1
|
Neeli P, Maza PAMA, Chai D, Zhao D, Hoi XP, Chan KS, Young KH, Li Y. DNA vaccines against GPRC5D synergize with PD-1 blockade to treat multiple myeloma. NPJ Vaccines 2024; 9:180. [PMID: 39353958 PMCID: PMC11445568 DOI: 10.1038/s41541-024-00979-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 09/19/2024] [Indexed: 10/03/2024] Open
Abstract
Multiple myeloma (MM), a hematological malignancy of the bone marrow, remains largely incurable. The orphan G protein-coupled receptor, GPRC5D, which is uniquely expressed in plasma cells and highly expressed in MM, is a compelling candidate for immunotherapy. In this study, we investigated the efficacy of a combination of DNA vaccine encoding mouse GPRC5D and PD-1 blockade in preventing and treating MM using the 5TGM1 murine model of MM. The mouse vaccine alone was effective in preventing myeloma growth but required PD-1 antibodies to inhibit established MM tumors. We next evaluated the prophylactic and therapeutic efficacy of a nanoplasmid vector encoding human GPRC5D in several murine syngeneic tumor models. Similar results for tumor inhibition were observed, as human GPRC5D-specific T cells and antibodies were induced by DNA vaccines. Taken together, these findings underscore the potential of GPRC5D-targeted DNA vaccines as versatile platforms for the treatment and prevention of MM.
Collapse
Affiliation(s)
- Praveen Neeli
- Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA.
| | | | - Dafei Chai
- Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Dan Zhao
- Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Xen Ping Hoi
- Department of Urology, Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, USA
| | - Keith Syson Chan
- Department of Urology, Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, USA
| | - Ken H Young
- Department of Pathology, Division of Hematopathology, Duke University Medical Center, Durham, NC, USA
| | - Yong Li
- Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
2
|
Yoneyama M, Kato H, Fujita T. Physiological functions of RIG-I-like receptors. Immunity 2024; 57:731-751. [PMID: 38599168 DOI: 10.1016/j.immuni.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 02/19/2024] [Accepted: 03/04/2024] [Indexed: 04/12/2024]
Abstract
RIG-I-like receptors (RLRs) are crucial for pathogen detection and triggering immune responses and have immense physiological importance. In this review, we first summarize the interferon system and innate immunity, which constitute primary and secondary responses. Next, the molecular structure of RLRs and the mechanism of sensing non-self RNA are described. Usually, self RNA is refractory to the RLR; however, there are underlying host mechanisms that prevent immune reactions. Studies have revealed that the regulatory mechanisms of RLRs involve covalent molecular modifications, association with regulatory factors, and subcellular localization. Viruses have evolved to acquire antagonistic RLR functions to escape the host immune reactions. Finally, the pathologies caused by the malfunction of RLR signaling are described.
Collapse
Affiliation(s)
- Mitsutoshi Yoneyama
- Division of Molecular Immunology, Medical Mycology Research Center, Chiba University, Chiba, Japan; Division of Pandemic and Post-disaster Infectious Diseases, Research Institute of Disaster Medicine, Chiba University, Chiba, Japan
| | - Hiroki Kato
- Institute of Cardiovascular Immunology, Medical Faculty, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Takashi Fujita
- Institute of Cardiovascular Immunology, Medical Faculty, University Hospital Bonn, University of Bonn, Bonn, Germany; Laboratory of Regulatory Information, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan.
| |
Collapse
|
3
|
Pushko P, Lukashevich IS, Johnson DM, Tretyakova I. Single-Dose Immunogenic DNA Vaccines Coding for Live-Attenuated Alpha- and Flaviviruses. Viruses 2024; 16:428. [PMID: 38543793 PMCID: PMC10974764 DOI: 10.3390/v16030428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 03/04/2024] [Accepted: 03/07/2024] [Indexed: 04/01/2024] Open
Abstract
Single-dose, immunogenic DNA (iDNA) vaccines coding for whole live-attenuated viruses are reviewed. This platform, sometimes called immunization DNA, has been used for vaccine development for flavi- and alphaviruses. An iDNA vaccine uses plasmid DNA to launch live-attenuated virus vaccines in vitro or in vivo. When iDNA is injected into mammalian cells in vitro or in vivo, the RNA genome of an attenuated virus is transcribed, which starts replication of a defined, live-attenuated vaccine virus in cell culture or the cells of a vaccine recipient. In the latter case, an immune response to the live virus vaccine is elicited, which protects against the pathogenic virus. Unlike other nucleic acid vaccines, such as mRNA and standard DNA vaccines, iDNA vaccines elicit protection with a single dose, thus providing major improvement to epidemic preparedness. Still, iDNA vaccines retain the advantages of other nucleic acid vaccines. In summary, the iDNA platform combines the advantages of reverse genetics and DNA immunization with the high immunogenicity of live-attenuated vaccines, resulting in enhanced safety and immunogenicity. This vaccine platform has expanded the field of genetic DNA and RNA vaccines with a novel type of immunogenic DNA vaccines that encode entire live-attenuated viruses.
Collapse
Affiliation(s)
- Peter Pushko
- Medigen, Inc., 8420 Gas House Pike Suite S, Frederick, MD 21701, USA;
| | - Igor S. Lukashevich
- Department of Pharmacology and Toxicology, School of Medicine, Center for Predictive Medicine and Emerging Infectious Diseases, University of Louisville, 505 S Hancock St., Louisville, KY 40202, USA;
| | - Dylan M. Johnson
- Department of Biotechnology & Bioengineering, Sandia National Laboratories, Livermore, CA 945501, USA;
| | - Irina Tretyakova
- Medigen, Inc., 8420 Gas House Pike Suite S, Frederick, MD 21701, USA;
| |
Collapse
|
4
|
Marie C, Scherman D. Antibiotic-Free Gene Vectors: A 25-Year Journey to Clinical Trials. Genes (Basel) 2024; 15:261. [PMID: 38540320 PMCID: PMC10970329 DOI: 10.3390/genes15030261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/07/2024] [Accepted: 02/15/2024] [Indexed: 06/15/2024] Open
Abstract
Until very recently, the major use, for gene therapy, specifically of linear or circular DNA, such as plasmids, was as ancillary products for viral vectors' production or as a genetic template for mRNA production. Thanks to targeted and more efficient physical or chemical delivery techniques and to the refinement of their structure, non-viral plasmid DNA are now under intensive consideration as pharmaceutical drugs. Plasmids traditionally carry an antibiotic resistance gene for providing the selection pressure necessary for maintenance in a bacterial host. Nearly a dozen different antibiotic-free gene vectors have now been developed and are currently assessed in preclinical assays and phase I/II clinical trials. Their reduced size leads to increased transfection efficiency and prolonged transgene expression. In addition, associating non-viral gene vectors and DNA transposons, which mediate transgene integration into the host genome, circumvents plasmid dilution in dividing eukaryotic cells which generate a loss of the therapeutic gene. Combining these novel molecular tools allowed a significantly higher yield of genetically engineered T and Natural Killer cells for adoptive immunotherapies due to a reduced cytotoxicity and increased transposition rate. This review describes the main progresses accomplished for safer, more efficient and cost-effective gene and cell therapies using non-viral approaches and antibiotic-free gene vectors.
Collapse
Affiliation(s)
- Corinne Marie
- Université Paris Cité, CNRS, Inserm, UTCBS, 75006 Paris, France;
- Chimie ParisTech, Université PSL, 75005 Paris, France
| | - Daniel Scherman
- Université Paris Cité, CNRS, Inserm, UTCBS, 75006 Paris, France;
- Fondation Maladies Rares, 75014 Paris, France
| |
Collapse
|
5
|
Lassaunière R, Polacek C, Linnea Tingstedt J, Fomsgaard A. Preclinical evaluation of a SARS-CoV-2 variant B.1.351-based candidate DNA vaccine. Vaccine 2023; 41:6505-6513. [PMID: 37726179 DOI: 10.1016/j.vaccine.2023.09.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/22/2023] [Accepted: 09/12/2023] [Indexed: 09/21/2023]
Abstract
The SARS-CoV-2 pandemic revealed the critical shortfalls of global vaccine availability for emergent pathogens and the need for exploring additional vaccine platforms with rapid update potential in response to new variants. Thus, it remains essential, for the present evolving SARS-CoV-2/Covid-19 and future pandemics, to continuously develop and characterize new and different vaccine platforms. Here, we describe an expression-optimized DNA vaccine candidate based on the SARS-CoV-2 spike protein of the Beta variant (B.1.351), pNTC-Spike.351, and, in animal models, compare its immunogenicity with a similar DNA vaccine encoding the ancestral index strain spike protein, pNTC-Spike. Both DNA vaccines induced neutralizing antibodies and a Th1 biased immune response. In contrast to the index-specific vaccine, the Beta-specific DNA vaccine induced antibodies in mice and rabbits that, even at low levels, efficiently neutralize the otherwise antibody resistant Beta variant. It similarly neutralized unrelated variants bearing the neutralization resistant E484K spike mutation. Intensive priming using two vaccinations with pNTC-Spike and a single booster immunization with the pNTC-Spike.351 induced a more robust neutralizing antibody response with comparable magnitude against different variants of concern. Thus, DNA vaccine technology with heterologous spike protein prime-boost should be explored further using the Beta derived pNTC-Spike.351 to broaden neutralizing antibody responses against emerging variants of concern.
Collapse
Affiliation(s)
- Ria Lassaunière
- Department of Virus and Microbiological Special Diagnostic, Statens Serum Institut, Copenhagen, Denmark
| | - Charlotta Polacek
- Department of Virus and Microbiological Special Diagnostic, Statens Serum Institut, Copenhagen, Denmark
| | - Jeanette Linnea Tingstedt
- Department of Virus and Microbiological Special Diagnostic, Statens Serum Institut, Copenhagen, Denmark
| | - Anders Fomsgaard
- Department of Virus and Microbiological Special Diagnostic, Statens Serum Institut, Copenhagen, Denmark; Infectious Disease Research Unit, Clinical Institute, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
6
|
Wang H, Xu J, Xiang L. Microneedle-Mediated Transcutaneous Immunization: Potential in Nucleic Acid Vaccination. Adv Healthc Mater 2023; 12:e2300339. [PMID: 37115817 DOI: 10.1002/adhm.202300339] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/07/2023] [Indexed: 04/29/2023]
Abstract
Efforts aimed at exploring economical and efficient vaccination have taken center stage to combat frequent epidemics worldwide. Various vaccines have been developed for infectious diseases, among which nucleic acid vaccines have attracted much attention from researchers due to their design flexibility and wide application. However, the lack of an efficient delivery system considerably limits the clinical translation of nucleic acid vaccines. As mass vaccinations via syringes are limited by low patient compliance and high costs, microneedles (MNs), which can achieve painless, cost-effective, and efficient drug delivery, can provide an ideal vaccination strategy. The MNs can break through the stratum corneum barrier in the skin and deliver vaccines to the immune cell-rich epidermis and dermis. In addition, the feasibility of MN-mediated vaccination is demonstrated in both preclinical and clinical studies and has tremendous potential for the delivery of nucleic acid vaccines. In this work, the current status of research on MN vaccines is reviewed. Moreover, the improvements of MN-mediated nucleic acid vaccination are summarized and the challenges of its clinical translation in the future are discussed.
Collapse
Affiliation(s)
- Haochen Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Junhua Xu
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lin Xiang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
7
|
Neeli P, Chai D, Wang X, Sobhani N, Udeani G, Li Y. Comparison of DNA vaccines with AddaS03 as an adjuvant and an mRNA vaccine against SARS-CoV-2. iScience 2023; 26:107120. [PMID: 37361876 PMCID: PMC10271916 DOI: 10.1016/j.isci.2023.107120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 03/16/2023] [Accepted: 06/09/2023] [Indexed: 06/28/2023] Open
Abstract
Emerging variants of SARS-CoV-2 call for frequent changes in vaccine antigens. Nucleic acid-based vaccination strategies are superior as the coding sequences can be easily altered with little impact on downstream production. mRNA vaccines, including variant-specific boosters, are approved for SARS-CoV-2. Here, we tested the efficacy of DNA vaccines against the SARS-CoV-2 Spike aided by the AddaS03 adjuvant using electroporation and compared their immunogenicity with an approved mRNA vaccine (mRNA-1273). DNA vaccination elicited robust humoral and cellular immune responses in C57BL/6 mice with Spike-specific antibody neutralization and T cells produced from 20 μg DNA vaccines similar to that from 0.5 μg mRNA-1273. Furthermore, a Nanoplasmid-based vector further increased the immunogenicity. Our results indicate that adjuvants are critical to the efficacy of DNA vaccines in stimulating robust immune responses against Spike, highlighting the feasibility of plasmid DNA as a rapid nucleic acid-based vaccine approach against SARS-CoV-2 and other emerging infectious diseases.
Collapse
Affiliation(s)
- Praveen Neeli
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Dafei Chai
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xu Wang
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Navid Sobhani
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - George Udeani
- Department of Pharmacy Practice, Irma Lerma Rangel School of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA
| | - Yong Li
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
8
|
Williams JA, Paez PA. Improving cell and gene therapy safety and performance using next-generation Nanoplasmid vectors. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 32:494-503. [PMID: 37346980 PMCID: PMC10280095 DOI: 10.1016/j.omtn.2023.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/23/2023]
Abstract
The cell and gene therapy industry has employed the same plasmid technology for decades in vaccination, cell and gene therapy, and as a raw material in viral vector and RNA production. While canonical plasmids contain antibiotic resistance markers in bacterial backbones greater than 2,000 base pairs, smaller backbones increase expression level and durability and reduce the cell-transfection-associated toxicity and transgene silencing that can occur with canonical plasmids. Therefore, the small backbone and antibiotic-free selection method of Nanoplasmid vectors have proven to be a transformative replacement in a wide variety of applications, offering a greater safety profile and efficiency than traditional plasmids. This review provides an overview of the Nanoplasmid technology and highlights its specific benefits for various applications with examples from recent publications.
Collapse
Affiliation(s)
- James A. Williams
- Research & Development, Aldevron, 4055 41st Avenue S, Fargo, ND 58104, USA
| | - Patrick A. Paez
- Research & Development, Aldevron, 4055 41st Avenue S, Fargo, ND 58104, USA
| |
Collapse
|
9
|
Wang J, Ma Y, Li J, Peng R, Mao T, Sun X, Duan Z. An oral NoV-rAd5 vaccine with built-in dsRNA adjuvant elicits systemic immune responses in mice. Int Immunopharmacol 2023; 116:109801. [PMID: 36780828 DOI: 10.1016/j.intimp.2023.109801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/21/2023] [Accepted: 01/25/2023] [Indexed: 02/13/2023]
Abstract
Norovirus (NoV) is an enteric pathogen notorious for causing epidemics of acute gastroenteritis. An effective vaccine against NoV is therefore urgently needed. A short double-stranded RNA (dsRNA) has been described that acts as a retinoic-acid-inducible gene-I agonist to induce the production of type I interferon; it also exhibits adjuvant activity. Using built-in dsRNA of different lengths (DS1 and DS2), we developed a recombinant adenovirus 5 (rAd5) expressing NoV VP1, and evaluated its immunogenicity following oral administration in a mouse model. An in vitro study demonstrated that the dsRNA adjuvants significantly enhanced VP1 protein expression in infected cells. The oral administration of both rAd5-VP1-DS vaccines elicited high serum levels of VP1-specific IgG and blocking antibodies, as well as strong and long-lasting mucosal immunity. There was no apparent difference in immunostimulatory effects in immunised mice between the two dsRNA adjuvants. This study indicates that an oral NoV-rAd5 vaccine with a built-in dsRNA adjuvant may be developed to prevent NoV infection in humans.
Collapse
Affiliation(s)
- Jindong Wang
- Department of Pathogenic Biology, Weifang Medical University, Weifang 261053, China; National Institute for Viral Diseases Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Yalin Ma
- National Institute for Viral Diseases Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China; School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, China
| | - Jinsong Li
- National Institute for Viral Diseases Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Rui Peng
- National Institute for Viral Diseases Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Tongyao Mao
- National Institute for Viral Diseases Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Xiaoman Sun
- National Institute for Viral Diseases Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Zhaojun Duan
- National Institute for Viral Diseases Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China.
| |
Collapse
|
10
|
Kommineni N, Butreddy A, Sainaga Jyothi VG, Angsantikul P. Freeze-drying for the preservation of immunoengineering products. iScience 2022; 25:105127. [PMID: 36267916 PMCID: PMC9576584 DOI: 10.1016/j.isci.2022.105127] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Immunoengineering technologies harness the power of immune system modulators such as monoclonal antibodies, cytokines, and vaccines to treat myriad diseases. Immunoengineering innovations have showed great promise in various practices including oncology, infectious disease, autoimmune diseases, and transplantation. Despite the countless successes, the majority of immunoengineering products contain active moieties that are prone to instability. The current review aims to feature freeze-drying as a robust and scalable solution to the inherent stability challenges in immunoengineering products by preventing the active moiety from degradation. Furthermore, this review describes the stability issues related to immunoengineering products and the utility of the lyophilization process to preserve the integrity and efficacy of immunoengineering tools ranging from biologics to nanoparticle-based vaccines. The concept of the freeze-drying process is described highlighting the quality by design (QbD) for robust process optimization. Case studies of lyophilized immunoengineering technologies and relevant clinical studies using immunoengineering products are discussed.
Collapse
Affiliation(s)
| | - Arun Butreddy
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, Oxford, MS 38677, USA
| | - Vaskuri G.S. Sainaga Jyothi
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana 500037, India
| | | |
Collapse
|
11
|
Solstad A, Hogaboam O, Forero A, Hemann EA. RIG-I-like Receptor Regulation of Immune Cell Function and Therapeutic Implications. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:845-854. [PMID: 36130131 PMCID: PMC9512390 DOI: 10.4049/jimmunol.2200395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 06/30/2022] [Indexed: 01/04/2023]
Abstract
Retinoic acid-inducible gene I-like receptors (RLRs) are cytosolic RNA sensors critical for initiation of antiviral immunity. Activation of RLRs following RNA recognition leads to production of antiviral genes and IFNs for induction of broad antiviral immunity. Although the RLRs are ubiquitously expressed, much of our understanding of these molecules comes from their study in epithelial cells and fibroblasts. However, RLR activation is critical for induction of immune function and long-term protective immunity. Recent work has focused on the roles of RLRs in immune cells and their contribution to programming of effective immune responses. This new understanding of RLR function in immune cells and immune programming has led to the development of vaccines and therapeutics targeting the RLRs. This review covers recent advances in our understanding of the contribution of RLRs to immune cell function during infection and the emerging RLR-targeting strategies for induction of immunity against cancer and viral infection.
Collapse
Affiliation(s)
- Abigail Solstad
- Biomedical Sciences Graduate Program, College of Medicine, The Ohio State University, Columbus, OH
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH
| | - Octavia Hogaboam
- Biomedical Sciences Graduate Program, College of Medicine, The Ohio State University, Columbus, OH
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH
| | - Adriana Forero
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH
- Infectious Diseases Institute, The Ohio State University, Columbus, OH; and
| | - Emily A Hemann
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH;
- Infectious Diseases Institute, The Ohio State University, Columbus, OH; and
| |
Collapse
|
12
|
Maier C, Fuchs J, Irrgang P, Wißing MH, Beyerlein J, Tenbusch M, Lapuente D. Mucosal immunization with an adenoviral vector vaccine confers superior protection against RSV compared to natural immunity. Front Immunol 2022; 13:920256. [PMID: 36003372 PMCID: PMC9394428 DOI: 10.3389/fimmu.2022.920256] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 07/07/2022] [Indexed: 01/09/2023] Open
Abstract
Respiratory syncytial virus (RSV) infections are the leading cause of severe respiratory illness in early infancy. Although the majority of children and adults mount immune responses against RSV, recurrent infections are frequent throughout life. Humoral and cellular responses contribute to an effective immunity but also their localization at respiratory mucosae is increasingly recognized as an important factor. In the present study, we evaluate a mucosal vaccine based on an adenoviral vector encoding for the RSV fusion protein (Ad-F), and we investigate two genetic adjuvant candidates that encode for Interleukin (IL)-1β and IFN-β promoter stimulator I (IPS-1), respectively. While vaccination with Ad-F alone was immunogenic, the inclusion of Ad-IL-1β increased F-specific mucosal immunoglobulin A (IgA) and tissue-resident memory T cells (TRM). Consequently, immunization with Ad-F led to some control of virus replication upon RSV infection, but Ad-F+Ad-IL-1β was the most effective vaccine strategy in limiting viral load and weight loss. Subsequently, we compared the Ad-F+Ad-IL-1β-induced immunity with that provoked by a primary RSV infection. Systemic F-specific antibody responses were higher in immunized than in previously infected mice. However, the primary infection provoked glycoprotein G-specific antibodies as well eventually leading to similar neutralization titers in both groups. In contrast, mucosal antibody levels were low after infection, whereas mucosal immunization raised robust F-specific responses including IgA. Similarly, vaccination generated F-specific TRM more efficiently compared to a primary RSV infection. Although the primary infection resulted in matrix protein 2 (M2)-specific T cells as well, they did not reach levels of F-specific immunity in the vaccinated group. Moreover, the infection-induced T cell response was less biased towards TRM compared to vaccine-induced immunity. Finally, our vaccine candidate provided superior protection against RSV infection compared to a primary infection as indicated by reduced weight loss, virus replication, and tissue damage. In conclusion, our mucosal vaccine candidate Ad-F+Ad-IL-1β elicits stronger mucosal immune responses and a more effective protection against RSV infection than natural immunity generated by a previous infection. Harnessing mucosal immune responses by next-generation vaccines is therefore a promising option to establish effective RSV immunity and thereby tackle a major cause of infant hospitalization.
Collapse
Affiliation(s)
- Clara Maier
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Jana Fuchs
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Pascal Irrgang
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | | | - Jasmin Beyerlein
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Matthias Tenbusch
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany,*Correspondence: Matthias Tenbusch, ; Dennis Lapuente,
| | - Dennis Lapuente
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany,*Correspondence: Matthias Tenbusch, ; Dennis Lapuente,
| |
Collapse
|
13
|
Nair S, Wu Y, Nguyen TM, Fink K, Luo D, Ruedl C. Intranasal Delivery of RIG-I Agonist Drives Pulmonary Myeloid Cell Activation in Mice. Front Immunol 2022; 13:910192. [PMID: 35784329 PMCID: PMC9241514 DOI: 10.3389/fimmu.2022.910192] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/13/2022] [Indexed: 11/30/2022] Open
Abstract
Viral respiratory infections cause substantial health and economic burden. There is a pressing demand for efficacious vaccination strategies and, therefore, a need for a better understanding of the mechanisms of action of novel potential adjuvants. Here we investigated the effect of a synthetic RIG-I agonist, the dsRNA hairpin 3p10LA9, on the activation of pulmonary myeloid cells. Analysis of early innate immune responses revealed that a single intranasal 3p10LA9 dose induces a transient pulmonary interferon-stimulated gene (ISG) and pro-inflammatory cytokine/chemokine response, which leads to the maturation of three distinct dendritic cell subpopulations in the lungs. While lung resident dendritic cell decrease shortly after 3p10LA9 delivery, their numbers increase in the draining mediastinal lymph node, where they have migrated, maintaining their activated phenotype. At the same time, dsRNA hairpin-induced chemokines attract transiently infiltrating monocytes into the lungs, which causes a short temporary pulmonary inflammation. However, these monocytes are dispensable in controlling influenza infection since in CCR2 deficient mice, lacking these infiltrating cells, the virus load was similar to the wild type mice when infected with the influenza virus at a sublethal dose. In summary, our data suggest that intranasal delivery of dsRNA hairpins, used as a RIG-I targeting adjuvant, represents an attractive strategy to boost type I inteferon-mediated lung dendritic cell maturation, which supports viral reduction in the lungs during infection.
Collapse
Affiliation(s)
- Sajith Nair
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Yilun Wu
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Trinh Mai Nguyen
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Katja Fink
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Dahai Luo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Christiane Ruedl
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
- *Correspondence: Christiane Ruedl,
| |
Collapse
|
14
|
Jia J, Fu J, Tang H. Activation and Evasion of RLR Signaling by DNA Virus Infection. Front Microbiol 2022; 12:804511. [PMID: 34987495 PMCID: PMC8721196 DOI: 10.3389/fmicb.2021.804511] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/02/2021] [Indexed: 12/24/2022] Open
Abstract
Antiviral innate immune response triggered by nucleic acid recognition plays an extremely important role in controlling viral infections. The initiation of antiviral immune response against RNA viruses through ligand recognition of retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs) was extensively studied. RLR’s role in DNA virus infection, which is less known, is increasing attention. Here, we review the research progress of the ligand recognition of RLRs during the DNA virus infection process and the viral evasion mechanism from host immune responses.
Collapse
Affiliation(s)
- Junli Jia
- Department of Immunology, Nanjing Medical University, Nanjing, China
| | - Jiangan Fu
- Genor Biopharma Co., Ltd., Shanghai, China
| | - Huamin Tang
- Department of Immunology, Nanjing Medical University, Nanjing, China.,Laboratory Center for Basic Medical Sciences, Nanjing Medical University, Nanjing, China.,Key Laboratory of Antibody Technique of Ministry of Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
15
|
Preclinical evaluation of a candidate naked plasmid DNA vaccine against SARS-CoV-2. NPJ Vaccines 2021; 6:156. [PMID: 34930909 PMCID: PMC8688418 DOI: 10.1038/s41541-021-00419-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 11/24/2021] [Indexed: 11/12/2022] Open
Abstract
New generation plasmid DNA vaccines may be a safe, fast and simple emergency vaccine platform for preparedness against emerging viral pathogens. Applying platform optimization strategies, we tested the pre-clinical immunogenicity and protective effect of a candidate DNA plasmid vaccine specific for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The DNA vaccine induced spike-specific binding IgG and neutralizing antibodies in mice, rabbits, and rhesus macaques together with robust Th1 dominant cellular responses in small animals. Intradermal and intramuscular needle-free administration of the DNA vaccine yielded comparable immune responses. In a vaccination-challenge study of rhesus macaques, the vaccine demonstrated protection from viral replication in the lungs following intranasal and intratracheal inoculation with SARS-CoV-2. In conclusion, the candidate plasmid DNA vaccine encoding the SARS-CoV-2 spike protein is immunogenic in different models and confers protection against lung infection in nonhuman primates. Further evaluation of this DNA vaccine candidate in clinical trials is warranted.
Collapse
|
16
|
Thoresen D, Wang W, Galls D, Guo R, Xu L, Pyle AM. The molecular mechanism of RIG-I activation and signaling. Immunol Rev 2021; 304:154-168. [PMID: 34514601 PMCID: PMC9293153 DOI: 10.1111/imr.13022] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 08/10/2021] [Accepted: 08/17/2021] [Indexed: 12/25/2022]
Abstract
RIG‐I is our first line of defense against RNA viruses, serving as a pattern recognition receptor that identifies molecular features common among dsRNA and ssRNA viral pathogens. RIG‐I is maintained in an inactive conformation as it samples the cellular space for pathogenic RNAs. Upon encounter with the triphosphorylated terminus of blunt‐ended viral RNA duplexes, the receptor changes conformation and releases a pair of signaling domains (CARDs) that are selectively modified and interact with an adapter protein (MAVS), thereby triggering a signaling cascade that stimulates transcription of interferons. Here, we describe the structural determinants for specific RIG‐I activation by viral RNA, and we describe the strategies by which RIG‐I remains inactivated in the presence of host RNAs. From the initial RNA triggering event to the final stages of interferon expression, we describe the experimental evidence underpinning our working knowledge of RIG‐I signaling. We draw parallels with behavior of related proteins MDA5 and LGP2, describing evolutionary implications of their collective surveillance of the cell. We conclude by describing the cell biology and immunological investigations that will be needed to accurately describe the role of RIG‐I in innate immunity and to provide the necessary foundation for pharmacological manipulation of this important receptor.
Collapse
Affiliation(s)
- Daniel Thoresen
- Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Wenshuai Wang
- Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Drew Galls
- Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Rong Guo
- Chemistry, Yale University, New Haven, CT, USA
| | - Ling Xu
- Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Anna Marie Pyle
- Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA.,Chemistry, Yale University, New Haven, CT, USA.,Howard Hughes Medical Institute, Yale University, New Haven, CT, USA
| |
Collapse
|
17
|
Abbasi S, Uchida S. Multifunctional Immunoadjuvants for Use in Minimalist Nucleic Acid Vaccines. Pharmaceutics 2021; 13:644. [PMID: 34062771 PMCID: PMC8147386 DOI: 10.3390/pharmaceutics13050644] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/25/2021] [Accepted: 04/27/2021] [Indexed: 12/13/2022] Open
Abstract
Subunit vaccines based on antigen-encoding nucleic acids have shown great promise for antigen-specific immunization against cancer and infectious diseases. Vaccines require immunostimulatory adjuvants to activate the innate immune system and trigger specific adaptive immune responses. However, the incorporation of immunoadjuvants into nonviral nucleic acid delivery systems often results in fairly complex structures that are difficult to mass-produce and characterize. In recent years, minimalist approaches have emerged to reduce the number of components used in vaccines. In these approaches, delivery materials, such as lipids and polymers, and/or pDNA/mRNA are designed to simultaneously possess several functionalities of immunostimulatory adjuvants. Such multifunctional immunoadjuvants encode antigens, encapsulate nucleic acids, and control their pharmacokinetic or cellular fate. Herein, we review a diverse class of multifunctional immunoadjuvants in nucleic acid subunit vaccines and provide a detailed description of their mechanisms of adjuvanticity and induction of specific immune responses.
Collapse
Affiliation(s)
- Saed Abbasi
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Satoshi Uchida
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
- Medical Chemistry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 1-5 Shimogamohangi-cho, Sakyo-ku, Kyoto 606-0823, Japan
| |
Collapse
|
18
|
Behzadi M, Vakili B, Ebrahiminezhad A, Nezafat N. Iron nanoparticles as novel vaccine adjuvants. Eur J Pharm Sci 2021; 159:105718. [PMID: 33465476 DOI: 10.1016/j.ejps.2021.105718] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 12/12/2022]
Abstract
The poor immunogenicity of peptide vaccines compared to conventional ones re usually improved by applying different adjuvants. As chemical or biological substances, adjuvants are added to vaccines to enhance and prolong the immune response. According to considerable investigations over the recent years in the context of finding new adjuvants, a handful of vaccine adjuvants have been licensed for human use. Recently, engineered nanoparticles (NPs) have been introduced as novel alternatives to traditional vaccine adjuvant. Metallic nanoparticles (MeNPs) are among the most promising NPs used for vaccine adjuvant as well as the delivery system that can improve immune responses against pathogens. Iron NPs, as an important class of MeNPs, have gained increasing attention as novel vaccine adjuvants. These particles have shown acceptable results in preclinical studies. Hence, understanding the physicochemical properties of iron NPs, including size, surface properties, charge and route of administration, is of substantial importance. The aim of this review is to provide an overview of the immunomodulatory effects of iron NPs as novel adjuvants. Furthermore, physicochemical properties of these NPs were also discussed.
Collapse
Affiliation(s)
- Maryam Behzadi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bahareh Vakili
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alireza Ebrahiminezhad
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Navid Nezafat
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
19
|
Abstract
The field of bio-nano interfaces paves the way for a better understanding, development, and implementation of the advanced biotechnological process. Interfacing biomolecules with the nanomaterials will result in the development of new tools and techniques that, in turn, will enable to explore the fundamental process at the nano level and fabricate cost-effective portable devices. Fascinating biomolecules like DNA, RNA and proteins in the regime of nanoscale are intelligent materials that are capable of storing the information and controlling the basic structure and function of the complex biological systems. Following this concept, the current pandemic situation would be a natural selection process, where the selective pressure is on the ssRNA of Covid-19 to choose the suitable progeny for survival. Consequently, the interaction of human DNA invoking response with Covid-19 happens at the nanoscale and it could be a better candidate to provoke combat against the virus. The extent of this interaction would give us the insights at the nanotechnological level to tackle the prevention, diagnosis and treatment for Covid-19. Herein, the possible features and obstacles in Covid-19 and a probable solution from the advent of nanotechnology are discussed to address the current necessity. Moreover, the perspective sustainable green graph mask that can be prepared using green plant extract/graphene (Bio-Nano composite mask) is suggested for the possible protection of virus-like Covid-19. The composite material will not only effectively trap the virus but also inactivate the virus due to the presence of antiviral compounds in the plant extracts.
Collapse
|
20
|
Abstract
Retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs) are key sensors of virus infection, mediating the transcriptional induction of type I interferons and other genes that collectively establish an antiviral host response. Recent studies have revealed that both viral and host-derived RNAs can trigger RLR activation; this can lead to an effective antiviral response but also immunopathology if RLR activities are uncontrolled. In this Review, we discuss recent advances in our understanding of the types of RNA sensed by RLRs in the contexts of viral infection, malignancies and autoimmune diseases. We further describe how the activity of RLRs is controlled by host regulatory mechanisms, including RLR-interacting proteins, post-translational modifications and non-coding RNAs. Finally, we discuss key outstanding questions in the RLR field, including how our knowledge of RLR biology could be translated into new therapeutics.
Collapse
Affiliation(s)
- Jan Rehwinkel
- Medical Research Council Human Immunology Unit, Medical Research Council Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
| | - Michaela U Gack
- Department of Microbiology, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
21
|
Jung HE, Lee HK. Host Protective Immune Responses against Influenza A Virus Infection. Viruses 2020; 12:v12050504. [PMID: 32375274 PMCID: PMC7291249 DOI: 10.3390/v12050504] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 04/27/2020] [Accepted: 04/30/2020] [Indexed: 12/31/2022] Open
Abstract
Influenza viruses cause infectious respiratory disease characterized by fever, myalgia, and congestion, ranging in severity from mild to life-threating. Although enormous efforts have aimed to prevent and treat influenza infections, seasonal and pandemic influenza outbreaks remain a major public health concern. This is largely because influenza viruses rapidly undergo genetic mutations that restrict the long-lasting efficacy of vaccine-induced immune responses and therapeutic regimens. In this review, we discuss the virological features of influenza A viruses and provide an overview of current knowledge of the innate sensing of invading influenza viruses and the protective immune responses in the host.
Collapse
Affiliation(s)
- Hi Eun Jung
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
- Correspondence: (H.E.J.); (H.K.L.); Tel.: +82-42-350-4281 (H.K.L.)
| | - Heung Kyu Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
- KAIST Institute for Health Science and Technology, KAIST, Daejeon 34141, Korea
- Correspondence: (H.E.J.); (H.K.L.); Tel.: +82-42-350-4281 (H.K.L.)
| |
Collapse
|
22
|
Suschak JJ, Dupuy LC, Shoemaker CJ, Six C, Kwilas SA, Spik KW, Williams JA, Schmaljohn CS. Nanoplasmid Vectors Co-expressing Innate Immune Agonists Enhance DNA Vaccines for Venezuelan Equine Encephalitis Virus and Ebola Virus. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 17:810-821. [PMID: 32296729 PMCID: PMC7158766 DOI: 10.1016/j.omtm.2020.04.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 04/13/2020] [Indexed: 01/04/2023]
Abstract
DNA vaccines expressing codon-optimized Venezuelan equine encephalitis virus (VEEV) and Ebola virus (EBOV) glycoprotein genes provide protective immunity to mice and nonhuman primates when delivered by intramuscular (IM) electroporation (EP). To achieve equivalent protective efficacy in the absence of EP, we evaluated VEEV and EBOV DNA vaccines constructed using minimalized Nanoplasmid expression vectors that are smaller than conventional plasmids used for DNA vaccination. These vectors may also be designed to co-express type I interferon inducing innate immune agonist genes that have an adjuvant effect. Nanoplasmid vaccinated mice had increased antibody responses as compared to those receiving our conventional pWRG7077-based vaccines when delivered by IM injection, and these responses were further enhanced by the inclusion of the innate immune agonist genes. The Nanoplasmid VEEV DNA vaccines also significantly increased protection against aerosol VEEV challenge as compared to the pWRG7077 VEEV DNA vaccine. Although all mice receiving the pWRG7077 and Nanoplasmid EBOV DNA vaccines at the dose tested survived EBOV challenge, only mice receiving the Nanoplasmid EBOV DNA vaccine that co-expresses the innate immune agonist genes failed to lose weight after challenge. Our results suggest that Nanoplasmid vectors can improve the immunogenicity and protective efficacy of alphavirus and filovirus DNA vaccines.
Collapse
Affiliation(s)
- John J Suschak
- Virology Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | - Lesley C Dupuy
- Virology Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | - Charles J Shoemaker
- Diagnostic Systems Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | - Carolyn Six
- Virology Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | - Steven A Kwilas
- Virology Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | - Kristin W Spik
- Virology Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | | | - Connie S Schmaljohn
- Headquarters, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| |
Collapse
|
23
|
Lapuente D, Stab V, Storcksdieck Genannt Bonsmann M, Maaske A, Köster M, Xiao H, Ehrhardt C, Tenbusch M. Innate signalling molecules as genetic adjuvants do not alter the efficacy of a DNA-based influenza A vaccine. PLoS One 2020; 15:e0231138. [PMID: 32243477 PMCID: PMC7122823 DOI: 10.1371/journal.pone.0231138] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 03/14/2020] [Indexed: 01/07/2023] Open
Abstract
In respect to the heterogeneity among influenza A virus strains and the shortcomings of current vaccination programs, there is a huge interest in the development of alternative vaccines that provide a broader and more long-lasting protection. Gene-based approaches are considered as promising candidates for such flu vaccines. In our study, innate signalling molecules from the RIG-I and the NALP3 pathways were evaluated as genetic adjuvants in intramuscular DNA immunizations. Plasmids encoding a constitutive active form of RIG-I (cRIG-I), IPS-1, IL-1β, or IL-18 were co-administered with plasmids encoding the hemagglutinin and nucleoprotein derived from H1N1/Puerto Rico/8/1934 via electroporation in BALB/c mice. Immunogenicity was analysed in detail and efficacy was demonstrated in homologous and heterologous influenza challenge experiments. Although the biological activities of the adjuvants have been confirmed by in vitro reporter assays, their single or combined inclusion in the vaccine did not result in superior vaccine efficacy. With the exception of significantly increased levels of antigen-specific IgG1 after the co-administration of IL-1β, there were only minor alterations concerning the immunogenicity. Since DNA electroporation alone induced substantial inflammation at the injection site, as demonstrated in this study using Mx2-Luc reporter mice, it might override the adjuvants´ contribution to the inflammatory microenvironment and thereby minimizes the influence on the immunogenicity. Taken together, the DNA immunization was protective against subsequent challenge infections but could not be further improved by the genetic adjuvants analysed in this study.
Collapse
Affiliation(s)
- Dennis Lapuente
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Viktoria Stab
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum, Germany
| | | | - Andre Maaske
- Environmental Medicine, UNIKA-T Augsburg, Technische Universität München and Helmholtz Zentrum, Neuherberg, Germany
| | - Mario Köster
- Model Systems for Infection and Immunity, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Han Xiao
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Christina Ehrhardt
- Section of Experimental Virology, Institute of Medical Microbiology, University Hospital Jena, Jena, Germany
| | - Matthias Tenbusch
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
24
|
Bernelin-Cottet C, Urien C, Fretaud M, Langevin C, Trus I, Jouneau L, Blanc F, Leplat JJ, Barc C, Boulesteix O, Riou M, Dysart M, Mahé S, Studsrub E, Nauwynck H, Bertho N, Bourry O, Schwartz-Cornil I. A DNA Prime Immuno-Potentiates a Modified Live Vaccine against the Porcine Reproductive and Respiratory Syndrome Virus but Does Not Improve Heterologous Protection. Viruses 2019; 11:E576. [PMID: 31242645 PMCID: PMC6631340 DOI: 10.3390/v11060576] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 06/18/2019] [Accepted: 06/21/2019] [Indexed: 12/24/2022] Open
Abstract
The porcine reproductive and respiratory syndrome virus (PRRSV), an RNA virus inducing abortion in sows and respiratory disease in young pigs, is a leading infectious cause of economic losses in the swine industry. Modified live vaccines (MLVs) help in controlling the disease, but their efficacy is often compromised by the high genetic diversity of circulating viruses, leading to vaccine escape variants in the field. In this study, we hypothesized that a DNA prime with naked plasmids encoding PRRSV antigens containing conserved T-cell epitopes may improve the protection of MLV against a heterologous challenge. Plasmids were delivered with surface electroporation or needle-free jet injection and European strain-derived PRRSV antigens were targeted or not to the dendritic cell receptor XCR1. Compared to MLV-alone, the DNA-MLV prime- boost regimen slightly improved the IFNγ T-cell response, and substantially increased the antibody response against envelope motives and the nucleoprotein N. The XCR1-targeting of N significantly improved the anti-N specific antibody response. Despite this immuno-potentiation, the DNA-MLV regimen did not further decrease the serum viral load or the nasal viral shedding of the challenge strain over MLV-alone. Finally, the heterologous protection, achieved in absence of detectable effective neutralizing antibodies, was not correlated to the measured antibody or to the IFNγ T-cell response. Therefore, immune correlates of protection remain to be identified and represent an important gap of knowledge in PRRSV vaccinology. This study importantly shows that a naked DNA prime immuno-potentiates an MLV, more on the B than on the IFNγ T-cell response side, and has to be further improved to reach cross-protection.
Collapse
Affiliation(s)
- Cindy Bernelin-Cottet
- VIM, INRA, Université Paris-Saclay, Domaine de Vilvert, 78350 Jouy-en-Josas, France.
| | - Céline Urien
- VIM, INRA, Université Paris-Saclay, Domaine de Vilvert, 78350 Jouy-en-Josas, France.
| | - Maxence Fretaud
- VIM, INRA, Université Paris-Saclay, Domaine de Vilvert, 78350 Jouy-en-Josas, France.
| | - Christelle Langevin
- VIM, INRA, Université Paris-Saclay, Domaine de Vilvert, 78350 Jouy-en-Josas, France.
- VIM, EMERG'IN-Plateforme d'Infectiologie Expérimentale IERP, INRA, Domaine de Vilvert, 78352 Jouy-en-Josas, France.
| | - Ivan Trus
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820 Merelbeke, Belgium.
| | - Luc Jouneau
- VIM, INRA, Université Paris-Saclay, Domaine de Vilvert, 78350 Jouy-en-Josas, France.
| | - Fany Blanc
- GABI, INRA-AgroParisTech, Université Paris-Saclay, Domaine de Vilvert, 78350 Jouy-en-Josas, France.
| | - Jean-Jacques Leplat
- GABI, INRA-AgroParisTech, Université Paris-Saclay, Domaine de Vilvert, 78350 Jouy-en-Josas, France.
| | - Céline Barc
- Plate-Forme d'Infectiologie Expérimentale-PFIE-UE1277, Centre Val de Loire, INRA, 37380 Nouzilly, France.
| | - Olivier Boulesteix
- Plate-Forme d'Infectiologie Expérimentale-PFIE-UE1277, Centre Val de Loire, INRA, 37380 Nouzilly, France.
| | - Mickaël Riou
- Plate-Forme d'Infectiologie Expérimentale-PFIE-UE1277, Centre Val de Loire, INRA, 37380 Nouzilly, France.
| | - Marilyn Dysart
- Pharmajet, 400 Corporate Circle Suite N, Golden, CO 80401, USA.
| | - Sophie Mahé
- Unité Virologie et Immunologie Porcines, Laboratoire de Ploufragan-Plouzané-Niort, Anses, BP 53, 22440 Ploufragan, France.
| | | | - Hans Nauwynck
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820 Merelbeke, Belgium.
| | - Nicolas Bertho
- VIM, INRA, Université Paris-Saclay, Domaine de Vilvert, 78350 Jouy-en-Josas, France.
| | - Olivier Bourry
- Unité Virologie et Immunologie Porcines, Laboratoire de Ploufragan-Plouzané-Niort, Anses, BP 53, 22440 Ploufragan, France.
| | | |
Collapse
|
25
|
Hou J, Shrivastava S, Fraser CC, Loo HL, Wong LH, Ho V, Fink K, Ooi EE, Chen J. Dengue Mosaic Vaccines Enhance Cellular Immunity and Expand the Breadth of Neutralizing Antibody Against All Four Serotypes of Dengue Viruses in Mice. Front Immunol 2019; 10:1429. [PMID: 31281322 PMCID: PMC6596366 DOI: 10.3389/fimmu.2019.01429] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 06/06/2019] [Indexed: 11/13/2022] Open
Abstract
An estimated 400 million people in the world are infected with any of the four types of dengue virus (DENV) annually. The only licensed dengue vaccine cannot effectively prevent infection with all of the four DENVs, especially in those immunologically naïve at baseline. In this study, we explored a mosaic vaccine approach, which utilizes an artificial recombinant sequence for each serotype to achieve maximum coverage of variant epitopes in the four DENVs. We determined the immunogenicity and cross-reactivity of DNA plasmids encoding individual mosaic sequences or the natural sequences in mice. We show that the mosaic vaccines, particularly those targeting DENV serotype 1 and 2, improved vaccine immunogenicity by increasing the percentage of antigen-specific IFNγ- or TNFα-secreting CD4 and CD8 T cells, and titers of neutralizing antibodies. The mosaic vaccine diversified and broadened anti-dengue T cell responses and cross-reactive neutralizing antibodies against all four serotypes. The mosaic vaccines also induced higher level of antigen-specific B cell responses. These results suggest that mosaic vaccines comprising of DENV serotype 1 and 2 variant epitopes could stimulate strong and broad immune responses against all four serotypes.
Collapse
Affiliation(s)
- Jue Hou
- Interdisciplinary Research Group in Infectious Diseases, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
| | - Shubham Shrivastava
- Interdisciplinary Research Group in Infectious Diseases, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
| | - Christopher C Fraser
- Interdisciplinary Research Group in Infectious Diseases, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
| | - Hooi Linn Loo
- Interdisciplinary Research Group in Infectious Diseases, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
| | - Lan Hiong Wong
- Interdisciplinary Research Group in Infectious Diseases, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
| | - Victor Ho
- Singapore Immunology Network, AStar, Singapore, Singapore
| | - Katja Fink
- Singapore Immunology Network, AStar, Singapore, Singapore
| | - Eng Eong Ooi
- Interdisciplinary Research Group in Infectious Diseases, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore.,Emerging Infectious Diseases Program, Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Jianzhu Chen
- Interdisciplinary Research Group in Infectious Diseases, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore.,Koch Institute for Integrative Cancer Research and Departments of Biology, Massachusetts Institute of Technology, Cambridge, MA, United States
| |
Collapse
|
26
|
Thalmensi J, Pliquet E, Liard C, Chamel G, Kreuz C, Bestetti T, Escande M, Kostrzak A, Pailhes-Jimenez AS, Bourges E, Julithe M, Bourre L, Keravel O, Clayette P, Huet T, Wain-Hobson S, Langlade-Demoyen P. A DNA telomerase vaccine for canine cancer immunotherapy. Oncotarget 2019; 10:3361-3372. [PMID: 31164958 PMCID: PMC6534364 DOI: 10.18632/oncotarget.26927] [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: 05/20/2016] [Accepted: 04/29/2019] [Indexed: 12/23/2022] Open
Abstract
Telomerase reverse transcriptase (TERT) is highly expressed in more than 90% of canine cancer cells and low to absent in normal cells. Given that immune tolerance to telomerase is easily broken both naturally and experimentally, telomerase is an attractive tumor associated antigen for cancer immunotherapy. Indeed, therapeutic trials using human telomerase peptides have been performed. We have developed an immunogenic yet catalytically inactive human telomerase DNA construct that is in clinical trials with patients presenting solid tumors. Paralleling this human construct, we have developed a canine telomerase DNA vaccine, called pDUV5. When administered intradermally to mice combined with electrogene transfer, pDUV5 induced canine TERT specific cytotoxic T-cells as measured by IFN-γ ELISpot assay. Intradermal vaccination of healthy dogs with 400 μg of pDUV5 generated strong, broad and long lasting TERT specific cellular immune responses. In vitro immunization with cTERT peptides revealed the maintenance of cTERT specific T-cells in PBMCs from tumor bearing dogs showing that this repertoire was not depleted. This study highlights the potential of pDUV5 as a cancer vaccine and supports its evaluation for the treatment of spontaneous canine tumors.
Collapse
Affiliation(s)
| | | | | | | | - Christine Kreuz
- ImmunoPharmacology and Biosafety Lab, Bertin Pharma/CEA, Fontenay-aux-Roses 92265, France
| | | | | | | | | | | | | | | | | | - Pascal Clayette
- ImmunoPharmacology and Biosafety Lab, Bertin Pharma/CEA, Fontenay-aux-Roses 92265, France
| | | | - Simon Wain-Hobson
- Invectys, Paris BioPark, Paris 75013, France.,Molecular Retrovirology Unit, Institut Pasteur, CNRS-URA 3015, Paris 75015, France
| | - Pierre Langlade-Demoyen
- Invectys, Paris BioPark, Paris 75013, France.,Molecular Retrovirology Unit, Institut Pasteur, CNRS-URA 3015, Paris 75015, France
| |
Collapse
|
27
|
Aderibigbe BA, Naki T. Design and Efficacy of Nanogels Formulations for Intranasal Administration. Molecules 2018; 23:E1241. [PMID: 29789506 PMCID: PMC6100477 DOI: 10.3390/molecules23061241] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 04/27/2018] [Accepted: 05/02/2018] [Indexed: 12/12/2022] Open
Abstract
Nanogels are drug delivery systems that can bypass the blood-brain barrier and deliver drugs to the desired site when administered intranasally. They have been used as a drug delivery platform for the management of brain diseases such as Alzheimer disease, migraine, schizophrenia and depression. nanogels have also been developed as vaccine carriers for the protection of bacterial infections such as influenza, meningitis, pneumonia and as veterinary vaccine carriers for the protection of animals from encephalomyelitis and mouth to foot disease. It has been developed as vaccine carriers for the prevention of lifestyle disease such as obesity. Intranasal administration of therapeutics using nanogels for the management of brain diseases revealed that the drug transportation was via the olfactory nerve pathway resulting in rapid drug delivery to the brain with excellent neuroprotective effect. The application of nanogels as vaccine carriers also induced significant responses associated with protective immunity against selected bacterial and viral infections. This review provides a detailed information on the enhanced therapeutic effects, mechanisms and biological efficacy of nanogels for intranasal administration.
Collapse
Affiliation(s)
- Blessing A Aderibigbe
- Department of Chemistry, University of Fort Hare, Alice Campus, Eastern Cape 5700, South Africa.
| | - Tobeka Naki
- Department of Chemistry, University of Fort Hare, Alice Campus, Eastern Cape 5700, South Africa.
| |
Collapse
|
28
|
Suschak JJ, Williams JA, Schmaljohn CS. Advancements in DNA vaccine vectors, non-mechanical delivery methods, and molecular adjuvants to increase immunogenicity. Hum Vaccin Immunother 2017. [PMID: 28604157 DOI: 10.1080/21645515.2017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023] Open
Abstract
A major advantage of DNA vaccination is the ability to induce both humoral and cellular immune responses. DNA vaccines are currently used in veterinary medicine, but have not achieved widespread acceptance for use in humans due to their low immunogenicity in early clinical studies. However, recent clinical data have re-established the value of DNA vaccines, particularly in priming high-level antigen-specific antibody responses. Several approaches have been investigated for improving DNA vaccine efficacy, including advancements in DNA vaccine vector design, the inclusion of genetically engineered cytokine adjuvants, and novel non-mechanical delivery methods. These strategies have shown promise, resulting in augmented adaptive immune responses in not only mice, but also in large animal models. Here, we review advancements in each of these areas that show promise for increasing the immunogenicity of DNA vaccines.
Collapse
Affiliation(s)
- John J Suschak
- a U.S. Army Medical Research Institute of Infectious Diseases , Fort Detrick , MD , USA
| | | | - Connie S Schmaljohn
- a U.S. Army Medical Research Institute of Infectious Diseases , Fort Detrick , MD , USA
| |
Collapse
|
29
|
Suschak JJ, Williams JA, Schmaljohn CS. Advancements in DNA vaccine vectors, non-mechanical delivery methods, and molecular adjuvants to increase immunogenicity. Hum Vaccin Immunother 2017; 13:2837-2848. [PMID: 28604157 PMCID: PMC5718814 DOI: 10.1080/21645515.2017.1330236] [Citation(s) in RCA: 163] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
A major advantage of DNA vaccination is the ability to induce both humoral and cellular immune responses. DNA vaccines are currently used in veterinary medicine, but have not achieved widespread acceptance for use in humans due to their low immunogenicity in early clinical studies. However, recent clinical data have re-established the value of DNA vaccines, particularly in priming high-level antigen-specific antibody responses. Several approaches have been investigated for improving DNA vaccine efficacy, including advancements in DNA vaccine vector design, the inclusion of genetically engineered cytokine adjuvants, and novel non-mechanical delivery methods. These strategies have shown promise, resulting in augmented adaptive immune responses in not only mice, but also in large animal models. Here, we review advancements in each of these areas that show promise for increasing the immunogenicity of DNA vaccines.
Collapse
Affiliation(s)
- John J Suschak
- a U.S. Army Medical Research Institute of Infectious Diseases , Fort Detrick , MD , USA
| | | | - Connie S Schmaljohn
- a U.S. Army Medical Research Institute of Infectious Diseases , Fort Detrick , MD , USA
| |
Collapse
|
30
|
Lazarte JMS, Kim YR, Lee JS, Im SP, Kim SW, Jung JW, Kim J, Lee JH, Jung TS. Immunostimulatory effect of DDX41 of olive flounder (Paralichthys olivaceus). FOOD AGR IMMUNOL 2017. [DOI: 10.1080/09540105.2017.1318836] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Affiliation(s)
- Jassy Mary S. Lazarte
- Laboratory of Aquatic Animal Diseases, College of Veterinary Medicine, Gyeongsang National University, Jinju, South Korea
| | - Young Rim Kim
- Laboratory of Aquatic Animal Diseases, College of Veterinary Medicine, Gyeongsang National University, Jinju, South Korea
| | - Jung Seok Lee
- Laboratory of Aquatic Animal Diseases, College of Veterinary Medicine, Gyeongsang National University, Jinju, South Korea
| | - Se Pyeong Im
- Laboratory of Aquatic Animal Diseases, College of Veterinary Medicine, Gyeongsang National University, Jinju, South Korea
| | - Si Won Kim
- Laboratory of Aquatic Animal Diseases, College of Veterinary Medicine, Gyeongsang National University, Jinju, South Korea
| | - Jae Wook Jung
- Laboratory of Aquatic Animal Diseases, College of Veterinary Medicine, Gyeongsang National University, Jinju, South Korea
| | - Jaesung Kim
- Laboratory of Aquatic Animal Diseases, College of Veterinary Medicine, Gyeongsang National University, Jinju, South Korea
| | - Jeong-Ho Lee
- Inland Aquaculture Research Center, NIFS, Changwon, Korea
| | - Tae Sung Jung
- Laboratory of Aquatic Animal Diseases, College of Veterinary Medicine, Gyeongsang National University, Jinju, South Korea
| |
Collapse
|
31
|
Probst P, Grigg JB, Wang M, Muñoz E, Loo YM, Ireton RC, Gale M, Iadonato SP, Bedard KM. A small-molecule IRF3 agonist functions as an influenza vaccine adjuvant by modulating the antiviral immune response. Vaccine 2017; 35:1964-1971. [PMID: 28279563 PMCID: PMC11514956 DOI: 10.1016/j.vaccine.2017.01.053] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 01/12/2017] [Accepted: 01/18/2017] [Indexed: 11/17/2022]
Abstract
Vaccine adjuvants are essential to drive a protective immune response in cases where vaccine antigens are weakly immunogenic, where vaccine antigen is limited, or where an increase in potency is needed for a specific population, such as the elderly. To discover novel vaccine adjuvants, we used a high-throughput screen (HTS) designed to identify small-molecule agonists of the RIG-I-like receptor (RLR) pathway leading to interferon regulatory factor 3 (IRF3) activation. RLRs are a group of cytosolic pattern-recognition receptors that are essential for the recognition of viral nucleic acids during infection. Upon binding of viral nucleic acid ligands, the RLRs become activated and signal to transcription factors, including IRF3, to initiate an innate immune transcriptional program to control virus infection. Among our HTS hits were a series of benzothiazole compounds from which we designed the lead analog, KIN1148. KIN1148 induced dose-dependent IRF3 nuclear translocation and specific activation of IRF3-responsive promoters. Prime-boost immunization of mice with a suboptimal dose of a monovalent pandemic influenza split virus H1N1 A/California/07/2009 vaccine plus KIN1148 protected against a lethal challenge with mouse-adapted influenza virus (A/California/04/2009) and induced an influenza virus-specific IL-10 and Th2 response by T cells derived from lung and lung-draining lymph nodes. Prime-boost immunization with vaccine plus KIN1148, but not prime immunization alone, induced antibodies capable of inhibiting influenza virus hemagglutinin and neutralizing viral infectivity. Nevertheless, a single immunization with vaccine plus KIN1148 provided increased protection over vaccine alone and reduced viral load in the lungs after challenge. These findings suggest that protection was at least partially mediated by a cellular immune component and that the induction of Th2 and immunoregulatory cytokines by a KIN1148-adjuvanted vaccine may be particularly beneficial for ameliorating the immunopathogenesis that is associated with influenza viruses.
Collapse
Affiliation(s)
- Peter Probst
- Kineta, Inc., 219 Terry Ave. N., Seattle, WA 98109, USA
| | - John B Grigg
- Kineta, Inc., 219 Terry Ave. N., Seattle, WA 98109, USA
| | - Myra Wang
- Kineta, Inc., 219 Terry Ave. N., Seattle, WA 98109, USA
| | - Ernesto Muñoz
- Kineta, Inc., 219 Terry Ave. N., Seattle, WA 98109, USA
| | - Yueh-Ming Loo
- Department of Immunology, University of Washington, Seattle, WA 98195, USA; Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA 98195, USA
| | - Renee C Ireton
- Department of Immunology, University of Washington, Seattle, WA 98195, USA; Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA 98195, USA
| | - Michael Gale
- Department of Immunology, University of Washington, Seattle, WA 98195, USA; Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA 98195, USA
| | | | | |
Collapse
|
32
|
Pushko P, Lukashevich IS, Weaver SC, Tretyakova I. DNA-launched live-attenuated vaccines for biodefense applications. Expert Rev Vaccines 2016; 15:1223-34. [PMID: 27055100 PMCID: PMC5033646 DOI: 10.1080/14760584.2016.1175943] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A novel vaccine platform uses DNA immunization to launch live-attenuated virus vaccines in vivo. This technology has been applied for vaccine development against positive-strand RNA viruses with global public health impact including alphaviruses and flaviviruses. The DNA-launched vaccine represents the recombinant plasmid that encodes the full-length genomic RNA of live-attenuated virus downstream from a eukaryotic promoter. When administered in vivo, the genomic RNA of live-attenuated virus is transcribed. The RNA initiates limited replication of a genetically defined, live-attenuated vaccine virus in the tissues of the vaccine recipient, thereby inducing a protective immune response. This platform combines the strengths of reverse genetics, DNA immunization and the advantages of live-attenuated vaccines, resulting in a reduced chance of genetic reversions, increased safety, and improved immunization. With this vaccine technology, the field of DNA vaccines is expanded from those that express subunit antigens to include a novel type of DNA vaccines that launch live-attenuated viruses.
Collapse
Affiliation(s)
- Peter Pushko
- Medigen, Inc. 8420 Gas House Pike Suite S, Frederick, MD 21701, USA
| | - Igor S. Lukashevich
- Department of Pharmacology and Toxicology, School of Medicine, Center for Predictive Medicine and Emerging Infectious Diseases, University of Louisville, 505 S Hancock St., Louisville, KY 40202, USA
| | - Scott C. Weaver
- Institute for Human Infections and Immunity, Sealy Center for Vaccine Development and Department of Microbiology and Immunology, University of Texas Medical Branch, GNL, 301 University Blvd., Galveston, TX 77555, USA
| | - Irina Tretyakova
- Medigen, Inc. 8420 Gas House Pike Suite S, Frederick, MD 21701, USA
| |
Collapse
|
33
|
Abstract
DNA plasmids can be used to induce a protective (or therapeutic) immune response by delivering genes encoding vaccine antigens. That naked DNA (without the refinement of coat proteins or host evasion systems) can cross from outside the cell into the nucleus and be expressed is particularly remarkable given the sophistication of the immune system in preventing infection by pathogens. As a result of the ease, low cost, and speed of custom gene synthesis, DNA vaccines dangle a tantalizing prospect of the next wave of vaccine technology, promising individual designer vaccines for cancer or mass vaccines with a rapid response time to emerging pandemics. There is considerable enthusiasm for the use of DNA vaccination as an approach, but this enthusiasm should be tempered by the successive failures in clinical trials to induce a potent immune response. The technology is evolving with the development of improved delivery systems that increase expression levels, particularly electroporation and the incorporation of genetically encoded adjuvants. This review will introduce some key concepts in the use of DNA plasmids as vaccines, including how the DNA enters the cell and is expressed, how it induces an immune response, and a summary of clinical trials with DNA vaccines. The review also explores the advances being made in vector design, delivery, formulation, and adjuvants to try to realize the promise of this technology for new vaccines. If the immunogenicity and expression barriers can be cracked, then DNA vaccines may offer a step change in mass vaccination.
Collapse
|
34
|
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
|
35
|
Bryant CE, Orr S, Ferguson B, Symmons MF, Boyle JP, Monie TP. International Union of Basic and Clinical Pharmacology. XCVI. Pattern recognition receptors in health and disease. Pharmacol Rev 2015; 67:462-504. [PMID: 25829385 DOI: 10.1124/pr.114.009928] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Since the discovery of Toll, in the fruit fly Drosophila melanogaster, as the first described pattern recognition receptor (PRR) in 1996, many families of these receptors have been discovered and characterized. PRRs play critically important roles in pathogen recognition to initiate innate immune responses that ultimately link to the generation of adaptive immunity. Activation of PRRs leads to the induction of immune and inflammatory genes, including proinflammatory cytokines and chemokines. It is increasingly clear that many PRRs are linked to a range of inflammatory, infectious, immune, and chronic degenerative diseases. Several drugs to modulate PRR activity are already in clinical trials and many more are likely to appear in the near future. Here, we review the different families of mammalian PRRs, the ligands they recognize, the mechanisms of activation, their role in disease, and the potential of targeting these proteins to develop the anti-inflammatory therapeutics of the future.
Collapse
Affiliation(s)
- Clare E Bryant
- Departments of Veterinary Medicine (C.E.B., J.P.B., T.P.M.), Pathology (B.F.), and Biochemistry (M.F.S., J.P.B.), University of Cambridge, Cambridge, United Kingdom; and Institute of Infection and Immunity, Cardiff University, Cardiff, United Kingdom (S.O.)
| | - Selinda Orr
- Departments of Veterinary Medicine (C.E.B., J.P.B., T.P.M.), Pathology (B.F.), and Biochemistry (M.F.S., J.P.B.), University of Cambridge, Cambridge, United Kingdom; and Institute of Infection and Immunity, Cardiff University, Cardiff, United Kingdom (S.O.)
| | - Brian Ferguson
- Departments of Veterinary Medicine (C.E.B., J.P.B., T.P.M.), Pathology (B.F.), and Biochemistry (M.F.S., J.P.B.), University of Cambridge, Cambridge, United Kingdom; and Institute of Infection and Immunity, Cardiff University, Cardiff, United Kingdom (S.O.)
| | - Martyn F Symmons
- Departments of Veterinary Medicine (C.E.B., J.P.B., T.P.M.), Pathology (B.F.), and Biochemistry (M.F.S., J.P.B.), University of Cambridge, Cambridge, United Kingdom; and Institute of Infection and Immunity, Cardiff University, Cardiff, United Kingdom (S.O.)
| | - Joseph P Boyle
- Departments of Veterinary Medicine (C.E.B., J.P.B., T.P.M.), Pathology (B.F.), and Biochemistry (M.F.S., J.P.B.), University of Cambridge, Cambridge, United Kingdom; and Institute of Infection and Immunity, Cardiff University, Cardiff, United Kingdom (S.O.)
| | - Tom P Monie
- Departments of Veterinary Medicine (C.E.B., J.P.B., T.P.M.), Pathology (B.F.), and Biochemistry (M.F.S., J.P.B.), University of Cambridge, Cambridge, United Kingdom; and Institute of Infection and Immunity, Cardiff University, Cardiff, United Kingdom (S.O.)
| |
Collapse
|
36
|
R El-Attar LM, Thomas C, Luke J, A Williams J, Brownlie J. Enhanced neutralising antibody response to bovine viral diarrhoea virus (BVDV) induced by DNA vaccination in calves. Vaccine 2015; 33:4004-12. [PMID: 26079613 DOI: 10.1016/j.vaccine.2015.06.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 05/28/2015] [Accepted: 06/02/2015] [Indexed: 11/26/2022]
Abstract
DNA vaccination is effective in inducing potent immunity in mice; however it appears to be less so in large animals. Increasing the dose of DNA plasmid to activate innate immunity has been shown to improve DNA vaccine adaptive immunity. Retinoic acid-inducible gene I (RIG-I) is a critical cytoplasmic double-stranded RNA pattern receptor required for innate immune activation in response to viral infection. RIG-I recognise viral RNA and trigger antiviral response, resulting in type I interferon (IFN) and inflammatory cytokine production. In an attempt to enhance the antibody response induced by BVDV DNA in cattle, we expressed BVDV truncated E2 (E2t) and NS3 codon optimised antigens from antibiotic free-plasmid vectors expressing a RIG-I agonist and designated either NTC E2t(co) and NTC NS3(co). To evaluate vaccine efficacy, groups of five BVDV-free calves were intramuscularly injected three times with NTC E2t(co) and NTC NS3(co) vaccine plasmids individually or in combination. Animals vaccinated with our (previously published) conventional DNA vaccines pSecTag/E2 and pTriExNS3 and plasmids expressing RIG-I agonist only presented both the positive and mock-vaccine groups. Our results showed that vaccines coexpressing E2t with a RIG-I agonist induced significantly higher E2 antigen specific antibody response (p<0.05). Additionally, E2t augmented the immune response to NS3 when the two vaccines were delivered in combination. Despite the lack of complete protection, on challenge day 4/5 calves vaccinated with NTC E2t(co) alone or NTC E2t(co) plus NTC NS3(co) had neutralising antibody titres exceeding 1/240 compared to 1/5 in the mock vaccine control group. Based on our results we conclude that co-expression of a RIG-I agonist with viral antigen could enhance DNA vaccine potency in cattle.
Collapse
Affiliation(s)
- Laila M R El-Attar
- Pathology & Pathogen Biology, Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield AL9 7TA, Hertfordshire, UK.
| | - Carole Thomas
- Pathology & Pathogen Biology, Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield AL9 7TA, Hertfordshire, UK
| | - Jeremy Luke
- Nature Technology Corporation, 4701 Innovation Drive, Lincoln, NE 68521, USA
| | - James A Williams
- Nature Technology Corporation, 4701 Innovation Drive, Lincoln, NE 68521, USA
| | - Joe Brownlie
- Pathology & Pathogen Biology, Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield AL9 7TA, Hertfordshire, UK
| |
Collapse
|
37
|
Grunwald T, Ulbert S. Improvement of DNA vaccination by adjuvants and sophisticated delivery devices: vaccine-platforms for the battle against infectious diseases. Clin Exp Vaccine Res 2015; 4:1-10. [PMID: 25648133 PMCID: PMC4313101 DOI: 10.7774/cevr.2015.4.1.1] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 11/30/2014] [Accepted: 12/31/2014] [Indexed: 01/24/2023] Open
Abstract
Advantages of DNA vaccination against infectious diseases over more classical immunization methods include the possibilities for rapid manufacture, fast adaptation to newly emerging pathogens and high stability at ambient temperatures. In addition, upon DNA immunization the antigen is produced by the cells of the vaccinated individual, which leads to activation of both cellular and humoral immune responses due to antigen presentation via MHC I and MHC II molecules. However, so far DNA vaccines have shown most efficient immunogenicity mainly in small rodent models, whereas in larger animals including humans there is still the need to improve effectiveness. This is mostly due to inefficient delivery of the DNA plasmid into cells and nuclei. Here, we discuss technologies used to overcome this problem, including physical means such as in vivo electroporation and co-administration of adjuvants. Several of these methods have already entered clinical testing in humans.
Collapse
Affiliation(s)
- Thomas Grunwald
- Department of Immunology, Fraunhofer-Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Sebastian Ulbert
- Department of Immunology, Fraunhofer-Institute for Cell Therapy and Immunology, Leipzig, Germany
| |
Collapse
|
38
|
Kim MG, Park JY, Shon Y, Kim G, Shim G, Oh YK. Nanotechnology and vaccine development. Asian J Pharm Sci 2014. [DOI: 10.1016/j.ajps.2014.06.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
39
|
Activation of the RIG-I pathway during influenza vaccination enhances the germinal center reaction, promotes T follicular helper cell induction, and provides a dose-sparing effect and protective immunity. J Virol 2014; 88:13990-4001. [PMID: 25253340 DOI: 10.1128/jvi.02273-14] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
UNLABELLED Pattern recognition receptors (PRR) sense certain molecular patterns uniquely expressed by pathogens. Retinoic-acid-inducible gene I (RIG-I) is a cytosolic PRR that senses viral nucleic acids and induces innate immune activation and secretion of type I interferons (IFNs). Here, using influenza vaccine antigens, we investigated the consequences of activating the RIG-I pathway for antigen-specific adaptive immune responses. We found that mice immunized with influenza vaccine antigens coadministered with 5'ppp-double-stranded RNA (dsRNA), a RIG-I ligand, developed robust levels of hemagglutination-inhibiting antibodies, enhanced germinal center reaction, and T follicular helper cell responses. In addition, RIG-I activation enhanced antibody affinity maturation and plasma cell responses in the draining lymph nodes, spleen, and bone marrow and conferred protective immunity against virus challenge. Importantly, activation of the RIG-I pathway was able to reduce the antigen requirement by 10- to 100-fold in inducing optimal influenza-specific cellular and humoral responses, including protective immunity. The effects induced by 5'ppp-dsRNA were significantly dependent on type I IFN and IPS-1 (an adapter protein downstream of the RIG-I pathway) signaling but were independent of the MyD88- and TLR3-mediated pathways. Our results show that activation of the RIG-I-like receptor pathway programs the innate immunity to achieve qualitatively and quantitatively enhanced protective cellular adaptive immune responses even at low antigen doses, and this indicates the potential utility of RIG-I ligands as molecular adjuvants for viral vaccines. IMPORTANCE The recently discovered RNA helicase family of RIG-I-like receptors (RLRs) is a critical component of host defense mechanisms responsible for detecting viruses and triggering innate antiviral cytokines that help control viral replication and dissemination. In this study, we show that the RLR pathway can be effectively exploited to enhance adaptive immunity and protective immune memory against viral infection. Our results show that activation of the RIG-I pathway along with influenza vaccination programs the innate immunity to induce qualitatively and quantitatively superior protective adaptive immunity against pandemic influenza viruses. More importantly, RIG-I activation at the time of vaccination allows induction of robust adaptive responses even at low vaccine antigen doses. These results highlight the potential utility of exploiting the RIG-I pathway to enhance viral-vaccine-specific immunity and have broader implications for designing better vaccines in general.
Collapse
|
40
|
Lipid nanoparticles as carriers for RNAi against viral infections: current status and future perspectives. BIOMED RESEARCH INTERNATIONAL 2014; 2014:161794. [PMID: 25184135 PMCID: PMC4145386 DOI: 10.1155/2014/161794] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 07/14/2014] [Accepted: 07/14/2014] [Indexed: 12/15/2022]
Abstract
The efforts made to develop RNAi-based therapies have led to productive research in the field of infections in humans, such as hepatitis C virus (HCV), hepatitis B virus (HBV), human immunodeficiency virus (HIV), human cytomegalovirus (HCMV), herpetic keratitis, human papillomavirus, or influenza virus. Naked RNAi molecules are rapidly digested by nucleases in the serum, and due to their negative surface charge, entry into the cell cytoplasm is also hampered, which makes necessary the use of delivery systems to exploit the full potential of RNAi therapeutics. Lipid nanoparticles (LNP) represent one of the most widely used delivery systems for in vivo application of RNAi due to their relative safety and simplicity of production, joint with the enhanced payload and protection of encapsulated RNAs. Moreover, LNP may be functionalized to reach target cells, and they may be used to combine RNAi molecules with conventional drug substances to reduce resistance or improve efficiency. This review features the current application of LNP in RNAi mediated therapy against viral infections and aims to explore possible future lines of action in this field.
Collapse
|
41
|
Chai D, Yue Y, Xu W, Dong C, Xiong S. Mucosal co-immunization with AIM2 enhances protective SIgA response and increases prophylactic efficacy of chitosan-DNA vaccine against coxsackievirus B3-induced myocarditis. Hum Vaccin Immunother 2014; 10:1284-94. [PMID: 24614684 DOI: 10.4161/hv.28333] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Coxsackievirus B3 (CVB3) infection is considered as the most common cause of viral myocarditis with no available vaccine. Considering that CVB3 mainly invades through the gastrointestinal mucosa, the development of CVB3-specific mucosal vaccine, which is the most efficient way to induce mucosal immune responses, gains more and more attention. In this study, we used absent in melanoma 2 (AIM2) as a mucosal adjuvant to enhance the immunogenicity and immunoprotection of CVB3-specific chitosan-pVP1 vaccine. Mice were intranasally co-immunized with 50 μg chitosan-pAIM2 and equal amount of chitosan-pVP1 vaccine 4 times at 2 week-intervals, and then challenged with CVB3 2 weeks after the last immunization. Compared with chitosan-pVP1 vaccine immunization alone, chitosan-pAIM2 co-immunization enhanced resistance to CVB3-induced myocarditis evidenced by significantly enhanced ejection fractions from 55.40 ± 9.35 to 80.31 ± 11.35, improved myocarditis scores from 1.50 ± 0.45 to 0.30 ± 0.15, reduced viral load from 3.33 ± 0.50 to 0.50 ± 0.65, and increased survival rate from 40.0% to 75.5%. This increased immunoprotection might be attributed to the augmented level of CVB3-specific fecal SIgA with high affinity and neutralizing ability. In addition, co-immunization with chitosan-pAIM2 remarkably facilitated dendritic cells (DCs) recruitment to mesenteric lymph nodes (MLN), and promoted the expression of IgA-inducing factors (BAFF, APRIL, iNOS, RALDH1, IL-6, TGF-β), which might account for its mucosal adjuvant effect. This strategy may represent a promising prophylactic vaccine against CVB3-induced myocarditis.
Collapse
Affiliation(s)
- Dafei Chai
- Jiangsu Key Laboratory of Infection and Immunity; Institutes of Biology and Medical Sciences; Soochow University; Suzhou, PR China
| | - Yan Yue
- Jiangsu Key Laboratory of Infection and Immunity; Institutes of Biology and Medical Sciences; Soochow University; Suzhou, PR China
| | - Wei Xu
- Jiangsu Key Laboratory of Infection and Immunity; Institutes of Biology and Medical Sciences; Soochow University; Suzhou, PR China
| | - Chunsheng Dong
- Jiangsu Key Laboratory of Infection and Immunity; Institutes of Biology and Medical Sciences; Soochow University; Suzhou, PR China
| | - Sidong Xiong
- Jiangsu Key Laboratory of Infection and Immunity; Institutes of Biology and Medical Sciences; Soochow University; Suzhou, PR China
| |
Collapse
|
42
|
Luke JM, Carnes AE, Williams JA. Development of antibiotic-free selection system for safer DNA vaccination. Methods Mol Biol 2014; 1143:91-111. [PMID: 24715283 DOI: 10.1007/978-1-4939-0410-5_6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The use of antibiotic-resistance markers in DNA vaccines is discouraged by regulatory agencies due to various theoretical safety concerns. This chapter presents methodologies for the design and cloning of synthetic antigen genes into RNA-OUT encoding antibiotic-free DNA vaccine vectors that are additionally optimized to improve protein expression, and immunogenicity, compared to alternative kanamycin-resistant vectors. First, antigen targeting considerations are discussed in the context of immune response customization through MHC class I or class II directed antigen presentation; the example NTC868 series RNA-OUT vector system allows simultaneous cloning into multiple vectors that feature various transgene intracellular targeting destinations. Then a detailed flowchart for codon optimization and synthetic transgene design is presented. Finally in-depth methodologies for cloning transgenes into the NTC868 series RNA-OUT vector system are presented. The resultant antibiotic-free DNA vaccine vectors are a more potent, safer alternative to existing kanamycin resistance marker encoding vectors.
Collapse
Affiliation(s)
- Jeremy M Luke
- Nature Technology Corporation, 4701 Innovation Drive, Lincoln, NE, 68521, USA
| | | | | |
Collapse
|
43
|
Coban C, Kobiyama K, Jounai N, Tozuka M, Ishii KJ. DNA vaccines: a simple DNA sensing matter? Hum Vaccin Immunother 2013; 9:2216-21. [PMID: 23912600 PMCID: PMC3906407 DOI: 10.4161/hv.25893] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Since the introduction of DNA vaccines two decades ago, this attractive strategy has been hampered by its low immunogenicity in humans. Studies conducted to improve the immunogenicity of DNA vaccines have shown that understanding the mechanism of action of DNA vaccines might be the key to successfully improving their immunogenicity. Our current understanding is that DNA vaccines induce innate and adaptive immune responses in two ways: (1) encoded protein (or polypeptide) antigen(s) by the DNA plasmid can be expressed in stromal cells (i.e., muscle cells) as well as DCs, where these antigens are processed and presented to naïve CD4 or CD8 T cells either by direct or cross presentation, respectively; and (2) the transfected DNA plasmid itself may bind to an un-identified cytosolic DNA sensor and activate the TBK1-STING pathway and the production of type I interferons (IFNs) which function as an adjuvant. Recent studies investigating double-stranded cytosolic DNA sensor(s) have highlighted new mechanisms in which cytosolic DNA may release secondary metabolites, which are in turn recognized by a novel DNA sensing machinery. Here, we discuss these new metabolites and the possibilities of translating this knowledge into improved immunogenicity for DNA vaccines.
Collapse
Affiliation(s)
- Cevayir Coban
- Laboratory of Malaria Immunology; WPI Immunology Frontier Research Center (IFReC); Osaka University; Osaka, Japan
| | - Kouji Kobiyama
- Laboratory of Adjuvant Innovation; National Institute of Biomedical Innovation; Osaka, Japan; Laboratory of Vaccine Science; IFReC; Osaka University; Osaka, Japan
| | - Nao Jounai
- Laboratory of Adjuvant Innovation; National Institute of Biomedical Innovation; Osaka, Japan
| | - Miyuki Tozuka
- Laboratory of Adjuvant Innovation; National Institute of Biomedical Innovation; Osaka, Japan
| | - Ken J Ishii
- Laboratory of Adjuvant Innovation; National Institute of Biomedical Innovation; Osaka, Japan; Laboratory of Vaccine Science; IFReC; Osaka University; Osaka, Japan
| |
Collapse
|
44
|
Williams JA. Vector Design for Improved DNA Vaccine Efficacy, Safety and Production. Vaccines (Basel) 2013; 1:225-49. [PMID: 26344110 PMCID: PMC4494225 DOI: 10.3390/vaccines1030225] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 06/12/2013] [Accepted: 06/18/2013] [Indexed: 12/25/2022] Open
Abstract
DNA vaccination is a disruptive technology that offers the promise of a new rapidly deployed vaccination platform to treat human and animal disease with gene-based materials. Innovations such as electroporation, needle free jet delivery and lipid-based carriers increase transgene expression and immunogenicity through more effective gene delivery. This review summarizes complementary vector design innovations that, when combined with leading delivery platforms, further enhance DNA vaccine performance. These next generation vectors also address potential safety issues such as antibiotic selection, and increase plasmid manufacturing quality and yield in exemplary fermentation production processes. Application of optimized constructs in combination with improved delivery platforms tangibly improves the prospect of successful application of DNA vaccination as prophylactic vaccines for diverse human infectious disease targets or as therapeutic vaccines for cancer and allergy.
Collapse
Affiliation(s)
- James A Williams
- Nature Technology Corporation/Suite 103, 4701 Innovation Drive, Lincoln, NE 68521, USA.
| |
Collapse
|
45
|
Immunologic Mechanism of Patchouli Alcohol Anti-H1N1 Influenza Virus May Through Regulation of the RLH Signal Pathway In Vitro. Curr Microbiol 2013; 67:431-6. [DOI: 10.1007/s00284-013-0381-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 04/03/2013] [Indexed: 12/25/2022]
|
46
|
Awate S, Babiuk LA, Mutwiri G. Mechanisms of action of adjuvants. Front Immunol 2013; 4:114. [PMID: 23720661 PMCID: PMC3655441 DOI: 10.3389/fimmu.2013.00114] [Citation(s) in RCA: 482] [Impact Index Per Article: 40.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 04/29/2013] [Indexed: 12/15/2022] Open
Abstract
Adjuvants are used in many vaccines, but their mechanisms of action are not fully understood. Studies from the past decade on adjuvant mechanisms are slowly revealing the secrets of adjuvant activity. In this review, we have summarized the recent progress in our understanding of the mechanisms of action of adjuvants. Adjuvants may act by a combination of various mechanisms including formation of depot, induction of cytokines and chemokines, recruitment of immune cells, enhancement of antigen uptake and presentation, and promoting antigen transport to draining lymph nodes. It appears that adjuvants activate innate immune responses to create a local immuno-competent environment at the injection site. Depending on the type of innate responses activated, adjuvants can alter the quality and quantity of adaptive immune responses. Understanding the mechanisms of action of adjuvants will provide critical information on how innate immunity influences the development of adaptive immunity, help in rational design of vaccines against various diseases, and can inform on adjuvant safety.
Collapse
Affiliation(s)
- Sunita Awate
- Vaccine and Infectious Disease Organization-International Vaccine Centre, School of Public Health, University of Saskatchewan Saskatoon, SK, Canada ; Vaccinology and Immunotherapeutics program, School of Public Health, University of Saskatchewan Saskatoon, SK, Canada
| | | | | |
Collapse
|
47
|
Pothlichet J, Meunier I, Davis BK, Ting JPY, Skamene E, von Messling V, Vidal SM. Type I IFN triggers RIG-I/TLR3/NLRP3-dependent inflammasome activation in influenza A virus infected cells. PLoS Pathog 2013; 9:e1003256. [PMID: 23592984 PMCID: PMC3623797 DOI: 10.1371/journal.ppat.1003256] [Citation(s) in RCA: 183] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 02/04/2013] [Indexed: 12/25/2022] Open
Abstract
Influenza A virus (IAV) triggers a contagious and potentially lethal respiratory disease. A protective IL-1β response is mediated by innate receptors in macrophages and lung epithelial cells. NLRP3 is crucial in macrophages; however, which sensors elicit IL-1β secretion in lung epithelial cells remains undetermined. Here, we describe for the first time the relative roles of the host innate receptors RIG-I (DDX58), TLR3, and NLRP3 in the IL-1β response to IAV in primary lung epithelial cells. To activate IL-1β secretion, these cells employ partially redundant recognition mechanisms that differ from those described in macrophages. RIG-I had the strongest effect through a MAVS/TRIM25/Riplet-dependent type I IFN signaling pathway upstream of TLR3 and NLRP3. Notably, RIG-I also activated the inflammasome through interaction with caspase 1 and ASC in primary lung epithelial cells. Thus, NS1, an influenza virulence factor that inhibits the RIG-I/type I IFN pathway, strongly modulated the IL-1β response in lung epithelial cells and in ferrets. The NS1 protein derived from a highly pathogenic strain resulted in increased interaction with RIG-I and inhibited type I IFN and IL-1β responses compared to the least pathogenic virus strains. These findings demonstrate that in IAV-infected lung epithelial cells RIG-I activates the inflammasome both directly and through a type I IFN positive feedback loop.
Collapse
Affiliation(s)
- Julien Pothlichet
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
- McGill Centre for the Study of Host Resistance, McGill University, Montreal, Quebec, Canada
- Institut Pasteur, Centre François Jacob, Paris, France
| | | | - Beckley K. Davis
- Department of Microbiology and Immunology School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Jenny P-Y. Ting
- Department of Microbiology and Immunology School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Emil Skamene
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
- McGill Centre for the Study of Host Resistance, McGill University, Montreal, Quebec, Canada
| | | | - Silvia M. Vidal
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
- McGill Centre for the Study of Host Resistance, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
48
|
Park H, Adamson L, Ha T, Mullen K, Hagen SI, Nogueron A, Sylwester AW, Axthelm MK, Legasse A, Piatak M, Lifson JD, McElrath JM, Picker LJ, Seder RA. Polyinosinic-polycytidylic acid is the most effective TLR adjuvant for SIV Gag protein-induced T cell responses in nonhuman primates. THE JOURNAL OF IMMUNOLOGY 2013; 190:4103-15. [PMID: 23509365 DOI: 10.4049/jimmunol.1202958] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Prime-boost immunization with heterologous vaccines elicits potent cellular immunity. In this study, we assessed the influence of various TLR ligands on SIV Gag-specific T cell immunity and protection following prime-boost immunization. Rhesus macaques (RMs) were primed with SIV Gag protein emulsified in Montanide ISA51 with or without TLR3 (polyinosinic-polycytidylic acid [poly-IC]), TLR4 (monophosphoryl lipid A), TLR7/8 (3M-012), TLR9 (CpG), or TLR3 (poly-IC) combined with TLR7/8 ligands, then boosted with replication defective adenovirus 5 expressing SIV Gag (rAd5-Gag). After priming, RMs that received SIV Gag protein plus poly-IC developed significantly higher frequencies of SIV Gag-specific CD4(+) Th1 responses in blood and bronchoalveolar lavage (BAL) fluid lymphocytes compared with all other adjuvants, and low-level SIV Gag-specific CD8(+) T cell responses. After the rAd5-Gag boost, the magnitude and breadth of SIV Gag-specific CD8(+) T cell responses were significantly increased in RM primed with SIV Gag protein plus poly-IC, with or without the TLR7/8 ligand, or CpG. However, the anamnestic, SIV Gag-specific CD8(+) T cell response to SIVmac251 challenge was not significantly enhanced by SIV Gag protein priming with any of the adjuvants. In contrast, the anamnestic SIV Gag-specific CD4(+) T cell response in BAL was enhanced by SIV Gag protein priming with poly-IC or CpG, which correlated with partial control of early viral replication after SIVmac251 challenge. These results demonstrate that prime-boost vaccination with SIV Gag protein/poly-IC improves magnitude, breadth, and durability of CD4(+) T cell immune responses, which could have a role in the control of SIV viral replication.
Collapse
Affiliation(s)
- Haesun Park
- Department of Pathology, Vaccine and Gene Therapy Institute, and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Yin J, Liu S, Zhu Y. An overview of the highly pathogenic H5N1 influenza virus. Virol Sin 2013; 28:3-15. [PMID: 23325419 PMCID: PMC7090813 DOI: 10.1007/s12250-013-3294-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 12/31/2012] [Indexed: 11/17/2022] Open
Abstract
Since the first human case of H5N1 avian influenza virus infection was reported in 1997, this highly pathogenic virus has infected hundreds of people around the world and resulted in many deaths. The ability of H5N1 to cross species boundaries, and the presence of polymorphisms that enhance virulence, present challenges to developing clear strategies to prevent the pandemic spread of this highly pathogenic avian influenza (HPAI) virus. This review summarizes the current understanding of, and recent research on, the avian influenza H5N1 virus, including transmission, virulence, pathogenesis, clinical characteristics, treatment and prevention.
Collapse
Affiliation(s)
- Jingchuan Yin
- The State Key laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| | | | | |
Collapse
|
50
|
Orsi A, Ansaldi F, de Florentiis D, Ceravolo A, Parodi V, Canepa P, Coppelli M, Icardi G, Durando P. Cross-protection against drifted influenza viruses: options offered by adjuvanted and intradermal vaccines. Hum Vaccin Immunother 2013; 9:582-90. [PMID: 23295230 DOI: 10.4161/hv.23239] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Antigenic drift, the evolutionary mechanism of influenza viruses, results in an increased susceptibility of vaccinated subjects against circulating viruses. New vaccines able to grant a broader and cross-reactive immune response against drifted influenza variants are needed. Several strategies were explored to enhance the immunogenicity of plain vaccines: adjuvants, carriers and intradermal administration of influenza vaccine emerge as a promising options. To evaluate the ability of a MF59-adjuvanted and intradermal influenza vaccine to elicit an effective antibody response against circulating viruses presenting antigenic patterns different from those of the vaccine strains, we compared antibody responses elicited by "implemented" vaccines and conventional intramuscular trivalent inactivated vaccine against heterologous circulating influenza A viruses. Different studies, simulating different epidemiological pictures produced by the natural antigenic drift of seasonal influenza viruses, highlighted the superior cross-reactivity of the antibodies elicited by MF59 and intradermal vaccines, compared with subunit or split vaccine against heterologous viruses.
Collapse
Affiliation(s)
- Andrea Orsi
- Department of Health Sciences; University of Genoa; Genoa, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|