1
|
Kong LZ, Kim SM, Wang C, Lee SY, Oh SC, Lee S, Jo S, Kim TD. Understanding nucleic acid sensing and its therapeutic applications. Exp Mol Med 2023; 55:2320-2331. [PMID: 37945923 PMCID: PMC10689850 DOI: 10.1038/s12276-023-01118-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/16/2023] [Accepted: 08/20/2023] [Indexed: 11/12/2023] Open
Abstract
Nucleic acid sensing is involved in viral infections, immune response-related diseases, and therapeutics. Based on the composition of nucleic acids, nucleic acid sensors are defined as DNA or RNA sensors. Pathogen-associated nucleic acids are recognized by membrane-bound and intracellular receptors, known as pattern recognition receptors (PRRs), which induce innate immune-mediated antiviral responses. PRR activation is tightly regulated to eliminate infections and prevent abnormal or excessive immune responses. Nucleic acid sensing is an essential mechanism in tumor immunotherapy and gene therapies that target cancer and infectious diseases through genetically engineered immune cells or therapeutic nucleic acids. Nucleic acid sensing supports immune cells in priming desirable immune responses during tumor treatment. Recent studies have shown that nucleic acid sensing affects the efficiency of gene therapy by inhibiting translation. Suppression of innate immunity induced by nucleic acid sensing through small-molecule inhibitors, virus-derived proteins, and chemical modifications offers a potential therapeutic strategy. Herein, we review the mechanisms and regulation of nucleic acid sensing, specifically covering recent advances. Furthermore, we summarize and discuss recent research progress regarding the different effects of nucleic acid sensing on therapeutic efficacy. This study provides insights for the application of nucleic acid sensing in therapy.
Collapse
Affiliation(s)
- Ling-Zu Kong
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Seok-Min Kim
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Chunli Wang
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Soo Yun Lee
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Se-Chan Oh
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Sunyoung Lee
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
- Department of Life Sciences, Korea University, Seoul, 02841, Korea
| | - Seona Jo
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113, Korea
| | - Tae-Don Kim
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea.
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113, Korea.
- Biomedical Mathematics Group, Institute for Basic Science (IBS), Daejeon, Republic of Korea.
- Department of Biopharmaceutical Convergence, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea.
| |
Collapse
|
2
|
Piri-Gharaghie T, Doosti A, Mirzaei SA. Novel adjuvant nano-vaccine induced immune response against Acinetobacter baumannii. AMB Express 2023; 13:31. [PMID: 36905472 PMCID: PMC10008545 DOI: 10.1186/s13568-023-01531-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 02/21/2023] [Indexed: 03/12/2023] Open
Abstract
Developing adjuvant vaccines to combat rising multidrug-resistant (MDR) Acinetobacter baumannii (A. baumannii) infections is a promising and cost-effective approach. The aim of this analysis was to construct a pDNA-CPG C274-adjuvant nano-vaccine and investigate its immunogenicity and protection in BALB/c mice. The CPG ODN C274 adjuvant was chemically synthesized and cloned into pcDNA3.1( +), and the cloning was verified using PCR and BamHI/EcoRV restriction enzyme digestion. Then, utilizing a complex coacervation approach, pDNA-CPG C274 was encapsulated by chitosan (CS) nanoparticles (NPs). TEM and DLS are used to explore the properties of the pDNA/CSNP complex. TLR-9 pathway activation was investigated in human HEK-293 and RAW 264.7 mouse cells. The vaccine's immunogenicity and immune-protective effectiveness were investigated in BALB/c mice. The pDNA-CPG C274/CSNPs were small (mean size 79.21 ± 0.23 nm), positively charged (+ 38.87 mV), and appeared to be spherical. A continuous slow release pattern was achieved. TLR-9 activation was greatest in the mouse model with CpG ODN (C274) at concentrations of 5 and 10 μg/ml with 56% and 55%, respectively (**P < 0.01). However, in HEK-293 human cells, by increasing the concentration of CpG ODN (C274) from 1 to 50 μg/ml, the activation rate of TLR-9 also increased, so that the highest activation rate (81%) was obtained at the concentration of 50 μg/ml (***P < 0.001). pDNA-CPG C274/CSNPs immunized BALB/c mice produced increased amounts of total-IgG, as well as IFN-γ and IL-1B in serum samples, compared to non-encapsulated pDNA-CPG C274. Furthermore, liver and lung injuries, as well as bacterial loads in the liver, lung, and blood, were reduced, and BALB/c mice immunized with pDNA-CPG C274/CSNPs showed potent protection (50-75%) against acute fatal Intraperitoneal A. baumannii challenge. pDNA-CPG C274/CSNPs evoked total-IgG antibodies, Th1 cellular immunity, and the TLR-9 pathway, as well as protection against an acute fatal A. baumannii challenge. Our findings suggest that this nano-vaccine is a promising approach for avoiding A. baumannii infection when used as a powerful adjuvant.
Collapse
Affiliation(s)
- Tohid Piri-Gharaghie
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Abbas Doosti
- Biotechnology Research Center, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran.
| | - Seyed Abbas Mirzaei
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran.,Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
3
|
Vaccines platforms and COVID-19: what you need to know. Trop Dis Travel Med Vaccines 2022; 8:20. [PMID: 35965345 PMCID: PMC9537331 DOI: 10.1186/s40794-022-00176-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 06/22/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The novel SARS-CoV-2, responsible for the COVID-19 pandemic, is the third zoonotic coronavirus since the beginning of the 21 first century, and it has taken more than 6 million human lives because of the lack of immunity causing global economic losses. Consequently, developing a vaccine against the virus represents the fastest way to finish the threat and regain some "normality." OBJECTIVE Here, we provide information about the main features of the most important vaccine platforms, some of them already approved, to clear common doubts fostered by widespread misinformation and to reassure the public of the safety of the vaccination process and the different alternatives presented. METHODS Articles published in open access databases until January 2022 were identified using the search terms "SARS-CoV-2," "COVID-19," "Coronavirus," "COVID-19 Vaccines," "Pandemic," COVID-19, and LMICs or their combinations. DISCUSSION Traditional first-generation vaccine platforms, such as whole virus vaccines (live attenuated and inactivated virus vaccines), as well as second-generation vaccines, like protein-based vaccines (subunit and viral vector vaccines), and third-generation vaccines, such as nanoparticle and genetic vaccines (mRNA vaccines), are described. CONCLUSIONS SARS-CoV-2 sequence information obtained in a record time provided the basis for the fast development of a COVID-19 vaccine. The adaptability characteristic of the new generation of vaccines is changing our capability to react to emerging threats to future pandemics. Nevertheless, the slow and unfair distribution of vaccines to low- and middle-income countries and the spread of misinformation are a menace to global health since the unvaccinated will increase the chances for resurgences and the surge of new variants that can escape the current vaccines.
Collapse
|
4
|
Ulrich-Lewis JT, Draves KE, Roe K, O’Connor MA, Clark EA, Fuller DH. STING Is Required in Conventional Dendritic Cells for DNA Vaccine Induction of Type I T Helper Cell- Dependent Antibody Responses. Front Immunol 2022; 13:861710. [PMID: 35529875 PMCID: PMC9072870 DOI: 10.3389/fimmu.2022.861710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/14/2022] [Indexed: 02/02/2023] Open
Abstract
DNA vaccines elicit antibody, T helper cell responses and CD8+ T cell responses. Currently, little is known about the mechanism that DNA vaccines employ to induce adaptive immune responses. Prior studies have demonstrated that stimulator of interferon genes (STING) and conventional dendritic cells (cDCs) play critical roles in DNA vaccine induced antibody and T cell responses. STING activation by double stranded (dsDNA) sensing proteins initiate the production of type I interferon (IFN),but the DC-intrinsic effect of STING signaling is still unclear. Here, we investigated the role of STING within cDCs on DNA vaccine induction of antibody and T cell responses. STING knockout (STING-/- ) and conditional knockout mice that lack STING in cDCs (cDC STING cKO), were immunized intramuscularly with a DNA vaccine that expressed influenza A nucleoprotein (pNP). Both STING-/- and cDC STING cKO mice had significantly lower type I T helper (Th1) type antibody (anti-NP IgG2C) responses and lower frequencies of Th1 associated T cells (NP-specific IFN-γ+CD4+ T cells) post-immunization than wild type (WT) and cDC STING littermate control mice. In contrast, all mice had similar Th2-type NP-specific (IgG1) antibody titers. STING-/- mice developed significantly lower polyfunctional CD8+ T cells than WT, cDC STING cKO and cDC STING littermate control mice. These findings suggest that STING within cDCs mediates DNA vaccine induction of type I T helper responses including IFN-γ+CD4+ T cells, and Th1-type IgG2C antibody responses. The induction of CD8+ effector cell responses also require STING, but not within cDCs. These findings are the first to show that STING is required within cDCs to mediate DNA vaccine induced Th1 immune responses and provide new insight into the mechanism whereby DNA vaccines induce Th1 responses.
Collapse
Affiliation(s)
- Justin Theophilus Ulrich-Lewis
- Department of Microbiology, University of Washington, Seattle, WA, United States,Department of Immunology, University of Washington, Seattle, WA, United States
| | - Kevin E. Draves
- Department of Microbiology, University of Washington, Seattle, WA, United States,Department of Immunology, University of Washington, Seattle, WA, United States
| | - Kelsey Roe
- Department of Immunology, University of Washington, Seattle, WA, United States,Seattle Children's Hospital Center for Immunity and Immunotherapies Children’s Hospital, Seattle, WA, United States
| | - Megan A. O’Connor
- Department of Microbiology, University of Washington, Seattle, WA, United States
| | - Edward A. Clark
- Department of Microbiology, University of Washington, Seattle, WA, United States,Department of Immunology, University of Washington, Seattle, WA, United States
| | - Deborah Heydenburg Fuller
- Department of Microbiology, University of Washington, Seattle, WA, United States,*Correspondence: Deborah Heydenburg Fuller,
| |
Collapse
|
5
|
Shamseddine AA, Burman B, Lee NY, Zamarin D, Riaz N. Tumor Immunity and Immunotherapy for HPV-Related Cancers. Cancer Discov 2021; 11:1896-1912. [PMID: 33990345 PMCID: PMC8338882 DOI: 10.1158/2159-8290.cd-20-1760] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/27/2021] [Accepted: 03/16/2021] [Indexed: 12/17/2022]
Abstract
Human papillomavirus (HPV) infection drives tumorigenesis in the majority of cervical, oropharyngeal, anal, and vulvar cancers. Genetic and epidemiologic evidence has highlighted the role of immunosuppression in the oncogenesis of HPV-related malignancies. Here we review how HPV modulates the immune microenvironment and subsequent therapeutic implications. We describe the landscape of immunotherapies for these cancers with a focus on findings from early-phase studies exploring antigen-specific treatments, and discuss future directions. Although responses across these studies have been modest to date, a deeper understanding of HPV-related tumor biology and immunology may prove instrumental for the development of more efficacious immunotherapeutic approaches. SIGNIFICANCE: HPV modulates the microenvironment to create a protumorigenic state of immune suppression and evasion. Our understanding of these mechanisms has led to the development of immunomodulatory treatments that have shown early clinical promise in patients with HPV-related malignancies. This review summarizes our current understanding of the interactions of HPV and its microenvironment and provides insight into the progress and challenges of developing immunotherapies for HPV-related malignancies.
Collapse
Affiliation(s)
- Achraf A Shamseddine
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Bharat Burman
- Department of Medicine, Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nancy Y Lee
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Dmitriy Zamarin
- Department of Medicine, Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Nadeem Riaz
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
6
|
de Queiroz NMGP, Marinho FV, Chagas MA, Leite LCC, Homan EJ, de Magalhães MTQ, Oliveira SC. Vaccines for COVID-19: perspectives from nucleic acid vaccines to BCG as delivery vector system. Microbes Infect 2020; 22:515-524. [PMID: 32961274 PMCID: PMC7501874 DOI: 10.1016/j.micinf.2020.09.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 09/10/2020] [Indexed: 12/12/2022]
Abstract
This article discusses standard and new disruptive strategies in the race to develop an anti-COVID-19 vaccine. We also included new bioinformatic data from our group mapping immunodominant epitopes and structural analysis of the spike protein. Another innovative approach reviewed here is the use of BCG vaccine as priming strategy and/or delivery system expressing SARS-CoV-2 antigens.
Collapse
Affiliation(s)
- Nina Marí G P de Queiroz
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Fabio V Marinho
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Marcelo A Chagas
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luciana C C Leite
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, SP, Brazil
| | | | - Mariana T Q de Magalhães
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Sergio C Oliveira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), CNPq/MCT, BA, Brazil.
| |
Collapse
|
7
|
Hager S, Fittler FJ, Wagner E, Bros M. Nucleic Acid-Based Approaches for Tumor Therapy. Cells 2020; 9:E2061. [PMID: 32917034 PMCID: PMC7564019 DOI: 10.3390/cells9092061] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/06/2020] [Accepted: 09/07/2020] [Indexed: 12/24/2022] Open
Abstract
Within the last decade, the introduction of checkpoint inhibitors proposed to boost the patients' anti-tumor immune response has proven the efficacy of immunotherapeutic approaches for tumor therapy. Furthermore, especially in the context of the development of biocompatible, cell type targeting nano-carriers, nucleic acid-based drugs aimed to initiate and to enhance anti-tumor responses have come of age. This review intends to provide a comprehensive overview of the current state of the therapeutic use of nucleic acids for cancer treatment on various levels, comprising (i) mRNA and DNA-based vaccines to be expressed by antigen presenting cells evoking sustained anti-tumor T cell responses, (ii) molecular adjuvants, (iii) strategies to inhibit/reprogram tumor-induced regulatory immune cells e.g., by RNA interference (RNAi), (iv) genetically tailored T cells and natural killer cells to directly recognize tumor antigens, and (v) killing of tumor cells, and reprograming of constituents of the tumor microenvironment by gene transfer and RNAi. Aside from further improvements of individual nucleic acid-based drugs, the major perspective for successful cancer therapy will be combination treatments employing conventional regimens as well as immunotherapeutics like checkpoint inhibitors and nucleic acid-based drugs, each acting on several levels to adequately counter-act tumor immune evasion.
Collapse
Affiliation(s)
- Simone Hager
- Department of Chemistry and Pharmacy, Ludwig-Maximilians-University (LMU), 81377 Munich, Germany;
| | | | - Ernst Wagner
- Department of Chemistry and Pharmacy, Ludwig-Maximilians-University (LMU), 81377 Munich, Germany;
| | - Matthias Bros
- Department of Dermatology, University Medical Center, 55131 Mainz, Germany;
| |
Collapse
|
8
|
Putzke S, Feldhues E, Heep I, Ilg T, Lamprecht A. Cationic lipid/pDNA complex formation as potential generic method to generate specific IRF pathway stimulators. Eur J Pharm Biopharm 2020; 155:112-121. [PMID: 32798666 DOI: 10.1016/j.ejpb.2020.08.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 05/21/2020] [Accepted: 08/10/2020] [Indexed: 10/23/2022]
Abstract
Cationic liposome - CpG DNA complexes (lipoplexes) are known as stimulators of innate immunity via Toll-like receptor 9 (TLR9)-triggered activation of the nuclear factor kappa B (NF-κB) pathway. More recent reports suggest that DNA lipoplexes also engage DNA sensors in the cytosol leading to the stimulation of the interferon response factor (IRF) pathway. In this study a range of lipoplexes were formulated by using an invariable helper lipid, three different cationic lipids (DOTAP, DOTMA and DDA) and three different CpG-containing plasmids of different sizes. These lipoplexes exhibited similar hydrodynamic diameters, zeta-potentials and plasmid loading rates, despite the different lipid blends and CpG-containing plasmids. Binding and uptake of liposomal lipids by J774.A1 macrophages and JAWSII dendritic cells increased significantly (up to 4-fold) upon lipoplex formation. Cellular plasmid DNA uptake via lipoplexes compared to naked DNA was increased up to 18-fold. Analysis of signal transduction pathway activation in J774-DUAL™ reporter cells by liposomes or naked CpG plasmid DNA compared to their derived lipoplexes showed only minor activation of the NF-κB pathway, while the IRF pathway displayed massive activation factors of up to 46-fold. DOTAP- and DOTMA lipoplexes also led to massive interferon-alpha and -beta secretion of J774A.1 macrophages and JAWSII dendritic cells, which is a hallmark of IRF pathway activation. Cellular distribution studies on DOTAP lipoplexes suggest delivery of plasmid DNA via vesicular compartments into the cytosol. Taken together, the CpG plasmid DNA lipoplexes generated in this study appear to selectively stimulate DNA receptors activating the IRF pathway, while bypassing TLR9 and NF-κB activation.
Collapse
Affiliation(s)
- Simone Putzke
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, 53121 Bonn, Germany; Bayer Animal Health GmbH, 40789 Monheim am Rhein, Germany
| | | | - Iris Heep
- Bayer Animal Health GmbH, 40789 Monheim am Rhein, Germany
| | - Thomas Ilg
- Bayer Animal Health GmbH, 40789 Monheim am Rhein, Germany
| | - Alf Lamprecht
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, 53121 Bonn, Germany.
| |
Collapse
|
9
|
Adam L, Tchitchek N, Todorova B, Rosenbaum P, Joly C, Poux C, Chapon C, Spetz AL, Ustav M, Le Grand R, Martinon F. Innate Molecular and Cellular Signature in the Skin Preceding Long-Lasting T Cell Responses after Electroporated DNA Vaccination. THE JOURNAL OF IMMUNOLOGY 2020; 204:3375-3388. [PMID: 32385135 DOI: 10.4049/jimmunol.1900517] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 04/09/2020] [Indexed: 12/21/2022]
Abstract
DNA vaccines delivered with electroporation (EP) have shown promising results in preclinical models and are evaluated in clinical trials. In this study, we aim to characterize early mechanisms occurring in the skin after intradermal injection and EP of the auxoGTUmultiSIV DNA vaccine in nonhuman primates. First, we show that EP acts as an adjuvant by enhancing local inflammation, notably via granulocytes, monocytes/macrophages, and CD1aint-expressing cell recruitment. EP also induced Langerhans cell maturation, illustrated by CD86, CD83, and HLA-DR upregulation and their migration out of the epidermis. Second, we demonstrate the crucial role of the DNA vaccine in soluble factors release, such as MCP-1 or IL-15. Transcriptomic analysis showed that EP played a major role in gene expression changes postvaccination. However, the DNA vaccine is required to strongly upregulate several genes involved in inflammatory responses (e.g., Saa4), cell migration (e.g., Ccl3, Ccl5, or Cxcl10), APC activation (e.g., Cd86), and IFN-inducible genes (e.g., Ifit3, Ifit5, Irf7, Isg15, orMx1), illustrating an antiviral response signature. Also, AIM-2, a cytosolic DNA sensor, appeared to be strongly upregulated only in the presence of the DNA vaccine and trends to positively correlate with several IFN-inducible genes, suggesting the potential role of AIM-2 in vaccine sensing and the subsequent innate response activation leading to strong adaptive T cell responses. Overall, these results demonstrate that a combined stimulation of the immune response, in which EP and the auxoGTUmultiSIV vaccine triggered different components of the innate immunity, led to strong and persistent cellular recall responses.
Collapse
Affiliation(s)
- Lucille Adam
- Immunology of Viral Infections and Autoimmune Diseases, Infectious Disease Models and Innovative Therapies Department, Commissariat à l'Energie Atomique et aux Energies Alternatives, Université Paris-Sud 11, INSERM U1184, 92265 Fontenay-aux-Roses, France
| | - Nicolas Tchitchek
- Immunology of Viral Infections and Autoimmune Diseases, Infectious Disease Models and Innovative Therapies Department, Commissariat à l'Energie Atomique et aux Energies Alternatives, Université Paris-Sud 11, INSERM U1184, 92265 Fontenay-aux-Roses, France
| | - Biliana Todorova
- Immunology of Viral Infections and Autoimmune Diseases, Infectious Disease Models and Innovative Therapies Department, Commissariat à l'Energie Atomique et aux Energies Alternatives, Université Paris-Sud 11, INSERM U1184, 92265 Fontenay-aux-Roses, France
| | - Pierre Rosenbaum
- Immunology of Viral Infections and Autoimmune Diseases, Infectious Disease Models and Innovative Therapies Department, Commissariat à l'Energie Atomique et aux Energies Alternatives, Université Paris-Sud 11, INSERM U1184, 92265 Fontenay-aux-Roses, France
| | - Candie Joly
- Immunology of Viral Infections and Autoimmune Diseases, Infectious Disease Models and Innovative Therapies Department, Commissariat à l'Energie Atomique et aux Energies Alternatives, Université Paris-Sud 11, INSERM U1184, 92265 Fontenay-aux-Roses, France
| | - Candice Poux
- Immunology of Viral Infections and Autoimmune Diseases, Infectious Disease Models and Innovative Therapies Department, Commissariat à l'Energie Atomique et aux Energies Alternatives, Université Paris-Sud 11, INSERM U1184, 92265 Fontenay-aux-Roses, France
| | - Catherine Chapon
- Immunology of Viral Infections and Autoimmune Diseases, Infectious Disease Models and Innovative Therapies Department, Commissariat à l'Energie Atomique et aux Energies Alternatives, Université Paris-Sud 11, INSERM U1184, 92265 Fontenay-aux-Roses, France
| | - Anna-Lena Spetz
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden; and
| | - Mart Ustav
- Institute of Technology, University of Tartu, 50411 Tartu, Estonia
| | - Roger Le Grand
- Immunology of Viral Infections and Autoimmune Diseases, Infectious Disease Models and Innovative Therapies Department, Commissariat à l'Energie Atomique et aux Energies Alternatives, Université Paris-Sud 11, INSERM U1184, 92265 Fontenay-aux-Roses, France
| | - Frédéric Martinon
- Immunology of Viral Infections and Autoimmune Diseases, Infectious Disease Models and Innovative Therapies Department, Commissariat à l'Energie Atomique et aux Energies Alternatives, Université Paris-Sud 11, INSERM U1184, 92265 Fontenay-aux-Roses, France;
| |
Collapse
|
10
|
Inhibition of antigen-specific immune responses by co-application of an indoleamine 2,3-dioxygenase (IDO)-encoding vector requires antigen transgene expression focused on dendritic cells. Amino Acids 2020; 52:411-424. [PMID: 32008091 DOI: 10.1007/s00726-020-02817-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 01/10/2020] [Indexed: 12/17/2022]
Abstract
We have previously shown that particle-mediated epidermal delivery (PMED) of plasmids encoding β-galactosidase (βGal) under control of the fascin-1 promoter (pFascin-βGal) yielded selective production of the protein in skin dendritic cells (DCs), and suppressed Th2 responses in a mouse model of type I allergy by inducing Th1/Tc1 cells. However, intranasal challenge of mice immunized with pFascin-βGal induced airway hyperreactivity (AHR) and neutrophilic inflammation in the lung. The tryptophan-catabolizing enzyme indoleamine 2,3-dioxygenase (IDO) has been implicated in immune suppression and tolerance induction. Here we investigated the consequences of co-application of an IDO-encoding vector on the modulatory effect of DNA vaccination by PMED using pFascin-βGal in models of eosinophilic allergic and non-eosinophilic intrinsic airway inflammation. IDO-encoding plasmids and pFascin-βGal or pCMV-βGal were co-applied to abdominal skin of BALB/c mice without, before or after sensitization with βGal protein. Immune responses in the lung were analysed after intranasal provocation and airway reactivity was determined by whole body plethysmography. Co-application of pCMV-IDO with pFascin-βGal, but not pCMV-βGal inhibited the Th1/Tc1 immune response after PMED. Moreover, AHR in those mice was attenuated following intranasal challenge. Therapeutic vaccination of βGal-sensitized mice with pFascin-βGal plus pCMV-IDO slightly suppressed airway inflammation and AHR after provocation with βGal protein, while prophylactic vaccination was not effective. Altogether, our data suggest that only the combination of DC-restricted antigen and ubiquitous IDO expression attenuated asthma responses in mice, most probably by forming a tryptophan-depleted and kynurenine-enriched micromilieu known to affect neutrophils and T cells.
Collapse
|
11
|
New Rabies Vaccines for Use in Humans. Vaccines (Basel) 2019; 7:vaccines7020054. [PMID: 31226750 PMCID: PMC6631309 DOI: 10.3390/vaccines7020054] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 06/17/2019] [Accepted: 06/19/2019] [Indexed: 12/11/2022] Open
Abstract
Although vaccines are available, rabies still claims more than 55,000 human lives each year. In most cases, rabies vaccines are given to humans after their exposure to a rabid animal; pre-exposure vaccination is largely reserved for humans at high risk for contacts with the virus. Most cases of human rabies are transmitted by dogs. Dog rabies control by mass canine vaccination campaigns combined with intensive surveillance programs has led to a decline of human rabies in many countries but has been unsuccessful in others. Animal vaccination programs are also not suited to control human rabies caused by bat transmission, which is common in some Central American countries. Alternatively, or in addition, more widespread pre-exposure vaccination, especially in highly endemic remote areas, could be implemented. With the multiple dose regimens of current vaccines, pre-exposure vaccination is not cost effective for most countries and this warrants the development of new rabies vaccines, which are as safe as current vaccines, but achieve protective immunity after a single dose, and most importantly, are less costly. This chapter discusses novel rabies vaccines that are in late stage pre-clinical testing or have undergone clinical testing and their potential for replacing current vaccines.
Collapse
|
12
|
Abstract
Neuroblastoma (NB) is a common and deadly malignancy mostly observed in children. Evolution of therapeutic options for NB led to the addition of immunotherapeutic modalities to the previously recruited chemotherapeutic options. Molecular studies of the NB cells resulted in the discovery of many tumor-associated genes and antigens such as MYCN gene and GD2. MYCN gene and GD2 surface antigen are two of the most practical discoveries regarding immunotherapy of neuroblastoma. The GD2 antigen has been targeted in many animal and human studies including Phase III clinical trials. Even though these antigens have changed the face of pediatric neuroblastoma, they do not take as much credit in immunotherapy of adult-onset neuroblastoma. Monoclonal antibodies have been designed to detect this antigen on the surface of NB tumor cells. Despite bettering the outcomes for NB patients, current therapies still fail in many cases. Studies are underway to discover more specific tumor-associated antigens and more effective treatment options. In the current narrative, immunotherapy of NB - from emerging of this therapeutic backbone in NB to the latest discoveries regarding this malignancy - has been reviewed.
Collapse
Affiliation(s)
- Parnian Jabbari
- Research Center for Immunodeficiencies (RCID), Children's Medical Center, Tehran University of Medical Sciences (TUMS), Tehran, Iran.,Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Sara Hanaei
- Research Center for Immunodeficiencies (RCID), Children's Medical Center, Tehran University of Medical Sciences (TUMS), Tehran, Iran.,Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies (RCID), Children's Medical Center, Tehran University of Medical Sciences (TUMS), Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran.,Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| |
Collapse
|
13
|
Hobernik D, Bros M. DNA Vaccines-How Far From Clinical Use? Int J Mol Sci 2018; 19:ijms19113605. [PMID: 30445702 PMCID: PMC6274812 DOI: 10.3390/ijms19113605] [Citation(s) in RCA: 301] [Impact Index Per Article: 50.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/07/2018] [Accepted: 11/09/2018] [Indexed: 12/12/2022] Open
Abstract
Two decades ago successful transfection of antigen presenting cells (APC) in vivo was demonstrated which resulted in the induction of primary adaptive immune responses. Due to the good biocompatibility of plasmid DNA, their cost-efficient production and long shelf life, many researchers aimed to develop DNA vaccine-based immunotherapeutic strategies for treatment of infections and cancer, but also autoimmune diseases and allergies. This review aims to summarize our current knowledge on the course of action of DNA vaccines, and which factors are responsible for the poor immunogenicity in human so far. Important optimization steps that improve DNA transfection efficiency comprise the introduction of DNA-complexing nano-carriers aimed to prevent extracellular DNA degradation, enabling APC targeting, and enhanced endo/lysosomal escape of DNA. Attachment of virus-derived nuclear localization sequences facilitates nuclear entry of DNA. Improvements in DNA vaccine design include the use of APC-specific promotors for transcriptional targeting, the arrangement of multiple antigen sequences, the co-delivery of molecular adjuvants to prevent tolerance induction, and strategies to circumvent potential inhibitory effects of the vector backbone. Successful clinical use of DNA vaccines may require combined employment of all of these parameters, and combination treatment with additional drugs.
Collapse
Affiliation(s)
- Dominika Hobernik
- Department of Dermatology, University Medical Center, 55131 Mainz, Germany.
| | - Matthias Bros
- Department of Dermatology, University Medical Center, 55131 Mainz, Germany.
| |
Collapse
|
14
|
Sadati SF, Jamali A, Abdoli A, Abedi-Valugerdi M, Gholami S, Alipour S, Soleymani S, Kheiri MT, Atyabi F. Simultaneous formulation of influenza vaccine and chitosan nanoparticles within CpG oligodesoxi nucleotides leads to dose-sparing and protects against lethal challenge in the mouse model. Pathog Dis 2018; 76:5089974. [PMID: 30184220 DOI: 10.1093/femspd/fty070] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 09/01/2018] [Indexed: 12/22/2022] Open
Abstract
Lack of efficient delivery systems for transporting antigenic molecules to the cytosol of antigen-presenting cells presents a major obstacle for antigen uptake by immune cells. To this end, influenza whole inactivated virus vaccines were formulated with chitosan nanoparticles and CpG oligonucleotide as a biodegradable delivery system and a Th1-specific adjuvant, respectively. Intradermal injections of a single high dose and low dose of formulated candidate vaccines were carried out. Thirty days after injection, cell proliferation assay (MTT), IFN-gamma and IL-4 ELISpot assays were conducted. Sera samples were collected 21 days after immunization to measure IgG1 and IgG2a levels. In addition, the mice challenged with mouse-adopted virus were monitored for weight loss. The results show a significant stimulation of both humoral and cellular immunities; also, weight gain and a decrease in mortality in the mice receiving both dosages of inactivated influenza virus vaccines with CpG and Chitosan coating were observed. Based on the results, it can be concluded that formulation of inactivated influenza virus with CpG and its delivery by chitosan as low-dose can return the same results as with high-dose balanced between cellular and humeral immune responses. This formulation could potentially lead to a significant saving in vaccine production.
Collapse
Affiliation(s)
- Seyed Farid Sadati
- Department of Medical Microbiology, Ondokuz Mayis University Medical School, Samsun, Turkey.,Amasya University Research Laboratory Center, Ipekkoy Campus, Amasya, Turkey
| | - Abbas Jamali
- Influenza Unit, Pasteur Institute of Iran, Tehran, Iran
| | - Asghar Abdoli
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | | | - Shima Gholami
- Department of Pharmaceutical Biotechnology, Pasteur Institute of Iran, Tehran, Iran
| | - Samira Alipour
- Department of Biological Sciences, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan 45195-1159, Iran
| | - Sepehr Soleymani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | | | - Fatemeh Atyabi
- Department of Pharmaceutical Nanotechnology, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Nicoli F, Paul S, Appay V. Harnessing the Induction of CD8 + T-Cell Responses Through Metabolic Regulation by Pathogen-Recognition-Receptor Triggering in Antigen Presenting Cells. Front Immunol 2018; 9:2372. [PMID: 30410483 PMCID: PMC6209652 DOI: 10.3389/fimmu.2018.02372] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 09/24/2018] [Indexed: 01/03/2023] Open
Abstract
Cytotoxic CD8+ T-cells are key players of the immune responses against viruses. During the priming of a CD8+ T-cell response, the activation of a naïve T-cell by a professional antigen presenting cell (APC) involves the induction of various intracellular and metabolic pathways. The modulation of these pathways at the level of APCs or T-cells offers great potential to enhance the induction of robust effector cells and the generation of long-lived memory cells. On the one hand, signaling through pathogen recognition receptors (PRRs) expressed by APCs can greatly influence T-cell priming, and the potential of several PRR ligands as adjuvants are being studied. On the other hand, the engagement of several metabolic processes, at play in APCs and T-cells upon stimulation, implies that modulating cellular metabolism can impact on priming efficacy. Here, we review recent efforts to understand the interplay between PRR mediated signaling and metabolic pathway modulation in this context, through three examples: interplay between TLR4 and fatty acid metabolism, between TLR9 and IDO, and between STING and autophagy. These initial works highlight the potential for harnessing the induction of antiviral CD8+ T-cell responses using synergistic modulation of metabolic and PRR pathways.
Collapse
Affiliation(s)
- Francesco Nicoli
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Stéphane Paul
- GIMAP/EA3064, Université de Lyon, CIC 1408 Vaccinology, Saint-Etienne, France
| | - Victor Appay
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses, Paris, France.,International Research Center of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
16
|
Allen A, Wang C, Caproni LJ, Sugiyarto G, Harden E, Douglas LR, Duriez PJ, Karbowniczek K, Extance J, Rothwell PJ, Orefo I, Tite JP, Stevenson FK, Ottensmeier CH, Savelyeva N. Linear doggybone DNA vaccine induces similar immunological responses to conventional plasmid DNA independently of immune recognition by TLR9 in a pre-clinical model. Cancer Immunol Immunother 2018; 67:627-638. [PMID: 29330557 PMCID: PMC5860099 DOI: 10.1007/s00262-017-2111-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 12/20/2017] [Indexed: 12/29/2022]
Abstract
Vaccination with DNA that encodes cancer antigens is a simple and convenient way to raise immunity against cancer and has already shown promise in the clinical setting. Conventional plasmid DNA is commonly used which together with the encoded antigen also includes bacterial immunostimulatory CpG motifs to target the DNA sensor Toll-like receptor 9. Recently DNA vaccines using doggybone DNA (dbDNA™), have been developed without the use of bacteria. The cell-free process relies on the use of Phi29 DNA polymerase to amplify the template followed by protelomerase TelN to complete individual closed linear DNA. The resulting DNA contains the required antigenic sequence, a promoter and a poly A tail but lacks bacterial sequences such as an antibiotic resistance gene, prompting the question of immunogenicity. Here we compared the ability of doggybone DNA vaccine with plasmid DNA vaccine to induce adaptive immunity using clinically relevant oncotargets E6 and E7 from HPV. We demonstrate that despite the inability to trigger TLR9, doggybone DNA was able to induce similar levels of cellular and humoral immunity as plasmid DNA, with suppression of established TC-1 tumours.
Collapse
Affiliation(s)
- Alex Allen
- Cancer Sciences Unit and Cancer Research UK and Experimental Cancer Medicine Centre Protein Core Facility, Faculty of Medicine, University of Southampton, Tremona road, Southampton, SO16 6YD, UK
| | - Chuan Wang
- Cancer Sciences Unit and Cancer Research UK and Experimental Cancer Medicine Centre Protein Core Facility, Faculty of Medicine, University of Southampton, Tremona road, Southampton, SO16 6YD, UK
| | - Lisa J Caproni
- Touchlight Genetics Ltd, Morelands and Riverdale Buildings, Lower Sunbury Road, Hampton, London, TW12 2ER, UK
| | - Gessa Sugiyarto
- Cancer Sciences Unit and Cancer Research UK and Experimental Cancer Medicine Centre Protein Core Facility, Faculty of Medicine, University of Southampton, Tremona road, Southampton, SO16 6YD, UK
| | - Elena Harden
- Cancer Sciences Unit and Cancer Research UK and Experimental Cancer Medicine Centre Protein Core Facility, Faculty of Medicine, University of Southampton, Tremona road, Southampton, SO16 6YD, UK
| | - Leon R Douglas
- Cancer Sciences Unit and Cancer Research UK and Experimental Cancer Medicine Centre Protein Core Facility, Faculty of Medicine, University of Southampton, Tremona road, Southampton, SO16 6YD, UK
| | - Patrick J Duriez
- Cancer Sciences Unit and Cancer Research UK and Experimental Cancer Medicine Centre Protein Core Facility, Faculty of Medicine, University of Southampton, Tremona road, Southampton, SO16 6YD, UK
| | - Kinga Karbowniczek
- Touchlight Genetics Ltd, Morelands and Riverdale Buildings, Lower Sunbury Road, Hampton, London, TW12 2ER, UK
| | - Jon Extance
- Touchlight Genetics Ltd, Morelands and Riverdale Buildings, Lower Sunbury Road, Hampton, London, TW12 2ER, UK
| | - Paul J Rothwell
- Touchlight Genetics Ltd, Morelands and Riverdale Buildings, Lower Sunbury Road, Hampton, London, TW12 2ER, UK
| | - Ifeayinwa Orefo
- Touchlight Genetics Ltd, Morelands and Riverdale Buildings, Lower Sunbury Road, Hampton, London, TW12 2ER, UK
| | - John P Tite
- Touchlight Genetics Ltd, Morelands and Riverdale Buildings, Lower Sunbury Road, Hampton, London, TW12 2ER, UK
| | - Freda K Stevenson
- Cancer Sciences Unit and Cancer Research UK and Experimental Cancer Medicine Centre Protein Core Facility, Faculty of Medicine, University of Southampton, Tremona road, Southampton, SO16 6YD, UK
| | - Christian H Ottensmeier
- Cancer Sciences Unit and Cancer Research UK and Experimental Cancer Medicine Centre Protein Core Facility, Faculty of Medicine, University of Southampton, Tremona road, Southampton, SO16 6YD, UK
| | - Natalia Savelyeva
- Cancer Sciences Unit and Cancer Research UK and Experimental Cancer Medicine Centre Protein Core Facility, Faculty of Medicine, University of Southampton, Tremona road, Southampton, SO16 6YD, UK.
| |
Collapse
|
17
|
Teixeira D, Ishimura ME, Apostólico JDS, Viel JM, Passarelli VC, Cunha-Neto E, Rosa DS, Longo-Maugéri IM. Propionibacterium acnes Enhances the Immunogenicity of HIVBr18 Human Immunodeficiency Virus-1 Vaccine. Front Immunol 2018; 9:177. [PMID: 29467764 PMCID: PMC5808300 DOI: 10.3389/fimmu.2018.00177] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 01/19/2018] [Indexed: 02/01/2023] Open
Abstract
Immunization of BALB/c mice with HIVBr18, a DNA vaccine containing 18 CD4+ T cell epitopes from human immunodeficiency virus (HIV), induced specific CD4+ and CD8+ T cell responses in a broad, polyfunctional and persistent manner. With the aim of increasing the immunogenicity of this vaccine, the effect of Propionibacterium acnes as an adjuvant was evaluated. The adjuvant effects of this bacterium have been extensively demonstrated in both experimental and clinical settings. Herein, administration of two doses of HIVBr18, in the presence of P. acnes, increased the proliferation of HIV-1-specific CD4+ and CD8+ T lymphocytes, the polyfunctional profile of CD4+ T cells, the production of IFN-γ, and the number of recognized vaccine-encoded peptides. One of the bacterial components responsible for most of the adjuvant effects observed was a soluble polysaccharide extracted from the P. acnes cell wall. Furthermore, within 10 weeks after immunization, the proliferation of specific T cells and production of IFN-γ were maintained when the whole bacterium was administered, demonstrating a greater effect on the longevity of the immune response by P. acnes. Even with fewer immunization doses, P. acnes was found to be a potent adjuvant capable of potentiating the effects of the HIVBr18 vaccine. Therefore, P. acnes may be a potential adjuvant to aid this vaccine in inducing immunity or for therapeutic use.
Collapse
Affiliation(s)
- Daniela Teixeira
- Division of Immunology, Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil
| | - Mayari Eika Ishimura
- Division of Immunology, Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil
| | - Juliana de Souza Apostólico
- Division of Immunology, Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil
| | - Jacqueline Miyuki Viel
- Division of Immunology, Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil
| | - Victor Cabelho Passarelli
- Division of Immunology, Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil
| | - Edecio Cunha-Neto
- Laboratory of Clinical Immunology and Allergy-LIM60, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Daniela Santoro Rosa
- Division of Immunology, Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil
| | - Ieda Maria Longo-Maugéri
- Division of Immunology, Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
18
|
Su H, Liao Z, Yuan G, Su J. A plasmid containing CpG ODN as vaccine adjuvant against grass carp reovirus in grass carp Ctenopharyngodon idella. Oncotarget 2017; 8:86576-86591. [PMID: 29156818 PMCID: PMC5689708 DOI: 10.18632/oncotarget.21245] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 08/17/2017] [Indexed: 02/07/2023] Open
Abstract
CpG oligodeoxynucleotides (ODNs) are proved to have strong immune stimulatory activity. Plasmids containing CpG ODNs could be conveniently and low-costly used as vaccine adjuvant. However, they are different among various plasmids, motif repeats, species, etc. In the present study, plasmid pcDNA3.1 (+) containing five repetitions of CpG ODN 1670A named pcDNA3.1-1670A*5 with strong immunostimulation was screened out from twelve recombinant plasmids and three empty vectors by cell proliferation activity, interferon promoter activities and immune related gene expressions in CIK cells. It works through TLR9-mediated signaling pathway, triggering the immune related genes expression. Furthermore, the potentiality of pcDNA3.1-1670A*5 as adjuvant was tested in vivo. pcDNA3.1-1670A*5 was co-inoculated with inactivated GCRV vaccine on grass carp fingerlings. Immunoglobulins (IgM, IgD, IgZ), TLR9, IFNγ2, IFN1, TNF-α, Mx2 and VP4 were examined. Ultimately, pcDNA3.1-1670A*5 significantly enhanced the expressions of IgM in serum, head kidney and spleen, recognition receptor TLR9 as well as antiviral effector molecule Mx2, and inhibited GCRV proliferation in head kidney and spleen tissues. The present study explored the application and mechanism of plasmid containing CpG ODN as high-efficient adjuvant to promote efficiency of vaccine and control disease in grass carp, which will contribute to the development of new type CpG ODN adjuvant in aquaculture industry.
Collapse
Affiliation(s)
- Hang Su
- College of Fisheries, Huazhong Agricultural University, Wuhan, China.,Hubei Engineering Technology Research Center for Aquatic Animal Disease Control and Prevention, Wuhan, China
| | - Zhiwei Liao
- College of Fisheries, Huazhong Agricultural University, Wuhan, China.,Hubei Engineering Technology Research Center for Aquatic Animal Disease Control and Prevention, Wuhan, China
| | - Gailing Yuan
- College of Fisheries, Huazhong Agricultural University, Wuhan, China.,Hubei Engineering Technology Research Center for Aquatic Animal Disease Control and Prevention, Wuhan, China
| | - Jianguo Su
- College of Fisheries, Huazhong Agricultural University, Wuhan, China.,Hubei Engineering Technology Research Center for Aquatic Animal Disease Control and Prevention, Wuhan, China.,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| |
Collapse
|
19
|
Ilg T. Investigations on the molecular mode of action of the novel immunostimulator ZelNate: Activation of the cGAS-STING pathway in mammalian cells. Mol Immunol 2017; 90:182-189. [PMID: 28802127 DOI: 10.1016/j.molimm.2017.07.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 07/18/2017] [Accepted: 07/25/2017] [Indexed: 02/06/2023]
Abstract
Bovine respiratory disease (BRD) is usually prevented or treated with vaccines and/or antibiotics. The use of antibiotics is, however, of concern due to the potential promotion of microbial resistance and the occurrence of residues. Recently an alternative aid in the treatment of BRD, the cationic lipid/bacterial plasmid DNA liposome-based immunomodulator ZelNate, has entered the veterinary market. In the present study, we provide data on the molecular mode of action of ZelNate. Despite the presence of numerous non-methylated CpG motifs in its plasmid DNA, ZelNate proved to be inactive on human and mouse toll-like receptor 9 (TLR9) in cell culture, in both recombinant and natural cellular receptor settings. However, in the human monocyte cell line THP1 and in the mouse melanoma cell line B16, ZelNate activates strongly the stimulator of interferon genes (STING) pathway, which is known to lead predominantly to interferon response factor 3 (IRF3) activation. Further analysis in THP1 cells suggests that the ZelNate plasmid DNA activates STING via interaction with cyclic guanylate adenylate synthase (cGAS), but not via interferon induced gene 16 (IFI16). Our in vitro observations suggest that ZelNate may act predominantly via the cGAS/STING/IRF3 pathway.
Collapse
Affiliation(s)
- Thomas Ilg
- Bayer Animal Health GmbH, Alfred-Nobel-Strasse 50, 40789 Monheim, Germany.
| |
Collapse
|
20
|
Ma Z, Liu J, Wu W, Zhang E, Zhang X, Li Q, Zelinskyy G, Buer J, Dittmer U, Kirschning CJ, Lu M. The IL-1R/TLR signaling pathway is essential for efficient CD8 + T-cell responses against hepatitis B virus in the hydrodynamic injection mouse model. Cell Mol Immunol 2017; 14:997-1008. [PMID: 28757610 PMCID: PMC5719144 DOI: 10.1038/cmi.2017.43] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 05/13/2017] [Accepted: 05/13/2017] [Indexed: 12/18/2022] Open
Abstract
The outcome of hepatitis B viral (HBV) infection is determined by the complex interactions between replicating HBV and the immune system. While the role of the adaptive immune system in the resolution of HBV infection has been studied extensively, the contribution of innate immune mechanisms remains to be defined. Here we examined the role of the interleukin-1 receptor/Toll-like receptor (IL-1R/TLR) signaling pathway in adaptive immune responses and viral clearance by exploring the HBV mouse model. Hydrodynamic injection with a replication-competent HBV genome was performed in wild-type mice (WT) and a panel of mouse strains lacking specific innate immunity component expression. We found higher levels of HBV protein production and replication in Tlr2−/−, Tlr23479−/−, 3d/Tlr24−/−, Myd88/Trif−/− and Irak4−/− mice, which was associated with reduced HBV-specific CD8+ T-cell responses in these mice. Importantly, HBV clearance was delayed for more than 2 weeks in 3d/Tlr24−/−, Myd88/Trif−/− and Irak4−/− mice compared to WT mice. HBV-specific CD8+ T-cell responses were functionally impaired for producing the cytokines IFN-γ, TNF-α and IL-2 in TLR signaling-deficient mice compared to WT mice. In conclusion, the IL-1R/TLR signaling pathway might contribute to controlling HBV infection by augmenting HBV-specific CD8+ T-cell responses.
Collapse
Affiliation(s)
- Zhiyong Ma
- Institute of Virology, University Hospital of Essen, University of Duisburg-Essen, 45122 Essen, Germany
| | - Jia Liu
- Institute of Virology, University Hospital of Essen, University of Duisburg-Essen, 45122 Essen, Germany
| | - Weimin Wu
- Institute of Virology, University Hospital of Essen, University of Duisburg-Essen, 45122 Essen, Germany
| | - Ejuan Zhang
- Institute of Virology, University Hospital of Essen, University of Duisburg-Essen, 45122 Essen, Germany
| | - Xiaoyong Zhang
- Institute of Virology, University Hospital of Essen, University of Duisburg-Essen, 45122 Essen, Germany
| | - Qian Li
- Institute of Virology, University Hospital of Essen, University of Duisburg-Essen, 45122 Essen, Germany
| | - Gennadiy Zelinskyy
- Institute of Virology, University Hospital of Essen, University of Duisburg-Essen, 45122 Essen, Germany
| | - Jan Buer
- Institute of Medical Microbiology, University Hospital of Essen, University of Duisburg-Essen, 45122 Essen, Germany
| | - Ulf Dittmer
- Institute of Virology, University Hospital of Essen, University of Duisburg-Essen, 45122 Essen, Germany
| | - Carsten J Kirschning
- Institute of Medical Microbiology, University Hospital of Essen, University of Duisburg-Essen, 45122 Essen, Germany
| | - Mengji Lu
- Institute of Virology, University Hospital of Essen, University of Duisburg-Essen, 45122 Essen, Germany
| |
Collapse
|
21
|
Abstract
In the two decades since their initial discovery, DNA vaccines technologies have come a long way. Unfortunately, when applied to human subjects inadequate immunogenicity is still the biggest challenge for practical DNA vaccine use. Many different strategies have been tested in preclinical models to address this problem, including novel plasmid vectors and codon optimization to enhance antigen expression, new gene transfection systems or electroporation to increase delivery efficiency, protein or live virus vector boosting regimens to maximise immune stimulation, and formulation of DNA vaccines with traditional or molecular adjuvants. Better understanding of the mechanisms of action of DNA vaccines has also enabled better use of the intrinsic host response to DNA to improve vaccine immunogenicity. This review summarizes recent advances in DNA vaccine technologies and related intracellular events and how these might impact on future directions of DNA vaccine development.
Collapse
Affiliation(s)
- Lei Li
- a Vaxine Pty Ltd, Bedford Park , Adelaide , Australia.,b Department of Diabetes and Endocrinology , Flinders University, Flinders Medical Centre , Adelaide , SA , Australia
| | - Nikolai Petrovsky
- a Vaxine Pty Ltd, Bedford Park , Adelaide , Australia.,b Department of Diabetes and Endocrinology , Flinders University, Flinders Medical Centre , Adelaide , SA , Australia
| |
Collapse
|
22
|
Lambricht L, Lopes A, Kos S, Sersa G, Préat V, Vandermeulen G. Clinical potential of electroporation for gene therapy and DNA vaccine delivery. Expert Opin Drug Deliv 2015; 13:295-310. [PMID: 26578324 DOI: 10.1517/17425247.2016.1121990] [Citation(s) in RCA: 152] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Electroporation allows efficient delivery of DNA into cells and tissues, thereby improving the expression of therapeutic or immunogenic proteins that are encoded by plasmid DNA. This simple and versatile method holds a great potential and could address unmet medical needs such as the prevention or treatment of many cancers or infectious diseases. AREAS COVERED This review explores the electroporation mechanism and the parameters affecting its efficacy. An analysis of past and current clinical trials focused on DNA electroporation is presented. The pathologies addressed, the protocol used, the treatment outcome and the tolerability are highlighted. In addition, several of the possible optimization strategies for improving patient compliance and therapeutic efficacy are discussed such as plasmid design, use of genetic adjuvants for DNA vaccines, choice of appropriate delivery site and electrodes as well as pulse parameters. EXPERT OPINION The growing number of clinical trials and the results already available underline the strong potential of DNA electroporation which combines both safety and efficiency. Nevertheless, it remains critical to further increase fundamental knowledge to refine future strategies, to develop concerted and common DNA electroporation protocols and to continue exploring new electroporation-based therapeutic options.
Collapse
Affiliation(s)
- Laure Lambricht
- a Université catholique de Louvain, Louvain Drug Research Institute , Advanced Drug Delivery and Biomaterials , Brussels , Belgium
| | - Alessandra Lopes
- a Université catholique de Louvain, Louvain Drug Research Institute , Advanced Drug Delivery and Biomaterials , Brussels , Belgium
| | - Spela Kos
- b Institute of Oncology Ljubljana , Department of Experimental Oncology , Ljubljana , Slovenia
| | - Gregor Sersa
- b Institute of Oncology Ljubljana , Department of Experimental Oncology , Ljubljana , Slovenia
| | - Véronique Préat
- a Université catholique de Louvain, Louvain Drug Research Institute , Advanced Drug Delivery and Biomaterials , Brussels , Belgium
| | - Gaëlle Vandermeulen
- a Université catholique de Louvain, Louvain Drug Research Institute , Advanced Drug Delivery and Biomaterials , Brussels , Belgium
| |
Collapse
|
23
|
Fontoura IC, Trombone A, Almeida LP, Lorenzi JCC, Rossetti RAM, Malardo T, Padilha E, Schluchting W, Silva RLL, Gembre AF, Fiuza JEC, Silva CL, Panunto-Castelo A, Coelho-Castelo AAM. B cells expressing IL-10 mRNA modulate memory T cells after DNA-Hsp65 immunization. Braz J Med Biol Res 2015; 48:1095-100. [PMID: 26397973 PMCID: PMC4661025 DOI: 10.1590/1414-431x20154409] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Accepted: 06/30/2015] [Indexed: 11/26/2022] Open
Abstract
In DNA vaccines, the gene of interest is cloned into a bacterial plasmid that is engineered to induce protein production for long periods in eukaryotic cells. Previous research has shown that the intramuscular immunization of BALB/c mice with a naked plasmid DNA fragment encoding the Mycobacterium leprae 65-kDa heat-shock protein (pcDNA3-Hsp65) induces protection against M. tuberculosis challenge. A key stage in the protective immune response after immunization is the generation of memory T cells. Previously, we have shown that B cells capture plasmid DNA-Hsp65 and thereby modulate the formation of CD8+ memory T cells after M. tuberculosis challenge in mice. Therefore, clarifying how B cells act as part of the protective immune response after DNA immunization is important for the development of more-effective vaccines. The aim of this study was to investigate the mechanisms by which B cells modulate memory T cells after DNA-Hsp65 immunization. C57BL/6 and BKO mice were injected three times, at 15-day intervals, with 100 µg naked pcDNA-Hsp65 per mouse. Thirty days after immunization, the percentages of effector memory T (TEM) cells (CD4+ and CD8+/CD44high/CD62Llow) and memory CD8+ T cells (CD8+/CD44high/CD62Llow/CD127+) were measured with flow cytometry. Interferon γ, interleukin 12 (IL-12), and IL-10 mRNAs were also quantified in whole spleen cells and purified B cells (CD43-) with real-time qPCR. Our data suggest that a B-cell subpopulation expressing IL-10 downregulated proinflammatory cytokine expression in the spleen, increasing the survival of CD4+ TEM cells and CD8+ TEM/CD127+ cells.
Collapse
Affiliation(s)
- I. C. Fontoura
- Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo,
Ribeirão Preto, SP, Brasil
| | | | - L. P. Almeida
- Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo,
Ribeirão Preto, SP, Brasil
| | - J. C. C. Lorenzi
- Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo,
Ribeirão Preto, SP, Brasil
| | - R. A. M. Rossetti
- Instituto de Ciências Biomédicas, Universidade de São Paulo, São
Paulo, SP, Brasil
| | - T. Malardo
- Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo,
Ribeirão Preto, SP, Brasil
| | - E. Padilha
- Universidade Paranaense, Cascavel, PR,
Brasil
| | - W. Schluchting
- Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo,
Ribeirão Preto, SP, Brasil
| | - R. L. L. Silva
- Departamento de Educação em Saúde, Universidade Federal de Sergipe,
Lagarto, SE, Brasil
| | - A. F. Gembre
- Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo,
Ribeirão Preto, SP, Brasil
| | - J. E. C. Fiuza
- Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo,
Ribeirão Preto, SP, Brasil
| | - C. L. Silva
- Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo,
Ribeirão Preto, SP, Brasil
| | - A. Panunto-Castelo
- Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto,
Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - A. A. M. Coelho-Castelo
- Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo,
Ribeirão Preto, SP, Brasil
| |
Collapse
|
24
|
Suschak JJ, Wang S, Fitzgerald KA, Lu S. A cGAS-Independent STING/IRF7 Pathway Mediates the Immunogenicity of DNA Vaccines. THE JOURNAL OF IMMUNOLOGY 2015; 196:310-6. [PMID: 26590319 DOI: 10.4049/jimmunol.1501836] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 10/29/2015] [Indexed: 11/19/2022]
Abstract
It has been known since the discovery of DNA vaccines >20 y ago that DNA vaccines can function as adjuvants. Our recent study reported the involvement of Aim2 as the sensor of DNA vaccines in eliciting Ag-specific Ab responses. Our findings indicated the presence of previously unrecognized innate immune response pathways in addition to the TLR9 pathway, which is mainly activated by the CpG motifs of DNA vaccines. Our data further demonstrated the requirement of type I IFN in DNA vaccine-induced immune responses via the Aim2 pathway, but the exact downstream molecular mechanism was not characterized. In the present study, we investigated the roles of the putative DNA sensor cyclic GMP-AMP synthase (cGas), as well as the downstream IFN regulatory factors (IRF) 3 and 7 in type I IFN induction and Ag-specific immune responses elicited by DNA vaccination. Our results showed that DNA vaccine-induced, Irf7-dependent signaling, as part of the Sting pathway, was critical for generation of both innate cytokine signaling and Ag-specific B and T cell responses. In contrast, Irf3 was not as critical as expected in this pathway and, more surprisingly, immune responses elicited by DNA vaccines were not cGas-dependent in vivo. Data from this study provide more details on the innate immune mechanisms involved in DNA vaccination and further enrich our understanding on the potential utility of DNA vaccines in generating Ag-specific immune responses.
Collapse
Affiliation(s)
- John J Suschak
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655; and
| | - Shixia Wang
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655; and
| | - Katherine A Fitzgerald
- Program in Innate Immunity, Division of Infectious Diseases, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655
| | - Shan Lu
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655; and
| |
Collapse
|
25
|
Abstract
Detecting pathogenic DNA by intracellular receptors termed "sensors" is critical toward galvanizing host immune responses and eliminating microbial infections. Emerging evidence has challenged the dogma that sensing of viral DNA occurs exclusively in sub-cellular compartments normally devoid of cellular DNA. The interferon-inducible protein IFI16 was shown to bind nuclear viral DNA and initiate immune signaling, culminating in antiviral cytokine secretion. Here, we review the newly characterized nucleus-originating immune signaling pathways, their links to other crucial host defenses, and unique mechanisms by which viruses suppress their functions. We frame these findings in the context of human pathologies associated with nuclear replicating DNA viruses.
Collapse
Affiliation(s)
- Benjamin A Diner
- From the Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544
| | - Krystal K Lum
- From the Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544
| | - Ileana M Cristea
- From the Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544
| |
Collapse
|
26
|
Stermann A, Huebener N, Seidel D, Fest S, Eschenburg G, Stauder M, Schramm A, Eggert A, Lode HN. Targeting of MYCN by means of DNA vaccination is effective against neuroblastoma in mice. Cancer Immunol Immunother 2015; 64:1215-27. [PMID: 26076666 PMCID: PMC11028418 DOI: 10.1007/s00262-015-1733-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Accepted: 06/04/2015] [Indexed: 01/21/2023]
Abstract
The MYCN oncogene is a strong genetic marker associated with poor prognosis in neuroblastoma (NB). Therefore, MYCN gene amplification and subsequent overexpression provide a possible target for new treatment approaches in NB. We first identified an inverse correlation of MYCN expression with CD45 mRNA in 101 NB tumor samples. KEGG mapping further revealed that MYCN expression was associated with immune-suppressive pathways characterized by a down-regulation of T cell activation and up-regulation of T cell inhibitory gene transcripts. We then aimed to investigate whether DNA vaccination against MYCN is effective to induce an antigen-specific and T cell-mediated immune response. For this purpose, we generated a MYCN-expressing syngeneic mouse model by MYCN gene transfer to NXS2 cells. MYCN-DNA vaccines were engineered based on the pCMV-F3Ub plasmid backbone to drive ubiquitinated full-length MYCN-cDNA and minigene expression. Vaccines were delivered orally with attenuated S. typhimurium strain SL7207 as a carrier. Immunization with both MYCN-DNA vaccines significantly reduced primary tumor growth of MYCN-expressing NB cells in contrast to negative controls. The immune response was mediated by tumor-infiltrating T cells in vivo, which revealed MYCN-specific and MHC class I-restricted lysis of inducible MYCN-expressing NB target cells in vitro. Finally, these antigen-specific T cells also killed MYCN-negative mammary carcinoma cells pulsed with MYCN peptides in contrast to controls. In summary, we demonstrate proof of concept that MYCN can be targeted by DNA vaccination, which may provide an approach to overcoming MYCN immune-suppressive activities in patients with MYCN-amplified disease.
Collapse
MESH Headings
- Administration, Oral
- Animals
- Carcinoma/immunology
- Carcinoma/microbiology
- Cell Line, Tumor
- Cytotoxicity, Immunologic
- Epitopes, B-Lymphocyte/genetics
- Epitopes, B-Lymphocyte/metabolism
- Gene Expression Regulation, Neoplastic
- Humans
- Lymphocytes, Tumor-Infiltrating/immunology
- Mammary Neoplasms, Animal/immunology
- Mammary Neoplasms, Animal/microbiology
- Mice
- Mice, Inbred Strains
- N-Myc Proto-Oncogene Protein
- Neoplasm Transplantation
- Neoplasms, Experimental
- Neuroblastoma/genetics
- Neuroblastoma/immunology
- Neuroblastoma/microbiology
- Peptide Fragments
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- Salmonella Vaccines/administration & dosage
- Salmonella typhimurium/immunology
- Transgenes/genetics
- Tumor Burden
- Vaccination
- Vaccines, Attenuated/administration & dosage
- Vaccines, DNA/administration & dosage
Collapse
Affiliation(s)
- Alexander Stermann
- Pediatric Hematology and Oncology, University Medicine Greifswald, Ferdinand-Sauerbruch-Str 1, 17475 Greifswald, Germany
| | - Nicole Huebener
- Genetics of Metabolic and Reproductive Disorders, Max Delbrück Center for Molecular Medicine Berlin, Berlin, Germany
| | - Diana Seidel
- Pediatric Hematology and Oncology, University Medicine Greifswald, Ferdinand-Sauerbruch-Str 1, 17475 Greifswald, Germany
| | - Stefan Fest
- Department of Pediatrics, Otto-von-Guericke University, Magdeburg, Germany
| | - Georg Eschenburg
- Department and Clinic of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Stauder
- Department of Pediatric Oncology and Hematology, University Children’s Hospital Essen, Essen, Germany
| | - Alexander Schramm
- Department of Pediatric Oncology and Hematology, University Children’s Hospital Essen, Essen, Germany
| | - Angelika Eggert
- Pediatric Oncology and Hematology, Charité University Medicine Berlin, Berlin, Germany
| | - Holger N. Lode
- Pediatric Hematology and Oncology, University Medicine Greifswald, Ferdinand-Sauerbruch-Str 1, 17475 Greifswald, Germany
| |
Collapse
|
27
|
Vandermeulen G, Vanvarenberg K, De Beuckelaer A, De Koker S, Lambricht L, Uyttenhove C, Reschner A, Vanderplasschen A, Grooten J, Préat V. The site of administration influences both the type and the magnitude of the immune response induced by DNA vaccine electroporation. Vaccine 2015; 33:3179-85. [PMID: 25980430 DOI: 10.1016/j.vaccine.2015.05.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 03/10/2015] [Accepted: 05/04/2015] [Indexed: 11/28/2022]
Abstract
We investigated the influence of the site of administration of DNA vaccine on the induced immune response. DNA vaccines were administered by electroporation at three different sites: tibial cranial muscle, abdominal skin and ear pinna. Aiming to draw general conclusions about DNA vaccine delivery, we successively used several plasmids encoding either luciferase and ovalbumin as models or gp160 and P1A as vaccines against HIV and P815 mastocytoma, respectively. Low levels and duration of luciferase transgene expression were observed after electroporation of the abdominal skin, partly explaining its lower immunogenic performance as compared to the other sites of administration. Analyses of OT-I CD8+ and OT-II CD4+ T cell responses highlighted the differential impact of the delivery site on the elicited immune response. Muscle electroporation induced the strongest humoral immune response and both muscle and ear pinna sites induced cellular immunity against gp160. Ear pinna delivery generated the highest level of CTL responses against P1A but electroporation of muscle and ear pinna were equally efficient in delaying P815 growth and improving mice survival. The present study demonstrated that the site of administration is a key factor to be tested in the development of DNA vaccine.
Collapse
Affiliation(s)
- Gaëlle Vandermeulen
- Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Brussels, Belgium
| | - Kevin Vanvarenberg
- Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Brussels, Belgium
| | - Ans De Beuckelaer
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Stefaan De Koker
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Laure Lambricht
- Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Brussels, Belgium
| | - Catherine Uyttenhove
- Université catholique de Louvain, Ludwig Institute for Cancer Research, Brussels Branch and de Duve Institute, Brussels, Belgium
| | - Anca Reschner
- Immunology-Vaccinology, Department of Infectious and Parasitic Diseases, Fundamental and Applied Research for Animals & Health (FARAH), Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Alain Vanderplasschen
- Immunology-Vaccinology, Department of Infectious and Parasitic Diseases, Fundamental and Applied Research for Animals & Health (FARAH), Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Johan Grooten
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Véronique Préat
- Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Brussels, Belgium.
| |
Collapse
|
28
|
Xu Y, Yuen PW, Lam JKW. Intranasal DNA Vaccine for Protection against Respiratory Infectious Diseases: The Delivery Perspectives. Pharmaceutics 2014; 6:378-415. [PMID: 25014738 PMCID: PMC4190526 DOI: 10.3390/pharmaceutics6030378] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 06/20/2014] [Accepted: 06/24/2014] [Indexed: 11/16/2022] Open
Abstract
Intranasal delivery of DNA vaccines has become a popular research area recently. It offers some distinguished advantages over parenteral and other routes of vaccine administration. Nasal mucosa as site of vaccine administration can stimulate respiratory mucosal immunity by interacting with the nasopharyngeal-associated lymphoid tissues (NALT). Different kinds of DNA vaccines are investigated to provide protection against respiratory infectious diseases including tuberculosis, coronavirus, influenza and respiratory syncytial virus (RSV) etc. DNA vaccines have several attractive development potential, such as producing cross-protection towards different virus subtypes, enabling the possibility of mass manufacture in a relatively short time and a better safety profile. The biggest obstacle to DNA vaccines is low immunogenicity. One of the approaches to enhance the efficacy of DNA vaccine is to improve DNA delivery efficiency. This review provides insight on the development of intranasal DNA vaccine for respiratory infections, with special attention paid to the strategies to improve the delivery of DNA vaccines using non-viral delivery agents.
Collapse
Affiliation(s)
- Yingying Xu
- Department of Pharmacology & Pharmacy, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Pokfulam, 21 Sassoon Road, Hong Kong, China.
| | - Pak-Wai Yuen
- Department of Pharmacology & Pharmacy, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Pokfulam, 21 Sassoon Road, Hong Kong, China.
| | - Jenny Ka-Wing Lam
- Department of Pharmacology & Pharmacy, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Pokfulam, 21 Sassoon Road, Hong Kong, China.
| |
Collapse
|
29
|
Comprehensive analysis of contributions from protein conformational stability and major histocompatibility complex class II-peptide binding affinity to CD4+ epitope immunogenicity in HIV-1 envelope glycoprotein. J Virol 2014; 88:9605-15. [PMID: 24920818 DOI: 10.1128/jvi.00789-14] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
UNLABELLED Helper T-cell epitope dominance in human immunodeficiency virus type 1 (HIV-1) envelope glycoprotein gp120 is not adequately explained by peptide binding to major histocompatibility complex (MHC) proteins. Antigen processing potentially influences epitope dominance, but few, if any, studies have attempted to reconcile the influences of antigen processing and MHC protein binding for all helper T-cell epitopes of an antigen. Epitopes of gp120 identified in both humans and mice occur on the C-terminal flanks of flexible segments that are likely to be proteolytic cleavage sites. In this study, the influence of gp120 conformation on the dominance pattern in gp120 from HIV strain 89.6 was examined in CBA mice, whose MHC class II protein has one of the most well defined peptide-binding preferences. Only one of six dominant epitopes contained the most conserved element of the I-Ak binding motif, an aspartic acid. Destabilization of the gp120 conformation by deletion of single disulfide bonds preferentially enhanced responses to the cryptic I-Ak motif-containing sequences, as reported by T-cell proliferation or cytokine secretion. Conversely, inclusion of CpG in the adjuvant with gp120 enhanced responses to the dominant CD4+ T-cell epitopes. The gp120 destabilization affected secretion of some cytokines more than others, suggesting that antigen conformation could modulate T-cell functions through mechanisms of antigen processing. IMPORTANCE CD4+ helper T cells play an essential role in protection against HIV and other pathogens. Thus, the sites of helper T-cell recognition, the dominant epitopes, are targets for vaccine design; and the corresponding T cells may provide markers for monitoring infection and immunity. However, T-cell epitopes are difficult to identify and predict. It is also unclear whether CD4+ T cells specific for one epitope are more protective than T cells specific for other epitopes. This work shows that the three-dimensional (3D) structure of an HIV protein partially determines which epitopes are dominant, most likely by controlling the breakdown of HIV into peptides. Moreover, some types of signals from CD4+ T cells are affected by the HIV protein 3D structure; and thus the protectiveness of a particular peptide vaccine could be related to its location in the 3D structure.
Collapse
|
30
|
Sheng KC, Day S, Wright MD, Stojanovska L, Apostolopoulos V. Enhanced Dendritic Cell-Mediated Antigen-Specific CD4+ T Cell Responses: IFN-Gamma Aids TLR Stimulation. JOURNAL OF DRUG DELIVERY 2013; 2013:516749. [PMID: 23781340 PMCID: PMC3679806 DOI: 10.1155/2013/516749] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 02/18/2013] [Indexed: 12/20/2022]
Abstract
Phenotypic maturation and T cell stimulation are two functional attributes of DCs critical for immune induction. The combination of antigens, including those from cancer, with Toll-like receptor (TLR) ligands induces far superior cellular immune responses compared to antigen alone. In this study, IFN-gamma treatment of bone marrow-derived DC, followed by incubation with the TLR2, TLR4, or TLR9 agonists, enhanced DC activation compared to TLR ligation alone. Most notably, the upregulation of CD40 with LPS stimulation and CD86 with CpG stimulation was observed in in vitro cultures. Similarly, IFN-gamma coinjected with TLR ligands was able to promote DC activation in vivo, with DCs migrating from the site of immunization to the popliteal lymph nodes demonstrating increased expression of CD80 and CD86. The heightened DC activation translated to a drastic increase in T cell stimulatory capacity in both antigen independent and antigen dependent fashions. This is the first time that IFN-gamma has been shown to have a combined effect with TLR ligation to enhance DC activation and function. The results demonstrate the novel use of IFN-gamma together with TLR agonists to enhance antigen-specific T cell responses, for applications in the development of enhanced vaccines and drug targets against diseases including cancer.
Collapse
Affiliation(s)
- Kuo-Ching Sheng
- Immunology and Vaccine Laboratory, Burnet Institute, Melbourne, VIC 3004, Australia
- Institute for Glycomics, Griffith University, Gold Coast, QLD 4215, Australia
| | - Stephanie Day
- Immunology and Vaccine Laboratory, Burnet Institute, Melbourne, VIC 3004, Australia
- Department of Immunology, Monash University, Melbourne, VIC 3004, Australia
| | - Mark D. Wright
- Department of Immunology, Monash University, Melbourne, VIC 3004, Australia
| | - Lily Stojanovska
- College of Health and Biomedicine, Victoria University, VIC 3021, Australia
| | - Vasso Apostolopoulos
- Immunology and Vaccine Laboratory, Burnet Institute, Melbourne, VIC 3004, Australia
- VA Consulting Services, Melbourne, VIC 3030, Australia
| |
Collapse
|
31
|
Berger E, Soldati R, Huebener N, Hohn O, Stermann A, Durmus T, Lobitz S, Zenclussen AC, Christiansen H, Lode HN, Fest S. Salmonella SL7207 application is the most effective DNA vaccine delivery method for successful tumor eradication in a murine model for neuroblastoma. Cancer Lett 2013; 331:167-73. [DOI: 10.1016/j.canlet.2012.12.026] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Accepted: 12/25/2012] [Indexed: 12/17/2022]
|
32
|
Zhou Q, Zhu K, Cheng H. Toll-like receptors in human papillomavirus infection. Arch Immunol Ther Exp (Warsz) 2013; 61:203-15. [PMID: 23435874 DOI: 10.1007/s00005-013-0220-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 02/13/2013] [Indexed: 01/02/2023]
Abstract
Infection with human papillomaviruses (HPVs) often causes cutaneous benign lesions, cervical cancer, and a number of other tumors. The mechanisms of host immune system to prevent and control HPV infection still remain poorly understood. Toll-like receptors (TLRs) are specific pattern recognition molecules that bind to microbial components to trigger innate immunity and direct adaptive immunity in the face of immunological danger. TLRs have been established to play an essential role in sensing and initiating antiviral immune responses. Recent accumulating evidence demonstrated that HPVs modulate TLR expression and interfere with TLR signaling pathways, leading to persistent viral infection and carcinogenesis. This review summarizes current knowledge on the roles of TLR during HPV infection, focusing on TLR recognition, modulation of TLR expression and signaling, regulatory receptors involved in TLR signaling, and cross-talk of TLRs with antimicrobial peptides. Immunotherapeutic strategies based on TLR agonists have emerged to be one of the novel promising avenues in treatment of HPV-associated diseases in the future.
Collapse
Affiliation(s)
- Qiang Zhou
- Department of Dermatology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 Qingchun Road East, Hangzhou 310016, People's Republic of China
| | | | | |
Collapse
|
33
|
Luo Z, Shi H, Zhang H, Li M, Zhao Y, Zhang J, Guo F, Luo S, Sun P, Zhang D, Qian Z, Yang L. Plasmid DNA containing multiple CpG motifs triggers a strong immune response to hepatitis B surface antigen when combined with incomplete Freund's adjuvant but not aluminum hydroxide. Mol Med Rep 2012; 6:1309-14. [PMID: 22971976 DOI: 10.3892/mmr.2012.1079] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Accepted: 07/04/2012] [Indexed: 02/05/2023] Open
Abstract
Adjuvants are important components of recombinant protein vaccines which are often poorly immunogenic. For decades, the search for new vaccine adjuvants has been predominantly empirical. In addition, combinations of more than one adjuvant plus antigen have been systematically studied. Plasmid DNA containing additional oligodeoxynucleotides with unmethylated CpG motifs (CpG ODN) entrapped in liposomes has been used as an adjuvant for DNA vaccines and has shown powerful immunostimulatory functions. In our study, the combination of plasmid DNA containing 16 additional CpG ODNs (pv-16CpG) and aluminum hydroxide (AL) or incomplete Freund's adjuvant (IFA) was used as an adjuvant for a hepatitis B surface antigen (HBsAg) vaccine to immunize C57BL/6J mice. ELISA and ELISPOT assays were used to analyze the immunological effects of the novel vaccine. A significant enhancement of the anti-HBs titer and seroconversion was observed when the CpG plasmid was combined with IFA, but not with AL. In addition, anti-HBs antibody isotype analysis revealed that the combination of CpG plasmid and IFA induced a strong HBsAg-specific IgG2a response. Moreover, the ELISPOT assays suggested that pv-16CpG suspended in IFA evoked a strong T helper 1 (Th1) immune response and high IFN-γ production. These results demonstrate that pv-16CpG suspended in IFA is able to induce cellular and humoral immune responses to HBsAg, and confirm its potential as an adjuvant for use in protein vaccines.
Collapse
Affiliation(s)
- Zichao Luo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
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: 125] [Impact Index Per Article: 10.4] [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
|
35
|
Grant EV, Thomas M, Fortune J, Klibanov AM, Letvin NL. Enhancement of plasmid DNA immunogenicity with linear polyethylenimine. Eur J Immunol 2012; 42:2937-48. [DOI: 10.1002/eji.201242410] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Revised: 06/05/2012] [Accepted: 08/01/2012] [Indexed: 11/11/2022]
Affiliation(s)
- Evita V. Grant
- Division of Viral Pathogenesis; Beth Israel Deaconess Medical Center; Harvard Medical School; Boston Massachusetts USA
| | - Mini Thomas
- Department of Chemistry; Purdue University; West Lafayette Indiana USA
| | - Jennifer Fortune
- Department of Chemistry and Division of Biological Engineering; Massachusetts Institute of Technology; Cambridge Massachusetts USA
| | - Alexander M. Klibanov
- Department of Chemistry and Division of Biological Engineering; Massachusetts Institute of Technology; Cambridge Massachusetts USA
| | - Norman L. Letvin
- Division of Viral Pathogenesis; Beth Israel Deaconess Medical Center; Harvard Medical School; Boston Massachusetts USA
| |
Collapse
|
36
|
Abstract
This review provides a detailed look at the attributes and immunologic mechanisms of plasmid DNA vaccines and their utility as laboratory tools as well as potential human vaccines. The immunogenicity and efficacy of DNA vaccines in a variety of preclinical models is used to illustrate how they differ from traditional vaccines in novel ways due to the in situ antigen production and the ease with which they are constructed. The ability to make new DNA vaccines without needing to handle a virulent pathogen or to adapt the pathogen for manufacturing purposes demonstrates the potential value of this vaccine technology for use against emerging and epidemic pathogens. Similarly, personalized anti-tumor DNA vaccines can also readily be made from a biopsy. Because DNA vaccines bias the T-helper (Th) cell response to a Th1 phenotype, DNA vaccines are also under development for vaccines against allergy and autoimmune diseases. The licensure of four animal health products, including two prophylactic vaccines against infectious diseases, one immunotherapy for cancer, and one gene therapy delivery of a hormone for a food animal, provides evidence of the efficacy of DNA vaccines in multiple species including horses and pigs. The size of these target animals provides evidence that the somewhat disappointing immunogenicity of DNA vaccines in a number of human clinical trials is not due simply to the larger mass of humans compared with most laboratory animals. The insights gained from the mechanisms of protection in the animal vaccines, the advances in the delivery and expression technologies for increasing the potency of DNA vaccines, and encouragingly potent human immune responses in certain clinical trials, provide insights for future efforts to develop DNA vaccines into a broadly useful vaccine and immunotherapy platform with applications for human and animal health.
Collapse
|
37
|
Almeida LP, Trombone AP, Lorenzi JC, Rocha CD, Malardo T, Fontoura IC, Gembre AF, Silva RL, Silva CL, Castelo AP, Coelho-Castelo AA. B cells Can Modulate the CD8 Memory T Cell after DNA Vaccination Against Experimental Tuberculosis. GENETIC VACCINES AND THERAPY 2011; 9:5. [PMID: 21401938 PMCID: PMC3066104 DOI: 10.1186/1479-0556-9-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Accepted: 03/14/2011] [Indexed: 11/21/2022]
Abstract
Background Although B cells are important as antigen presenting cells (APC) during the immune response, their role in DNA vaccination models is unknown. Methods In this study in vitro and in vivo experiments were performed to evaluate the ability of B cells to protect mice against Mycobacterium tuberculosis challenge. Results In vitro and in vivo studies showed that B cells efficiently present antigens after naked plasmid pcDNA3 encoding M. leprae 65-kDa heat shock protein (pcDNA3-Hsp65) internalization and protect B knock-out (BKO) mice against Mycobacterium tuberculosis infection. pcDNA3-Hsp65-transfected B cells adoptively transferred into BKO mice rescued the memory phenotypes and reduced the number of CFU compared to wild-type mice. Conclusions These data not only suggest that B cells play an important role in the induction of CD8 T cells but also that they improve bacterial clearance in DNA vaccine model.
Collapse
Affiliation(s)
- Luciana P Almeida
- Department of Biochemistry and Immunology, Medical School of Ribeirão Preto, University of São Paulo, Brazil.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
|
39
|
Abstract
Efforts to make vaccines against infectious diseases as well as immunotherapies for cancer, autoimmune diseases and allergy have utilized a variety of heterologous expression systems, including viral and bacterial vectors, as well as DNA and RNA constructs. This review explores the immunologic rationale and provides an update of insights obtained from preclinical and clinical studies of such vaccines.
Collapse
|
40
|
Wikström FH, Fossum C, Fuxler L, Kruse R, Lövgren T. Cytokine induction by immunostimulatory DNA in porcine PBMC is impaired by a hairpin forming sequence motif from the genome of Porcine Circovirus type 2 (PCV2). Vet Immunol Immunopathol 2010; 139:156-66. [PMID: 20980058 DOI: 10.1016/j.vetimm.2010.09.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Revised: 09/22/2010] [Accepted: 09/28/2010] [Indexed: 01/07/2023]
Abstract
Porcine Circovirus type 2 (PCV2) can cause postweaning multisystemic wasting syndrome (PMWS) in young pigs with severe immunosuppression as a major characteristic of the disease complex. Despite the dramatic involvement of the immune system, the interaction between PCV2 and the host is until date not well understood. The DNA genome of PCV2 contains sequences that in synthetic form (oligodeoxyribonucleotides; ODNs) can act immunomodulatory on porcine peripheral blood mononuclear cells (poPBMCs) in vitro. One such sequence (ODN PCV2/1) acts inhibitory on interferon (IFN)-α production induced by immunostimulatory DNA but not that induced by RNA, and the inhibitory activity is dependent on secondary structure formation. In the present study, the characteristic of ODN PCV2/1 was examined further by altering the nucleotide sequence to disrupt hairpin structure formation but still enable multimer structures through G-tetrads. This modification resulted in loss of IFN-α-inhibitory activity of the ODN and thus indicated the importance of hairpin structures. In addition, ODN PCV2/1 was compared to another inhibitory ODN (IRS 869) previously used in human and murine cells. In contrast to ODN PCV2/1, ODN IRS 869 did not inhibit IFN-α production induced by class A ODN 2216 but was a more efficient inhibitor of IFN-α production induced by plasmid DNA than ODN PCV2/1. In cultures induced by the RNA stimulator Poly I:C, however, a strong synergistic IFN-α stimulatory effect was seen in combination with ODN IRS 869. These results indicate that ODN PCV2/1 and ODN IRS 869 function through separate mechanisms to affect cytokine production by immune cells. The effect of ODN PCV2/1 was studied further by monitoring the expression of mRNA for IFN-α, IL-12p40, IL-10, IL-6, IFN-γ, IL-1β, TGF-β, and TNF-α in cultures of poPBMC stimulated with ODN 2216 or Poly I:C. Results from qPCR analyses showed that ODN PCV2/1 clearly inhibited the expression of IFN-α, IL-12p40, IL-10 and IL-6 when induced by ODN 2216, but did not seem to affect any of the cytokines examined when induced by Poly I:C. Initial studies using confocal microscopy and fluorochrome labelled ODNs indicate that ODN 2216 and ODN PCV2/1 co-localize in subpopulations of poPBMC.
Collapse
Affiliation(s)
- F Hasslung Wikström
- Department of Biomedical Sciences and Veterinary Public Health, Section for Immunology, Swedish University of Agricultural Sciences, BMC, P.O. Box 588, SE-751 23 Uppsala, Sweden
| | | | | | | | | |
Collapse
|
41
|
Rottembourg D, Filippi CM, Bresson D, Ehrhardt K, Estes EA, Oldham JE, von Herrath MG. Essential role for TLR9 in prime but not prime-boost plasmid DNA vaccination to activate dendritic cells and protect from lethal viral infection. THE JOURNAL OF IMMUNOLOGY 2010; 184:7100-7. [PMID: 20483769 DOI: 10.4049/jimmunol.0803935] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
One of the requirements for efficient vaccination against infection is to achieve the best combination of an adequate adjuvant with the antigenic information to deliver. Although plasmid DNA is a promising tool bearing the unique potential to activate humoral and cellular immunity, an actual challenge is to increase plasmid immunogenicity in human vaccination protocols in which efficacy has proven rather limited. Previous work showed that the bacterial DNA backbone of the plasmid has potent adjuvant properties because it contains CpG motifs that are particular activating nucleotidic sequences. Among TLRs, which are key sensors of microbial products, TLR9 can detect CpG motifs and confer activation of APCs, such as dendritic cells. However, whether the immunogenic properties of plasmid DNA involve TLR9 signaling has not been clearly established. In the current study, we demonstrate that TLR9 determines the effectiveness of vaccination against lethal lymphocytic choriomeningitis virus infection using plasmid DNA in a prime, but not prime-boost, vaccination regimen. Furthermore, we provide evidence that the presence of TLR9 in dendritic cells is necessary for effective and functional priming of virus-specific CD8+ T cells upon plasmid exposure in vitro or single-dose vaccination in vivo. Therefore, at single or low vaccine doses that are often used in human-vaccination protocols, CpG/TLR9 interactions participate in the immunogenicity of plasmid DNA. These results suggest that the TLR9 signaling pathway is involved in the efficacy of plasmid vaccination; therefore, it should remain a focus in the development or amelioration of vaccines to treat infections in humans.
Collapse
Affiliation(s)
- Diane Rottembourg
- La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
DNA vaccines: developing new strategies against cancer. J Biomed Biotechnol 2010; 2010:174378. [PMID: 20368780 PMCID: PMC2846346 DOI: 10.1155/2010/174378] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2009] [Accepted: 02/05/2010] [Indexed: 12/14/2022] Open
Abstract
Due to their rapid and widespread development, DNA vaccines have entered into a variety of human clinical trials for vaccines against various diseases including cancer. Evidence that DNA vaccines are well tolerated and have an excellent safety profile proved to be of advantage as many clinical trials combines the first phase with the second, saving both time and money. It is clear from the results obtained in clinical trials that such DNA vaccines require much improvement in antigen expression and delivery methods to make them sufficiently effective in the clinic. Similarly, it is clear that additional strategies are required to activate effective immunity against poorly immunogenic tumor antigens. Engineering vaccine design for manipulating antigen presentation and processing pathways is one of the most important aspects that can be easily handled in the DNA vaccine technology. Several approaches have been investigated including DNA vaccine engineering, co-delivery of immunomodulatory molecules, safe routes of administration, prime-boost regimen and strategies to break the immunosuppressive networks mechanisms adopted by malignant cells to prevent immune cell function. Combined or single strategies to enhance the efficacy and immunogenicity of DNA vaccines are applied in completed and ongoing clinical trials, where the safety and tolerability of the DNA platform are substantiated.
In this review on DNA vaccines, salient aspects on this topic going from basic research to the clinic are evaluated. Some representative DNA cancer vaccine studies are also discussed.
Collapse
|
43
|
Ohlschläger P, Spies E, Alvarez G, Quetting M, Groettrup M. The combination of TLR-9 adjuvantation and electroporation-mediated delivery enhances in vivo antitumor responses after vaccination with HPV-16 E7 encoding DNA. Int J Cancer 2010; 128:473-81. [PMID: 20309939 DOI: 10.1002/ijc.25344] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2009] [Accepted: 03/11/2010] [Indexed: 11/11/2022]
Abstract
Therapeutic DNA vaccination is an attractive adjuvant option to conventional methods in the fight against cancer, like surgery radiotherapy and chemotherapy. Despite strong antitumor effects that were observed in small animals with different antigens, DNA-based vaccines remain weakly immunogenic in large animals and primates compared to protein-based vaccines. Here, we sought to enhance the immunogenicity of a therapeutic nontransforming cervical cancer DNA vaccine (HPV-16 E7SH) by introduction of a highly optimized CpG cassette into the plasmid backbone as well as by an optimized DNA delivery using an advanced electroporation (EP) technology. By integrating the means for agent administration and EP into a single device, this technology enables a simple, one-step procedure that facilitates reproducibility. We found that highly optimized CpG motifs alone triggers an enhanced IFN-γ and granzyme B response in Elispot assays as well as stronger tumor regression. Furthermore, these effects could be dramatically enhanced when the CpG cassette containing plasmid was administered via the newly developed EP technology. These data suggest that an optimized application of CpG-enriched DNA vaccines may be an attractive strategy for the treatment of cancer. Collectively, these results provide a basis for the transfer of preclinical therapeutic DNA-based immunization studies into successful clinical cancer trials.
Collapse
Affiliation(s)
- Peter Ohlschläger
- Department of Immunology, University of Constance, 78457 Constance, Germany.
| | | | | | | | | |
Collapse
|
44
|
Laddy DJ, Weiner DB. From Plasmids to Protection: A Review of DNA Vaccines Against Infectious Diseases. Int Rev Immunol 2009; 25:99-123. [PMID: 16818367 DOI: 10.1080/08830180600785827] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
The field of DNA vaccine development began over 16 years ago with the observation that plasmid DNA could be injected into and expressed in vivo and drive adaptive immune responses. Since then, there has been great interest in developing this technology to create a new generation of vaccines with the ability to elicit both humoral and cellular immune responses from an inherently innocuous injection. However, DNA vaccines have yet to proceed past phase I/II clinical trials in humans--primarily due to a desire to induce more potent immune responses. This review will examine how DNA vaccines function to induce an immune response and how this information might be useful in future vaccine design.
Collapse
Affiliation(s)
- Dominick J Laddy
- Department of Pathology & Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | |
Collapse
|
45
|
Larsen KC, Spencer AJ, Goodman AL, Gilchrist A, Furze J, Rollier CS, Kiss-Toth E, Gilbert SC, Bregu M, Soilleux EJ, Hill AVS, Wyllie DH. Expression of tak1 and tram induces synergistic pro-inflammatory signalling and adjuvants DNA vaccines. Vaccine 2009; 27:5589-98. [PMID: 19646407 DOI: 10.1016/j.vaccine.2009.07.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2009] [Revised: 05/21/2009] [Accepted: 07/13/2009] [Indexed: 01/07/2023]
Abstract
Improving vaccine immunogenicity remains a major challenge in the fight against developing country diseases like malaria and AIDS. We describe a novel strategy to identify new DNA vaccine adjuvants. We have screened components of the Toll-like receptor signalling pathways for their ability to activate pro-inflammatory target genes in transient transfection assays and assessed in vivo adjuvant activity by expressing the activators from the DNA backbone of vaccines. We find that a robust increase in the immune response necessitates co-expression of two activators. Accordingly, the combination of tak1 and tram elicits synergistic reporter activation in transient transfection assays. In a mouse model this combination, but not the individual molecules, induced approximately twofold increases in CD8+ T-cell immune responses. These results indicate that optimal immunogenicity may require activation of distinct innate immune signalling pathways. Thus this strategy offers a novel route to the discovery of a new generation of adjuvants.
Collapse
|
46
|
Abstract
We have focused our research on understanding the basic biology of and developing novel therapeutic and prophylactic DNA vaccines. We have among others three distinct primary areas of interest which include: 1. Enhancing in vivo delivery and transfection of DNA vaccine vectors 2. Improving DNA vaccine construct immunogenicity 3. Using molecular adjuvants to modulate and skew immune responses. Key to the immunogenicity of DNA vaccines is the presentation of expressed antigen to antigen-presenting cells. To improve expression and presentation of antigen, we have investigated various immunization methods with current focus on a combination of intramuscular injection and electroporation. To improve our vaccine constructs, we also employed methods such as RNA/codon optimization and antigen consensus to enhance expression and cellular/humoral cross-reactivity, respectively. Our lab also researches the potential of various molecular adjuvants to skew Th1/Th2 responses, enhance cellular/humoral responses, and improve protection in various animal models. Through improving our understanding of basic immunology as it is related to DNA vaccine technology, our goal is to develop the technology to the point of utility for human and animal health.
Collapse
Affiliation(s)
- Shaheed A Abdulhaqq
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, 422 Curie Blvd, 505 Stellar-Chance Laboratories, Philadelphia, PA, 19104, USA
| | | |
Collapse
|
47
|
Nguyen DN, Green JJ, Chan JM, Longer R, Anderson DG. Polymeric Materials for Gene Delivery and DNA Vaccination. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2009; 21:847-867. [PMID: 28413262 PMCID: PMC5391878 DOI: 10.1002/adma.200801478] [Citation(s) in RCA: 177] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Gene delivery holds great potential for the treatment of many different diseases. Vaccination with DNA holds particular promise, and may provide a solution to many technical challenges that hinder traditional vaccine systems including rapid development and production and induction of robust cell-mediated immune responses. However, few candidate DNA vaccines have progressed past preclinical development and none have been approved for human use. This Review focuses on the recent progress and challenges facing materials design for nonviral DNA vaccine drug delivery systems. In particular, we highlight work on new polymeric materials and their effects on protective immune activation, gene delivery, and current efforts to optimize polymeric delivery systems for DNA vaccination.
Collapse
Affiliation(s)
- David N Nguyen
- Massachusetts Institute of Technology, 77 Massachusetts Ave, E25 Room 342, Cambridge, MA 02139 (USA)
| | - Jordan J Green
- Massachusetts Institute of Technology, 77 Massachusetts Ave, E25 Room 342, Cambridge, MA 02139 (USA)
| | - Juliana M Chan
- Massachusetts Institute of Technology, 77 Massachusetts Ave, E25 Room 342, Cambridge, MA 02139 (USA)
| | - Robert Longer
- Massachusetts Institute of Technology, 77 Massachusetts Ave, E25 Room 342, Cambridge, MA 02139 (USA)
| | - Daniel G Anderson
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Ave, E25 Room 342, Cambridge, MA 02139 (USA)
| |
Collapse
|
48
|
Ishii KJ, Kawagoe T, Koyama S, Matsui K, Kumar H, Kawai T, Uematsu S, Takeuchi O, Takeshita F, Coban C, Akira S. TANK-binding kinase-1 delineates innate and adaptive immune responses to DNA vaccines. Nature 2008; 451:725-9. [PMID: 18256672 DOI: 10.1038/nature06537] [Citation(s) in RCA: 487] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2007] [Accepted: 11/29/2007] [Indexed: 11/09/2022]
Abstract
Successful vaccines contain not only protective antigen(s) but also an adjuvant component that triggers innate immune activation and is necessary for their optimal immunogenicity. In the case of DNA vaccines, this consists of plasmid DNA; however, the adjuvant element(s) as well as its intra- and inter-cellular innate immune signalling pathway(s) leading to the encoded antigen-specific T- and B-cell responses remain unclear. Here we demonstrate in vivo that TANK-binding kinase 1 (TBK1), a non-canonical IkappaB kinase, mediates the adjuvant effect of DNA vaccines and is essential for its immunogenicity in mice. Plasmid-DNA-activated, TBK1-dependent signalling and the resultant type-I interferon receptor-mediated signalling was required for induction of antigen-specific B and T cells, which occurred even in the absence of innate immune signalling through a well known CpG DNA sensor-Toll-like receptor 9 (TLR9) or Z-DNA binding protein 1 (ZBP1, also known as DAI, which was recently reported as a potential B-form DNA sensor). Moreover, bone-marrow-transfer experiments revealed that TBK1-mediated signalling in haematopoietic cells was critical for the induction of antigen-specific B and CD4(+) T cells, whereas in non-haematopoietic cells TBK1 was required for CD8(+) T-cell induction. These data suggest that TBK1 is a key signalling molecule for DNA-vaccine-induced immunogenicity, by differentially controlling DNA-activated innate immune signalling through haematopoietic and non-haematopoietic cells.
Collapse
Affiliation(s)
- Ken J Ishii
- Exploratory Research for Advanced Technology (ERATO), Japan Science and Technology Agency (JST).
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Schmitz F, Heit A. Protective cancer immunotherapy: what can the innate immune system contribute? Expert Opin Biol Ther 2008; 8:31-43. [PMID: 18081535 DOI: 10.1517/14712598.8.1.31] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Despite significant efforts to induce protection against malignant diseases, the clinical effects of antitumour vaccines are poor. However, recent studies on a quadrivalent human papilloma virus vaccine suggest that protection against secondary tumour development is feasible. While this scenario benefits rather from antiviral protection than from direct antitumour responses, immunisation against cancers of non-viral origin demands strategies that rely on the circumvention of intrinsic regulatory mechanisms. Strong activation of innate immune cells seems to be key and, thus, the choice of adjuvant determines vaccination efficacy. The recently acquired knowledge about molecular and cellular recognition of microbial molecules suggests how one can modulate innate and adaptive immune reactions to potentially induce robust T- and B-cell reactions capable of prohibiting tumour development and progression. Here, the authors review the present knowledge of innate immune reactions, which may help to define rationales on the design of novel antitumour vaccines.
Collapse
Affiliation(s)
- Frank Schmitz
- Technical University Munich, Institute of Medical Microbiology, Immunology and Hygiene, Trogerstrasse 30, 81675 Munich, Germany.
| | | |
Collapse
|
50
|
Pavlenko M, Leder C, Moreno S, Levitsky V, Pisa P. Priming of CD8+ T-cell responses after DNA immunization is impaired in TLR9- and MyD88-deficient mice. Vaccine 2007; 25:6341-7. [PMID: 17628235 DOI: 10.1016/j.vaccine.2007.06.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2007] [Revised: 06/11/2007] [Accepted: 06/11/2007] [Indexed: 11/18/2022]
Abstract
The plasmid DNA vaccine not only provides expression of the antigen in vivo, but also activates cells of the innate immune system via unmethylated CpG-containing DNA sequences that are recognized by Toll like receptor 9 (TLR9). The requirement of such immunostimulatory activity for induction of CD8+ T-cell responses after DNA immunization is still controversial. In the present study we assessed induction of CD8+ T-cell responses against an immunodominant H-2D(b)-restricted epitope of human prostate-specific antigen in C57Bl/6 (wild-type), TLR9- and MyD88-deficient mice. A single DNA immunization resulted in efficient priming of CD8+ T responses in wild-type mice but not in TLR9- or MyD88-deficient mice. However, priming of CD8+ T cell responses was observed in TLR9-deficient but not in MyD88-deficient mice after multiple DNA immunizations. Moreover, induction of CD8+ T cell responses in TLR9-deficient mice was dependent on the presence of endotoxin contamination in plasmid DNA preparations. Collectively, these results demonstrate that TLR9-dependent immunostimulatory activity of plasmid DNA is essential for priming of CD8+ T-cell responses and that other bacterial compounds present in plasmid DNA preparations and acting via MyD88-dependent pathway could provide alternative signals necessary for priming of CD8+ T cells.
Collapse
Affiliation(s)
- Maxim Pavlenko
- Department of Oncology and Pathology, Cancer Center Karolinska, Immune and Gene Therapy Laboratory, Karolinska Institute, Stockholm, Sweden
| | | | | | | | | |
Collapse
|