuvant, which were delivered by the more efficacious in vivo constant-current electroporation. A very strong induction of antigen-specific responses to Gag, Env, Nef, and Pol, during ART (1.2-1.6% of SIV-specific T cells in the circulating T lymphocytes) was obtained with the improved vaccination method. Immunological responses were characterized by the production of IFN-gamma, IL-2, and TNF-alpha either alone, or in combination as double or triple cytokine positive multifunctional T cells. A significant induction of CD4(+) T cell responses, mainly targeting Gag, Nef, and Pol, as well as of CD8(+) T cells, mainly targeting Env, was found in both T cells with central memory and effector memory markers. After release from ART, the animals showed a virological benefit with a further approximately 1 log reduction in viremia. Vaccination with plasmid DNAs has several advantages over other vaccine modalities, including the possibility for repeated administration, and was shown to induce potent, efficacious, and long-lasting recall immune responses. Therefore, these data support the concept of adding DNA vaccination to the HAART regimen to boost the HIV-specific immune responses.
Collapse
|
40
|
De Keersmaecker B, Heirman C, Allard S, Bonehill A, Corthals J, Thielemans K, Aerts JL. Lumenal part of the DC-LAMP protein is not required for induction of antigen-specific T cell responses by means of antigen-DC-LAMP messenger RNA-electroporated dendritic cells. Hum Gene Ther 2010; 21:479-85. [PMID: 19903083 DOI: 10.1089/hum.2009.080] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Previous studies showed that stimulation of T cells derived from HIV-1-infected patients with autologous dendritic cells electroporated with mRNA encoding HIV antigens can induce antigen-specific T cell responses in vitro. Linking the antigen to an MHC class II-targeting sequence, such as dendritic cell lysosome-associated membrane protein (DC-LAMP), in the mRNA construct results in presentation of antigenic peptides in both MHC class I and class II molecules and therefore enhances the induced T cell responses. To analyze whether the lumenal domain of DC-LAMP is required for optimal induction of cellular immunity against HIV antigens, we compared fusion constructs with or without the lumenal domain of the DC-LAMP protein. A human codon-optimized consensus Gag sequence and a chimeric cDNA sequence encompassing Tat, Rev, and Nef codons (TaReNef ) were cloned into a vector containing the DC-LAMP sequence with or without its lumenal domain. The Gag protein lacking the DC-LAMP-derived sequence altogether elicited only weak T cell responses. DCs electroporated with Gag or TaReNef linked to DC-LAMP were able to elicit similar levels of antigen-specific CD4(+) and CD8(+) T cell responses for both Gag and TaReNef, irrespective of the addition of the DC-LAMP lumenal domain. These data show that DC-LAMP-mediated antigen targeting is absolutely required for optimal T cell stimulation, but that in our experimental setup, the lumenal part of DC-LAMP does not improve the overall induction of antigen-specific T cell responses.
Collapse
Affiliation(s)
- Brenda De Keersmaecker
- Laboratory of Molecular and Cellular Therapy, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | | | | | | | | | | | | |
Collapse
|
41
|
Carvalho JA, Azzoni AR, Prazeres DMF, Monteiro GA. Comparative analysis of antigen-targeting sequences used in DNA vaccines. Mol Biotechnol 2010; 44:204-12. [PMID: 20013075 DOI: 10.1007/s12033-009-9229-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Plasmid vectors can be optimized by including specific signals that promote antigen targeting to the major antigen presentation and processing pathways, increasing the immunogenicity and potency of DNA vaccines. A pVAX1-based backbone was used to encode the Green Fluorescence Protein (GFP) reporter gene fused either to ISG (Invariant Surface Glycoprotein) or to TSA (trans-sialidase) Trypanosoma brucei genes. The plasmids were further engineered to carry antigen-targeting sequences, which promote protein transport to the extracellular space (secretion signal), lysosomes (LAMP-1) and to the endoplasmic reticulum (adenovirus e1a). Transfection efficiency was not affected by differences in the size between each construct as no differences in the plasmid copy number per cell were found. This finding also suggests that the addition of both ISG gene and targeting sequences did not add sensitive regions prone to nuclease attack to the plasmid. Cells transfected with pVAX1GFP had a significant higher number of transcripts. This could be a result of lower mRNA stability and/or a lower transcription rate associated with the bigger transcripts. On the other hand, no differences were found between transcript levels of each ISG-GFP plasmids. Therefore, the addition of these targeting sequences does not affect the maturation/stability of the transcripts. Microscopy analysis showed differences in protein localization and fluorescent levels of cells transfected with pVAX1GFP and ISG constructs. Moreover, cells transfected with the lamp and secretory sequences presented a distinct distribution pattern when compared with ISG protein. Protein expression was quantified by flow cytometry. Higher cell fluorescence was observed in cells expressing the cytoplasmic fusion protein (ISG-GFP or TSA-GFP) compared with cells where the protein was transported to the lysosomal pathway. Protein transport to the endoplasmic reticulum does not lead to a decrease in the mean fluorescence values. The secretion signal was only effective when used in conjunction with TSA gene. Therefore, the characteristics of each protein (e.g., presence of transmembrane domains) might influence the efficacy of its cellular transport. This analysis constitutes a useful tool for the optimization of the design of DNA vaccines.
Collapse
Affiliation(s)
- Joana A Carvalho
- IBB-Institute for Biotechnology and Bioengineering, Centre for Biological and Chemical Engineering, Instituto Superior Técnico, Lisbon, Portugal
| | | | | | | |
Collapse
|
42
|
Starodubova E, Boberg A, Ivanov A, Latyshev O, Petrakova N, Kuzmenko Y, Litvina M, Chernousov A, Kochetkov S, Karpov V, Wahren B, Isaguliants MG. Potent cross-reactive immune response against the wild-type and drug-resistant forms of HIV reverse transcriptase after the chimeric gene immunization. Vaccine 2010; 28:1975-1986. [PMID: 20188253 DOI: 10.1016/j.vaccine.2009.10.098] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
HIV reverse transcriptase (RT) can be considered as a target and an instrument of immunotherapy aimed at limiting the emergence and spread of drug-resistant HIV. The chimeric genes coding for the wild-type and multi-drug-resistant RT (RT1.14) fused to lysosome-associated membrane protein 1 (LAMP-1) were injected intramuscularly into BALB/c mice. The immune response was assessed by ELISpot, cytokine ELISA intracellular IFN-gamma staining, and antibody ELISA. The genes for RT- and RT1.14-LAMP fusions (RT-LAMP and RT1.14-LAMP) were immunogenic generating a mixed Th1/Th2-profile of immune response, while the wild-type RT gene induced only weak immune response. Specific secretion of Th1-cytokines increased with increasing level of RT modification: RT
Collapse
Affiliation(s)
- Elizaveta Starodubova
- Swedish Institute for Infectious Disease Control, Nobelsvägen 18, 17182 Stockholm, Sweden.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Simon GG, Hu Y, Khan AM, Zhou J, Salmon J, Chikhlikar PR, Jung KO, Marques ETA, August JT. Dendritic cell mediated delivery of plasmid DNA encoding LAMP/HIV-1 Gag fusion immunogen enhances T cell epitope responses in HLA DR4 transgenic mice. PLoS One 2010; 5:e8574. [PMID: 20052293 PMCID: PMC2797323 DOI: 10.1371/journal.pone.0008574] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2009] [Accepted: 12/03/2009] [Indexed: 11/18/2022] Open
Abstract
This report describes the identification and bioinformatics analysis of HLA-DR4-restricted HIV-1 Gag epitope peptides, and the application of dendritic cell mediated immunization of DNA plasmid constructs. BALB/c (H-2d) and HLA-DR4 (DRA1*0101, DRB1*0401) transgenic mice were immunized with immature dendritic cells transfected by a recombinant DNA plasmid encoding the lysosome-associated membrane protein-1/HIV-1 Gag (pLAMP/gag) chimera antigen. Three immunization protocols were compared: 1) primary subcutaneous immunization with 1×105 immature dendritic cells transfected by electroporation with the pLAMP/gag DNA plasmid, and a second subcutaneous immunization with the naked pLAMP/gag DNA plasmid; 2) primary immunization as above, and a second subcutaneous immunization with a pool of overlapping peptides spanning the HIV-1 Gag sequence; and 3) immunization twice by subcutaneous injection of the pLAMP/gag DNA plasmid. Primary immunization with pLAMP/gag-transfected dendritic cells elicited the greatest number of peptide specific T-cell responses, as measured by ex vivo IFN-γ ELISpot assay, both in BALB/c and HLA-DR4 transgenic mice. The pLAMP/gag-transfected dendritic cells prime and naked DNA boost immunization protocol also resulted in an increased apparent avidity of peptide in the ELISpot assay. Strikingly, 20 of 25 peptide-specific T-cell responses in the HLA-DR4 transgenic mice contained sequences that corresponded, entirely or partially to 18 of the 19 human HLA-DR4 epitopes listed in the HIV molecular immunology database. Selection of the most conserved epitope peptides as vaccine targets was facilitated by analysis of their representation and variability in all reported sequences. These data provide a model system that demonstrates a) the superiority of immunization with dendritic cells transfected with LAMP/gag plasmid DNA, as compared to naked DNA, b) the value of HLA transgenic mice as a model system for the identification and evaluation of epitope-based vaccine strategies, and c) the application of variability analysis across reported sequences in public databases for selection of historically conserved HIV epitopes as vaccine targets.
Collapse
Affiliation(s)
- Gregory G Simon
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America.
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Dhalia R, Maciel M, Cruz FSP, Viana IFT, Palma ML, August T, Marques ETA. Membrane and envelope virus proteins co-expressed as lysosome associated membrane protein (LAMP) fused antigens: a potential tool to develop DNA vaccines against flaviviruses. AN ACAD BRAS CIENC 2009; 81:663-9. [PMID: 19893892 DOI: 10.1590/s0001-37652009000400005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2008] [Accepted: 03/03/2009] [Indexed: 11/21/2022] Open
Abstract
Vaccination is the most practical and cost-effective strategy to prevent the majority of the flavivirus infection to which there is an available vaccine. However, vaccines based on attenuated virus can potentially promote collateral side effects and even rare fatal reactions. Given this scenario, the development of alternative vaccination strategies such as DNA-based vaccines encoding specific flavivirus sequences are being considered. Endogenous cytoplasmic antigens, characteristically plasmid DNA-vaccine encoded, are mainly presented to the immune system through Major Histocompatibility Complex class I - MHC I molecules. The MHC I presentation via is mostly associated with a cellular cytotoxic response and often do not elicit a satisfactory humoral response. One of the main strategies to target DNA-encoded antigens to the MHC II compartment is expressing the antigen within the Lysosome-Associated Membrane Protein (LAMP). The flavivirus envelope protein is recognized as the major virus surface protein and the main target for neutralizing antibodies. Different groups have demonstrated that co-expression of flavivirus membrane and envelope proteins in mammalian cells, fused with the carboxyl-terminal of LAMP, is able to induce satisfactory levels of neutralizing antibodies. Here we reviewed the use of the envelope flavivirus protein co-expression strategy as LAMP chimeras with the aim of developing DNA vaccines for dengue, West Nile and yellow fever viruses.
Collapse
Affiliation(s)
- Rafael Dhalia
- Fundação Oswaldo Cruz, Centro de Pesquisas Aggeu Magalhães, Departamento de Virologia, Laboratório de Virologia e Terapia Experimental, Recife, PE, Brasil.
| | | | | | | | | | | | | |
Collapse
|
45
|
Yang K, Sun K, Srinivasan KN, Salmon J, Marques ET, Xu J, August JT. Immune responses to T-cell epitopes of SARS CoV-N protein are enhanced by N immunization with a chimera of lysosome-associated membrane protein. Gene Ther 2009; 16:1353-62. [PMID: 19727132 PMCID: PMC7091638 DOI: 10.1038/gt.2009.92] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2008] [Revised: 05/11/2009] [Accepted: 06/05/2009] [Indexed: 12/30/2022]
Abstract
In our previous study by Gupta et al, dominant T-cell epitopes of SARS CoV-N(N) protein were predicted by software. The spectrum of interferon (IFN)-gamma responses of Balb/c mice immunized against two different forms of SARS CoV-N plasmid was then analyzed. A cluster of dominant T-cell epitopes of SARS CoV-N protein was found in the N-terminus (amino acids 76-114). On the basis of this study, four different plasmids were constructed: (i) DNA encoding the unmodified N (p-N) or N(70-122) (p-N(70-122)) as an endogenous cytoplasmic protein or (ii) DNA encoding a lysosome-associated membrane protein (LAMP) chimera with N (p-LAMP/N) or N(70-122) (p-LAMP/N(70-122)). The immune responses of mice to these four constructs were evaluated. The results showed marked differences in the responses of the immunized mice. A single priming immunization with the p-LAMP/N construct was sufficient to elicit an antibody response. Enzyme-linked immunospot (ELISpot) assay indicated that p-LAMP/N(70-122) and p-LAMP/N plasmids both elicited a greater IFN-gamma response than p-N. p-N and p-N(70-122) constructs induced low or undetectable levels of cytokine secretion. We also found that the p-LAMP/N(70-122) construct promoted a long-lasting T-cell memory response without an additional boost 6 months after three immunizations. These findings show that DNA vaccines, even epitope-based DNA vaccines using LAMP as chimera, can elicit both humoral and cellular immune responses.
Collapse
Affiliation(s)
- K Yang
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD USA
- Department of Immunology, The Fourth Military Medical University, Xi'an, Shaanxi Province, PR China,
| | - K Sun
- Division of Biomedical Sciences, Johns Hopkins in Singapore, Singapore
| | - K N Srinivasan
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD USA
- Product Evaluation and Registration Division, Centre for Drug Administration, Health Sciences Authority, Singapore
| | - J Salmon
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - E T Marques
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD USA
- Division of Infectious Diseases, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - J Xu
- Department of Neurology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, PR China,
| | - J T August
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD USA
| |
Collapse
|
46
|
Valentin A, Chikhlikar P, Patel V, Rosati M, Maciel M, Chang KH, Silvera P, Felber BK, Pavlakis GN, August JT, Marques ETA. Comparison of DNA vaccines producing HIV-1 Gag and LAMP/Gag chimera in rhesus macaques reveals antigen-specific T-cell responses with distinct phenotypes. Vaccine 2009; 27:4840-9. [PMID: 19539586 PMCID: PMC2743166 DOI: 10.1016/j.vaccine.2009.05.093] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2009] [Revised: 05/16/2009] [Accepted: 05/21/2009] [Indexed: 11/27/2022]
Abstract
Optimized DNA expression vectors encoding the native HIV-1 Gag or a fusion of Gag with the lysosomal membrane associated protein 1 (LAMP) were compared for immunogenicity upon intramuscular DNA delivery in rhesus macaques. Both vaccines elicited CD4(+) T-cell responses, but with significant differences in the phenotype of the Gag-specific cells: the native Gag induced CD4(+) responses with a phenotype of central memory-like T cells (CD28(+) CD45RA(-)), whereas the LAMP/Gag chimera induced CD4(+) responses with effector memory phenotype (CD28(-) CD45RA(-)). Antigen-specific T cells producing both IFN-gamma and TNFalpha were found in the animals receiving the native Gag, whereas the LAMP/Gag chimera induced humoral responses faster. These results demonstrate that modification of intracellular Gag trafficking results in the induction of distinct immune responses. Combinations of DNA vectors encoding both forms of antigen may be more potent in eliciting anti-HIV-1 immunity.
Collapse
Affiliation(s)
- Antonio Valentin
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702-1201, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Kaur M, Rai A, Bhatnagar R. Rabies DNA vaccine: no impact of MHC class I and class II targeting sequences on immune response and protection against lethal challenge. Vaccine 2009; 27:2128-37. [PMID: 19356616 PMCID: PMC7115670 DOI: 10.1016/j.vaccine.2009.01.128] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2008] [Revised: 01/24/2009] [Accepted: 01/29/2009] [Indexed: 11/23/2022]
Abstract
Rabies is progressive fatal encephalitis. WHO estimates 55,000 rabies deaths and more than 10 million PEP every year world-wide. A variety of cell-culture derived vaccines are available for prophylaxis against rabies. However, their high cost restricts their usage in developing countries, where such cases are most often encountered. This is driving the quest for newer vaccine formulations; DNA vaccines being most promising amongst them. Here, we explored strategies of antigen trafficking to various cellular compartments aiming at improving both humoral and cellular immunity. These strategies include use of signal sequences namely Tissue Plasminogen Activator (TPA), Ubiquitin (UQ) and Lysosomal-Associated Membrane Protein-1 (LAMP-1). TPA, LAMP-1 and their combination were aimed at enhancing the CD4(+) T cell and antibody response. In contrast, the UQ tag was utilized for enhancing CD8(+) response. The potency of modified DNA vaccines assessed by total antibody response, antibody isotypes, cytokine profile, neutralizing antibody titer and protection conferred against in vivo challenge; was enhanced in comparison to native unmodified vaccine, but the response elicited did not pertain to the type of target sequence and the directed arm of immunity. Interestingly, the DNA vaccines that had been designed to generate different type of immune responses yielded in effect similar response. In conclusion, our data indicate that the directing target sequence is not the exclusive deciding factor for type and extent of immune response elicited and emphasizes on the antigen dependence of immune enhancement strategies.
Collapse
Key Words
- ab, antibody
- ig, immunoglobulin
- elisa, enzyme linked immunosorbent assay
- gp, glycoprotein
- lamp-1, lysosomal-associated membrane protein-1
- mhc, major histocompatibility complex
- mq, milli quartz water
- pmsf, phenyl methyl sulphonyl fluoride
- ripa, radioimmunoprecipitation assay buffer
- rffit, rapid fluorescence focus inhibition test
- tm, transmembrane
- tpa, tissue plasminogen activator
- tris, tris(hydroxymethyl) aminomethane
- uq, ubiquitin
- targeting sequence
- rabies virus-neutralizing antibody (rvna)
- survival
Collapse
Affiliation(s)
- Manpreet Kaur
- Laboratory Of Molecular Biology And Genetic Engineering, School Of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, Delhi, India
| | | | | |
Collapse
|
48
|
Rosati M, Valentin A, Jalah R, Patel V, von Gegerfelt A, Bergamaschi C, Alicea C, Weiss D, Treece J, Pal R, Markham PD, Marques ETA, August JT, Khan A, Draghia-Akli R, Felber BK, Pavlakis GN. Increased immune responses in rhesus macaques by DNA vaccination combined with electroporation. Vaccine 2008; 26:5223-9. [PMID: 18468743 DOI: 10.1016/j.vaccine.2008.03.090] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
We used optimized DNA expression vectors to compare two gene delivery methodologies in rhesus macaques, namely direct DNA injection and in vivo adaptive constant-current electroporation via the intramuscular route. The use of in vivo electroporation increased levels of gene expression and immune responses. We used an optimized HIV gag expression plasmid to show the development of new cellular immune responses in SIV-infected animals controlling viremia. Furthermore, after vaccination with SIV expression plasmids the recall responses to the SIV antigens were very high, indicating that DNA is a strong boost in the presence of antiretroviral treatment in SIV-infected animals. There was substantial animal-to-animal variability in DNA expression, revealed by plasma measurements of IL-15 produced by co-injected IL-15 DNA. IL-15 expression levels correlated with peak immune responses. Electroporation led to an expansion of antigen-specific CD4+ and CD8+ T cells of both central and effector memory phenotype. These results indicate that improved gene delivery and expression by electroporation dramatically increases immunogenicity of DNA vaccines. Electroporation is thus an important method to improve the effectiveness of DNA vaccination.
Collapse
Affiliation(s)
- Margherita Rosati
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702-1201, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Chung KS, Jang YJ, Kim NS, Park SY, Choi SJ, Kim JY, Ahn JH, Lee HJ, Lim JH, Song JH, Ji JH, Oh JH, Song KB, Yoo HS, Won M. Rapid screen of human genes for relevance to cancer using fission yeast. ACTA ACUST UNITED AC 2007; 12:568-77. [PMID: 17478483 DOI: 10.1177/1087057107301007] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
A total of 437 human full-length cDNAs isolated by microarray analysis of liver and/or gastric cancer tissues were evaluated for their relevance to cancer using the fission yeast Schizosaccharomyces pombe. Overexpression of 161 human cDNAs in S. pombe caused growth inhibition and/or morphological changes, which can be considered as cancer-related phenotypes of S. pombe. Sixteen genes causing growth defects and morphological changes at the same time were chosen to validate their ostensible oncogenic properties. They were highly expressed in liver and/or gastric cancer cell lines. Also, when the mouse embryonic fibroblast cell type NIH3T3 was transfected with these genes, the proliferation rates of cells were increased by 32% to 120%. This study demonstrates that fission yeast can be used as an advantageous and powerful tool for the rapid screening of human genes relevant to cancer. Furthermore, the human genes screened can be tested further as diagnostic markers and potential therapeutic targets for liver and stomach cancers. They also can be studied further for the elucidation of mechanisms involved in carcinogenesis.
Collapse
|
50
|
Young KR, McBurney SP, Karkhanis LU, Ross TM. Virus-like particles: designing an effective AIDS vaccine. Methods 2007; 40:98-117. [PMID: 16997718 DOI: 10.1016/j.ymeth.2006.05.024] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2005] [Accepted: 05/05/2006] [Indexed: 01/10/2023] Open
Abstract
Viruses that infect eukaryotic organisms have the unique characteristic of self-assembling into particles. The mammalian immune system is highly attuned to recognizing and attacking these viral particles following infection. The use of particle-based immunogens, often delivered as live-attenuated viruses, has been an effective vaccination strategy for a variety of viruses. The development of an effective vaccine against the human immunodeficiency virus (HIV) has proven to be a challenge, since HIV infects cells of the immune system causing severe immunodeficiency resulting in the syndrome known as AIDS. In addition, the ability of the virus to adapt to immune pressure and reside in an integrated form in host cells presents hurdles for vaccinologists to overcome. A particle-based vaccine strategy has promise for eliciting high titer, long-lived, immune responses to a diverse number of viral epitopes against different HIV antigens. Live-attenuated viruses are effective at generating both cellular and humoral immune responses. However, while these vaccines stimulate immunity, challenged animals rarely clear the viral infection and the degree of attenuation directly correlates with protection from disease. Further, a live-attenuated vaccine has the potential to revert to a pathogenic form. Alternatively, virus-like particles (VLPs) mimic the viral particle without causing an immunodeficiency disease. VLPs are self-assembling, non-replicating, non-pathogenic particles that are similar in size and conformation to intact virions. A variety of VLPs for lentiviruses are currently in preclinical and clinical trials. This review focuses on our current status of VLP-based AIDS vaccines, regarding issues of purification and immune design for animal and clinical trials.
Collapse
Affiliation(s)
- Kelly R Young
- Department of Medicine, Division of Infectious Diseases, University of Pittsburgh School of Medicine, PA 15261, USA
| | | | | | | |
Collapse
|