1
|
Peng S, Fan D, Tu HF, Cheng M, Arend RC, Levinson K, Tao J, Roden RBS, Hung CF, Wu TC. Improved efficacy of therapeutic HPV DNA vaccine using intramuscular injection with electroporation compared to conventional needle and needle-free jet injector methods. Cell Biosci 2024; 14:154. [PMID: 39722048 DOI: 10.1186/s13578-024-01338-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND We have previously developed a candidate therapeutic HPV DNA vaccine (pBI-11) encoding mycobacteria heat shock protein 70 linked to HPV16/18 E6/E7 proteins for the control of advanced HPV-associated oropharyngeal cancer (NCT05799144). While naked DNA vaccines are readily produced, stable, and well tolerated, their potency is limited by the delivery efficiency. Here we compared three different IM delivery strategies, including intramuscular (IM) injection, either with a needle alone or with electroporation at the injection site, and a needle-free injection system (NFIS), for their ability to elicit gene expression and to improve the potency of pBI-11 DNA vaccine. RESULTS We found that electroporation after IM injection significantly increases gene expression from a luciferase-encoding DNA construct compared to IM injection alone or NFIS. We also showed that single administration of pBI-11 DNA via electroporation-mediated delivery generates the greatest increase in HPV antigen-specific CD8 + T cell-mediated immune responses, resulting in the most potent antitumor effect compared to the other two methods. We further compared the response to three repeat immunizations via each of these different methods. We found that electroporation-mediated delivery of pBI-11 DNA generates the greatest HPV antigen-specific CD8 + T cell immune responses and therapeutic antitumor effects compared to the other two methods. Monitoring of mouse behaviors and body weight, and necropsy indicated that electroporation-mediated delivery of clinical grade pBI-11 DNA vaccine was well-tolerated and presented no evident local or systemic toxicity. CONCLUSIONS These findings provide rationale for clinical testing of pBI-11 DNA vaccine delivered by electroporation for the control of HPV16/18-associated infections and/or cancers.
Collapse
Affiliation(s)
- Shiwen Peng
- Department of Pathology, Johns Hopkins School of Medicine, CRB II Room 307, 1550 Orleans St, Baltimore, MD, USA
| | - Darrell Fan
- Department of Pathology, Johns Hopkins School of Medicine, CRB II Room 307, 1550 Orleans St, Baltimore, MD, USA
| | - Hsin-Fang Tu
- Department of Pathology, Johns Hopkins School of Medicine, CRB II Room 307, 1550 Orleans St, Baltimore, MD, USA
| | - Michelle Cheng
- Department of Pathology, Johns Hopkins School of Medicine, CRB II Room 307, 1550 Orleans St, Baltimore, MD, USA
| | - Rebecca C Arend
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham, O'Neal Comprehensive Cancer Center, Birmingham, AL, USA
| | - Kimberly Levinson
- Department of Oncology, Johns Hopkins School of Medicine, CRB II Room 307, 1550 Orleans St, Baltimore, MD, USA
- Department of Obstetrics and Gynecology, Johns Hopkins University, CRB II Room 307, 1550 Orleans St, Baltimore, MD, USA
| | - Julia Tao
- Department of Pathology, Johns Hopkins School of Medicine, CRB II Room 307, 1550 Orleans St, Baltimore, MD, USA
| | - Richard B S Roden
- Department of Pathology, Johns Hopkins School of Medicine, CRB II Room 307, 1550 Orleans St, Baltimore, MD, USA
- Department of Oncology, Johns Hopkins School of Medicine, CRB II Room 307, 1550 Orleans St, Baltimore, MD, USA
| | - Chien-Fu Hung
- Department of Pathology, Johns Hopkins School of Medicine, CRB II Room 307, 1550 Orleans St, Baltimore, MD, USA.
- Department of Oncology, Johns Hopkins School of Medicine, CRB II Room 307, 1550 Orleans St, Baltimore, MD, USA.
- Department of Obstetrics and Gynecology, Johns Hopkins University, CRB II Room 307, 1550 Orleans St, Baltimore, MD, USA.
| | - T-C Wu
- Department of Pathology, Johns Hopkins School of Medicine, CRB II Room 307, 1550 Orleans St, Baltimore, MD, USA.
- Department of Oncology, Johns Hopkins School of Medicine, CRB II Room 307, 1550 Orleans St, Baltimore, MD, USA.
- Department of Obstetrics and Gynecology, Johns Hopkins University, CRB II Room 307, 1550 Orleans St, Baltimore, MD, USA.
| |
Collapse
|
2
|
Garcia-Atutxa I, Mondragon-Teran P, Huerta-Saquero A, Villanueva-Flores F. Advancements in monkeypox vaccines development: a critical review of emerging technologies. Front Immunol 2024; 15:1456060. [PMID: 39464881 PMCID: PMC11502315 DOI: 10.3389/fimmu.2024.1456060] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 09/20/2024] [Indexed: 10/29/2024] Open
Abstract
Monkeypox (mpox) is a zoonotic illness caused by the monkeypox virus (MPXV), with higher health concerns among people who are pregnant, children, and persons who are immunocompromised, including people with untreated and advanced HIV disease. Significant progress has been made in developing vaccines against mpox, yet critical challenges and limitations persist in ensuring their effectiveness, safety, and accessibility. The pertinence of this review is highlighted by the World Health Organization's declaration of a global health emergency on August 14, 2024, due to the recent mpox outbreak, underscoring the critical necessity for effective vaccine solutions in the face of a rapidly evolving virus. Here, we comprehensively analyze various vaccine platforms utilized in mpox prevention, including attenuated and non-replicating virus vaccines, viral vector-based vaccines, recombinant protein vaccines, and DNA and mRNA vaccines. We evaluate the advantages and limitations of each platform, highlighting the urgent need for ongoing research and innovation to enhance vaccine efficacy and safety. Recent advancements, such as incorporating immunostimulatory sequences, improved delivery systems, and developing polyvalent vaccines, are explored for their potential to offer broader protection against diverse orthopoxvirus strains. This work underscores the need to optimize currently available vaccines and investigate novel vaccination strategies to address future public health emergencies effectively. By focusing on these advanced methodologies, we aim to contribute to the development of robust and adaptable vaccine solutions for mpox and other related viral threats.
Collapse
Affiliation(s)
- Igor Garcia-Atutxa
- Computer Science Department, Universidad Católica de Murcia (UCAM), Murcia, Spain
| | - Paul Mondragon-Teran
- Centro de Investigación en Ciencia Aplicada y Tecnología Avanzada (CICATA) Unidad Morelos del Instituto Politécnico Nacional (IPN), Xochitepec, Morelos, Mexico
| | - Alejandro Huerta-Saquero
- Departamento de Bionanotecnología, Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México (UNAM), Ensenada, Mexico
| | - Francisca Villanueva-Flores
- Centro de Investigación en Ciencia Aplicada y Tecnología Avanzada (CICATA) Unidad Morelos del Instituto Politécnico Nacional (IPN), Xochitepec, Morelos, Mexico
| |
Collapse
|
3
|
Eftekhari Z, Zohrabi H, Oghalaie A, Ebrahimi T, Shariati FS, Behdani M, Kazemi-Lomedasht F. Advancements and challenges in mRNA and ribonucleoprotein-based therapies: From delivery systems to clinical applications. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102313. [PMID: 39281702 PMCID: PMC11402252 DOI: 10.1016/j.omtn.2024.102313] [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: 09/18/2024]
Abstract
The use of mRNA and ribonucleoproteins (RNPs) as therapeutic agents is a promising strategy for treating diseases such as cancer and infectious diseases. This review provides recent advancements and challenges in mRNA- and RNP-based therapies, focusing on delivery systems such as lipid nanoparticles (LNPs), which ensure efficient delivery to target cells. Strategies such as microfluidic devices are employed to prepare LNPs loaded with mRNA and RNPs, demonstrating effective genome editing and protein expression in vitro and in vivo. These applications extend to cancer treatment and infectious disease management, with promising results in genome editing for cancer therapy using LNPs encapsulating Cas9 mRNA and single-guide RNA. In addition, tissue-specific targeting strategies offer potential for improved therapeutic outcomes and reduced off-target effects. Despite progress, challenges such as optimizing delivery efficiency and targeting remain. Future research should enhance delivery efficiency, explore tissue-specific targeting, investigate combination therapies, and advance clinical translation. In conclusion, mRNA- and RNP-based therapies offer a promising avenue for treating various diseases and have the potential to revolutionize medicine, providing new hope for patients worldwide.
Collapse
Affiliation(s)
- Zohre Eftekhari
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Horieh Zohrabi
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Akbar Oghalaie
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Tahereh Ebrahimi
- Department of Nanobiotechnology, New Technologies Research Group, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Fatemeh Sadat Shariati
- Department of Influenza and other Respiratory Viruses, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Mahdi Behdani
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Fatemeh Kazemi-Lomedasht
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran
| |
Collapse
|
4
|
Cui L, Wang J, Orlando F, Giacconi R, Malavolta M, Bartozzi B, Galeazzi R, Giorgini G, Pesce L, Cardarelli F, Quagliarini E, Renzi S, Xiao S, Pozzi D, Provinciali M, Caracciolo G, Marchini C, Amici A. Enhancing Immune Responses against SARS-CoV-2 Variants in Aged Mice with INDUK: A Chimeric DNA Vaccine Encoding the Spike S1-TM Subunits. ACS OMEGA 2024; 9:34624-34635. [PMID: 39157118 PMCID: PMC11325517 DOI: 10.1021/acsomega.4c03285] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 06/12/2024] [Accepted: 06/14/2024] [Indexed: 08/20/2024]
Abstract
Currently available vaccines against COVID-19 showed high efficacy against the original strain of SARS-CoV-2 but progressively lower efficacy against new variants. In response to emerging SARS-CoV-2 strains, we propose chimeric DNA vaccines encoding the spike antigen, including a combination of selected key mutations from different variants of concern. We developed two DNA vaccines, pVAX-S1-TM-D614G and pVAX-S1-TM-INDUK (INDUK), encoding the SARS-CoV-2 S1 spike subunit in fusion with the transmembrane region that allows protein trimerization as predicted by in silico analysis. pVAX-S1-TM-D614G included the dominant D614G substitution, while the chimeric vaccine INDUK contained additional selected mutations from the Delta (E484Q and L452R) and Alpha (N501Y and A570D) variants. Considering that aging is a risk factor for severe disease and that suboptimal vaccine responses were observed in older individuals, the immunogenicity of pVAX-S1-TM-D614G and INDUK was tested in both young and aged C57BL/6 mice. Two vaccine doses were able to trigger significant anti-SARS-CoV-2 antibody production, showing neutralizing activity. ELISA tests confirmed that antibodies induced by pVAX-S1-TM-D614G and INDUK were able to recognize both Wuhan Spike and Delta variant Spike as trimers, while neutralizing antibodies were detected by an ACE2:SARS-CoV-2 Spike S1 inhibitor screening assay, designed to assess the capacity of antibodies to block the interaction between the viral spike S1 protein and the ACE2 receptor. Although antibody titer declined within six months, a third booster dose significantly increased the magnitude of humoral response, even in aged individuals, suggesting that immune recall can improve antibody response durability. The analysis of cellular responses demonstrated that vaccination with INDUK elicited an increase in the percentage of SARS-CoV-2-specific IFN-γ producing T lymphocytes in immunized young mice and TNF-α-producing T lymphocytes in both young and aged mice. These findings not only hold immediate promise for addressing evolving challenges in SARS-CoV-2 vaccination but also open avenues to refine strategies and elevate the effectiveness of next-generation vaccines.
Collapse
Affiliation(s)
- Lishan Cui
- School
of Biosciences and Veterinary Medicine, University of Camerino, via Gentile III da Varano, 62032 Camerino, Italy
| | - Junbiao Wang
- School
of Biosciences and Veterinary Medicine, University of Camerino, via Gentile III da Varano, 62032 Camerino, Italy
| | - Fiorenza Orlando
- Experimental
Animal Models for Aging Unit, Scientific Technological Area, IRRCS INRCA, 60100 Ancona, Italy
| | - Robertina Giacconi
- Advanced
Technology Center for Aging Research, IRCCS
INRCA, 60100Ancona, Italy
| | - Marco Malavolta
- Advanced
Technology Center for Aging Research, IRCCS
INRCA, 60100Ancona, Italy
| | - Beatrice Bartozzi
- Advanced
Technology Center for Aging Research, IRCCS
INRCA, 60100Ancona, Italy
| | - Roberta Galeazzi
- Department
of Life and Environmental Sciences, Marche
Polytechnic University, 60131 Ancona, Italy
| | - Giorgia Giorgini
- Department
of Life and Environmental Sciences, Marche
Polytechnic University, 60131 Ancona, Italy
| | - Luca Pesce
- NEST
Laboratory, Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Francesco Cardarelli
- NEST
Laboratory, Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Erica Quagliarini
- NanoDelivery
Lab, Department of Molecular Medicine, Sapienza
University of Rome, viale
Regina Elena 291, 00161 Rome, Italy
| | - Serena Renzi
- NanoDelivery
Lab, Department of Molecular Medicine, Sapienza
University of Rome, viale
Regina Elena 291, 00161 Rome, Italy
| | - Siyao Xiao
- NanoDelivery
Lab, Department of Molecular Medicine, Sapienza
University of Rome, viale
Regina Elena 291, 00161 Rome, Italy
| | - Daniela Pozzi
- NanoDelivery
Lab, Department of Molecular Medicine, Sapienza
University of Rome, viale
Regina Elena 291, 00161 Rome, Italy
| | - Mauro Provinciali
- Experimental
Animal Models for Aging Unit, Scientific Technological Area, IRRCS INRCA, 60100 Ancona, Italy
| | - Giulio Caracciolo
- NanoDelivery
Lab, Department of Molecular Medicine, Sapienza
University of Rome, viale
Regina Elena 291, 00161 Rome, Italy
| | - Cristina Marchini
- School
of Biosciences and Veterinary Medicine, University of Camerino, via Gentile III da Varano, 62032 Camerino, Italy
| | - Augusto Amici
- School
of Biosciences and Veterinary Medicine, University of Camerino, via Gentile III da Varano, 62032 Camerino, Italy
| |
Collapse
|
5
|
Han L, Song S, Feng H, Ma J, Wei W, Si F. A roadmap for developing Venezuelan equine encephalitis virus (VEEV) vaccines: Lessons from the past, strategies for the future. Int J Biol Macromol 2023:125514. [PMID: 37353130 DOI: 10.1016/j.ijbiomac.2023.125514] [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: 04/11/2023] [Revised: 06/16/2023] [Accepted: 06/20/2023] [Indexed: 06/25/2023]
Abstract
Venezuelan equine encephalitis (VEE) is a zoonotic infectious disease caused by the Venezuelan equine encephalitis virus (VEEV), which can lead to severe central nervous system infections in both humans and animals. At present, the medical community does not possess a viable means of addressing VEE, rendering the prevention of the virus a matter of paramount importance. Regarding the prevention and control of VEEV, the implementation of a vaccination program has been recognized as the most efficient strategy. Nevertheless, there are currently no licensed vaccines or drugs available for human use against VEEV. This imperative has led to a surge of interest in vaccine research, with VEEV being a prime focus for researchers in the field. In this paper, we initially present a comprehensive overview of the current taxonomic classification of VEEV and the cellular infection mechanism of the virus. Subsequently, we provide a detailed introduction of the prominent VEEV vaccine types presently available, including inactivated vaccines, live attenuated vaccines, genetic, and virus-like particle vaccines. Moreover, we emphasize the challenges that current VEEV vaccine development faces and suggest urgent measures that must be taken to overcome these obstacles. Notably, based on our latest research, we propose the feasibility of incorporation codon usage bias strategies to create the novel VEEV vaccine. Finally, we prose several areas that future VEEV vaccine development should focus on. Our objective is to encourage collaboration between the medical and veterinary communities, expedite the translation of existing vaccines from laboratory to clinical applications, while also preparing for future outbreaks of new VEEV variants.
Collapse
Affiliation(s)
- Lulu Han
- Institute of Animal Science and Veterinary Medicine, Shanghai Academy of Agricultural Sciences, Shanghai Key Laboratory of Agricultural Genetics and Breeding, Shanghai Engineering Research Center of Breeding Pig, Shanghai 201106, China; Huaihe Hospital of Henan University, Clinical Medical College of Henan University, Kai Feng 475000, China
| | - Shuai Song
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, PR China
| | - Huilin Feng
- Kaifeng Key Laboratory of Infection and Biological Safety, School of Basic Medical Sciences of Henan University, Kai Feng 475000, China
| | - Jing Ma
- Huaihe Hospital of Henan University, Clinical Medical College of Henan University, Kai Feng 475000, China
| | - Wenqiang Wei
- Kaifeng Key Laboratory of Infection and Biological Safety, School of Basic Medical Sciences of Henan University, Kai Feng 475000, China.
| | - Fusheng Si
- Institute of Animal Science and Veterinary Medicine, Shanghai Academy of Agricultural Sciences, Shanghai Key Laboratory of Agricultural Genetics and Breeding, Shanghai Engineering Research Center of Breeding Pig, Shanghai 201106, China.
| |
Collapse
|
6
|
Mba IE, Sharndama HC, Anyaegbunam ZKG, Anekpo CC, Amadi BC, Morumda D, Doowuese Y, Ihezuo UJ, Chukwukelu JU, Okeke OP. Vaccine development for bacterial pathogens: Advances, challenges and prospects. Trop Med Int Health 2023; 28:275-299. [PMID: 36861882 DOI: 10.1111/tmi.13865] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
The advent and use of antimicrobials have played a key role in treating potentially life-threatening infectious diseases, improving health, and saving the lives of millions of people worldwide. However, the emergence of multidrug resistant (MDR) pathogens has been a significant health challenge that has compromised the ability to prevent and treat a wide range of infectious diseases that were once treatable. Vaccines offer potential as a promising alternative to fight against antimicrobial resistance (AMR) infectious diseases. Vaccine technologies include reverse vaccinology, structural biology methods, nucleic acid (DNA and mRNA) vaccines, generalised modules for membrane antigens, bioconjugates/glycoconjugates, nanomaterials and several other emerging technological advances that are offering a potential breakthrough in the development of efficient vaccines against pathogens. This review covers the opportunities and advancements in vaccine discovery and development targeting bacterial pathogens. We reflect on the impact of the already-developed vaccines targeting bacterial pathogens and the potential of those currently under different stages of preclinical and clinical trials. More importantly, we critically and comprehensively analyse the challenges while highlighting the key indices for future vaccine prospects. Finally, the issues and concerns of AMR for low-income countries (sub-Saharan Africa) and the challenges with vaccine integration, discovery and development in this region are critically evaluated.
Collapse
Affiliation(s)
- Ifeanyi Elibe Mba
- Department of Microbiology, University of Nigeria, Nsukka, Nigeria
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, University of Ibadan, Ibadan, Nigeria
| | | | - Zikora Kizito Glory Anyaegbunam
- Department of Microbiology, University of Nigeria, Nsukka, Nigeria
- Institute for Drug-Herbal Medicine-Excipient Research and Development, University of Nigeria, Nsukka, Nigeria
| | - Chijioke Chinedu Anekpo
- Department of Ear Nose and Throat, College of Medicine, Enugu State University of Science and Technology, Enugu, Nigeria
| | - Ben Chibuzo Amadi
- Pharmaceutical Technology and Industrial Pharmacy, University of Nigeria, Nsukka, Nigeria
| | - Daji Morumda
- Department of Microbiology, Federal University Wukari, Wukari, Taraba, Nigeria
| | - Yandev Doowuese
- Department of Microbiology, Federal University of Health Sciences, Otukpo, Nigeria
| | - Uchechi Justina Ihezuo
- Department of Microbiology, University of Nigeria, Nsukka, Nigeria
- Institute for Drug-Herbal Medicine-Excipient Research and Development, University of Nigeria, Nsukka, Nigeria
| | | | | |
Collapse
|
7
|
Kozak M, Hu J. The Integrated Consideration of Vaccine Platforms, Adjuvants, and Delivery Routes for Successful Vaccine Development. Vaccines (Basel) 2023; 11:vaccines11030695. [PMID: 36992279 DOI: 10.3390/vaccines11030695] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/01/2023] [Accepted: 03/02/2023] [Indexed: 03/31/2023] Open
Abstract
Vaccines have proven to be the most cost-efficient and reasonable way to fight and exterminate virulent pathogens. Vaccines can be designed using a variety of platforms including inactivated/attenuated pathogen or subunits of it. The most recent COVID mRNA vaccines have employed nucleic acid sequences for the antigen of interest to combat the pandemic. Different vaccine platforms have been chosen for different licensed vaccines which all have shown their ability to induce durable immune responses and protection. In addition to platforms, different adjuvants have been used to strengthen the immunogenicity of vaccines. Among the delivery routes, intramuscular injection has been the most common for vaccination. In this review, we present a historical overview of the integrated consideration of vaccine platforms, adjuvants, and delivery routes in the success of vaccine development. We also discuss the advantages and limitations of each choice in the efficacy of vaccine development.
Collapse
Affiliation(s)
- Michael Kozak
- The Jake Gittlen Laboratories for Cancer Research, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Department of Pathology and Laboratory Medicine, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Jiafen Hu
- The Jake Gittlen Laboratories for Cancer Research, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Department of Pathology and Laboratory Medicine, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
8
|
Malyško-Ptašinskė V, Staigvila G, Novickij V. Invasive and non-invasive electrodes for successful drug and gene delivery in electroporation-based treatments. Front Bioeng Biotechnol 2023; 10:1094968. [PMID: 36727038 PMCID: PMC9885012 DOI: 10.3389/fbioe.2022.1094968] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 12/28/2022] [Indexed: 01/17/2023] Open
Abstract
Electroporation is an effective physical method for irreversible or reversible permeabilization of plasma membranes of biological cells and is typically used for tissue ablation or targeted drug/DNA delivery into living cells. In the context of cancer treatment, full recovery from an electroporation-based procedure is frequently dependent on the spatial distribution/homogeneity of the electric field in the tissue; therefore, the structure of electrodes/applicators plays an important role. This review focuses on the analysis of electrodes and in silico models used for electroporation in cancer treatment and gene therapy. We have reviewed various invasive and non-invasive electrodes; analyzed the spatial electric field distribution using finite element method analysis; evaluated parametric compatibility, and the pros and cons of application; and summarized options for improvement. Additionally, this review highlights the importance of tissue bioimpedance for accurate treatment planning using numerical modeling and the effects of pulse frequency on tissue conductivity and relative permittivity values.
Collapse
Affiliation(s)
| | - Gediminas Staigvila
- Faculty of Electronics, Vilnius Gediminas Technical University, Vilnius, Lithuania
| | - Vitalij Novickij
- Faculty of Electronics, Vilnius Gediminas Technical University, Vilnius, Lithuania
- Department of Immunology, State Research Institute Centre of Innovative Medicine, Vilnius, Lithuania
| |
Collapse
|
9
|
Melo ARDS, de Macêdo LS, Invenção MDCV, de Moura IA, da Gama MATM, de Melo CML, Silva AJD, Batista MVDA, de Freitas AC. Third-Generation Vaccines: Features of Nucleic Acid Vaccines and Strategies to Improve Their Efficiency. Genes (Basel) 2022; 13:genes13122287. [PMID: 36553554 PMCID: PMC9777941 DOI: 10.3390/genes13122287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/23/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Gene immunization comprises mRNA and DNA vaccines, which stand out due to their simple design, maintenance, and high efficacy. Several studies indicate promising results in preclinical and clinical trials regarding immunization against ebola, human immunodeficiency virus (HIV), influenza, and human papillomavirus (HPV). The efficiency of nucleic acid vaccines has been highlighted in the fight against COVID-19 with unprecedented approval of their use in humans. However, their low intrinsic immunogenicity points to the need to use strategies capable of overcoming this characteristic and increasing the efficiency of vaccine campaigns. These strategies include the improvement of the epitopes' presentation to the system via MHC, the evaluation of immunodominant epitopes with high coverage against emerging viral subtypes, the use of adjuvants that enhance immunogenicity, and the increase in the efficiency of vaccine transfection. In this review, we provide updates regarding some characteristics, construction, and improvement of such vaccines, especially about the production of synthetic multi-epitope genes, widely employed in the current gene-based vaccines.
Collapse
Affiliation(s)
- Alanne Rayssa da Silva Melo
- Laboratory of Molecular Studies and Experimental Therapy—LEMTE, Department of Genetics, Federal University of Pernambuco, Recife 50670-901, Brazil
| | - Larissa Silva de Macêdo
- Laboratory of Molecular Studies and Experimental Therapy—LEMTE, Department of Genetics, Federal University of Pernambuco, Recife 50670-901, Brazil
| | - Maria da Conceição Viana Invenção
- Laboratory of Molecular Studies and Experimental Therapy—LEMTE, Department of Genetics, Federal University of Pernambuco, Recife 50670-901, Brazil
| | - Ingrid Andrêssa de Moura
- Laboratory of Molecular Studies and Experimental Therapy—LEMTE, Department of Genetics, Federal University of Pernambuco, Recife 50670-901, Brazil
| | - Marco Antonio Turiah Machado da Gama
- Laboratory of Molecular Studies and Experimental Therapy—LEMTE, Department of Genetics, Federal University of Pernambuco, Recife 50670-901, Brazil
| | - Cristiane Moutinho Lagos de Melo
- Laboratory of Immunological and Antitumor Analysis, Department of Antibiotics, Bioscience Center, and Keizo Asami Imunophatology Laboratory, Federal University of Pernambuco, Recife 50670-901, Brazil
| | - Anna Jéssica Duarte Silva
- Laboratory of Molecular Studies and Experimental Therapy—LEMTE, Department of Genetics, Federal University of Pernambuco, Recife 50670-901, Brazil
| | - Marcus Vinicius de Aragão Batista
- Laboratory of Molecular Genetics and Biotechnology (GMBio), Department of Biology, Center for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão 49100-000, Brazil
| | - Antonio Carlos de Freitas
- Laboratory of Molecular Studies and Experimental Therapy—LEMTE, Department of Genetics, Federal University of Pernambuco, Recife 50670-901, Brazil
- Correspondence: ; Tel.: +55-8199-6067-671
| |
Collapse
|
10
|
Wang Z, Wang G, Lu H, Li H, Tang M, Tong A. Development of therapeutic antibodies for the treatment of diseases. MOLECULAR BIOMEDICINE 2022; 3:35. [PMID: 36418786 PMCID: PMC9684400 DOI: 10.1186/s43556-022-00100-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/24/2022] [Indexed: 11/25/2022] Open
Abstract
Since the first monoclonal antibody drug, muromonab-CD3, was approved for marketing in 1986, 165 antibody drugs have been approved or are under regulatory review worldwide. With the approval of new drugs for treating a wide range of diseases, including cancer and autoimmune and metabolic disorders, the therapeutic antibody drug market has experienced explosive growth. Monoclonal antibodies have been sought after by many biopharmaceutical companies and scientific research institutes due to their high specificity, strong targeting abilities, low toxicity, side effects, and high development success rate. The related industries and markets are growing rapidly, and therapeutic antibodies are one of the most important research and development areas in the field of biology and medicine. In recent years, great progress has been made in the key technologies and theoretical innovations provided by therapeutic antibodies, including antibody-drug conjugates, antibody-conjugated nuclides, bispecific antibodies, nanobodies, and other antibody analogs. Additionally, therapeutic antibodies can be combined with technologies used in other fields to create new cross-fields, such as chimeric antigen receptor T cells (CAR-T), CAR-natural killer cells (CAR-NK), and other cell therapy. This review summarizes the latest approved or in regulatory review therapeutic antibodies that have been approved or that are under regulatory review worldwide, as well as clinical research on these approaches and their development, and outlines antibody discovery strategies that have emerged during the development of therapeutic antibodies, such as hybridoma technology, phage display, preparation of fully human antibody from transgenic mice, single B-cell antibody technology, and artificial intelligence-assisted antibody discovery.
Collapse
Affiliation(s)
- Zeng Wang
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Guoqing Wang
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu, China
| | - Huaqing Lu
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Hongjian Li
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Mei Tang
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Aiping Tong
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
11
|
Onzere CK, Fry LM, Bishop RP, Da Silva M, Madsen-Bouterse SA, Bastos RG, Knowles DP, Suarez CE. Theileria equi RAP-1a and RAP-1b proteins contain immunoreactive epitopes and are suitable candidates for vaccine and diagnostics development. Int J Parasitol 2022; 52:385-397. [DOI: 10.1016/j.ijpara.2022.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 11/16/2022]
|
12
|
Current view on novel vaccine technologies to combat human infectious diseases. Appl Microbiol Biotechnol 2022; 106:25-56. [PMID: 34889981 PMCID: PMC8661323 DOI: 10.1007/s00253-021-11713-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/19/2021] [Accepted: 11/24/2021] [Indexed: 02/06/2023]
Abstract
Inactivated and live attenuated vaccines have improved human life and significantly reduced morbidity and mortality of several human infectious diseases. However, these vaccines have faults, such as reactivity or suboptimal efficacy and expensive and time-consuming development and production. Additionally, despite the enormous efforts to develop vaccines against some infectious diseases, the traditional technologies have not been successful in achieving this. At the same time, the concerns about emerging and re-emerging diseases urge the need to develop technologies that can be rapidly applied to combat the new challenges. Within the last two decades, the research of vaccine technologies has taken several directions to achieve safe, efficient, and economic platforms or technologies for novel vaccines. This review will give a brief overview of the current state of the novel vaccine technologies, new vaccine candidates in clinical trial phases 1-3 (listed by European Medicines Agency (EMA) and Food and Drug Administration (FDA)), and vaccines based on the novel technologies which have already been commercially available (approved by EMA and FDA) with the special reference to pandemic COVID-19 vaccines. KEY POINTS: • Vaccines of the new generation follow the minimalist strategy. • Some infectious diseases remain a challenge for the vaccine development. • The number of new vaccine candidates in the late phase clinical trials remains low.
Collapse
|
13
|
Viana Invenção MDC, Melo ARDS, de Macêdo LS, da Costa Neves TSP, de Melo CML, Cordeiro MN, de Aragão Batista MV, de Freitas AC. Development of synthetic antigen vaccines for COVID-19. Hum Vaccin Immunother 2021; 17:3855-3870. [PMID: 34613880 PMCID: PMC8506811 DOI: 10.1080/21645515.2021.1974288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 08/04/2021] [Accepted: 08/24/2021] [Indexed: 11/04/2022] Open
Abstract
The current pandemic called COVID-19 caused by the SARS-CoV-2 virus brought the need for the search for fast alternatives to both control and fight the SARS-CoV-2 infection. Therefore, a race for a vaccine against COVID-19 took place, and some vaccines have been approved for emergency use in several countries in a record time. Ongoing prophylactic research has sought faster, safer, and precise alternatives by redirecting knowledge of other vaccines, and/or the development of new strategies using available tools, mainly in the areas of genomics and bioinformatics. The current review highlights the development of synthetic antigen vaccines, focusing on the usage of bioinformatics tools for the selection and construction of antigens on the different vaccine constructions under development, as well as strategies to optimize vaccines for COVID-19.
Collapse
Affiliation(s)
- Maria da Conceição Viana Invenção
- Laboratory of Molecular Studies and Experimental Therapy - LEMTE, Department of Genetics, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Alanne Rayssa da Silva Melo
- Laboratory of Molecular Studies and Experimental Therapy - LEMTE, Department of Genetics, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Larissa Silva de Macêdo
- Laboratory of Molecular Studies and Experimental Therapy - LEMTE, Department of Genetics, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Thaís Souto Paula da Costa Neves
- Laboratory of Molecular Studies and Experimental Therapy - LEMTE, Department of Genetics, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Cristiane Moutinho Lagos de Melo
- Laboratory of Immunological and Antitumor Analysis, Department of Antibiotics, Bioscience Center, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Marcelo Nazário Cordeiro
- Laboratory of Molecular Studies and Experimental Therapy - LEMTE, Department of Genetics, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Marcus Vinicius de Aragão Batista
- Laboratory of Molecular Genetics and Biotechnology, Department of Biology, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil
| | - Antonio Carlos de Freitas
- Laboratory of Molecular Studies and Experimental Therapy - LEMTE, Department of Genetics, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| |
Collapse
|
14
|
A Needle-Free Jet Injection System for Controlled Release and Repeated Biopharmaceutical Delivery. Pharmaceutics 2021; 13:pharmaceutics13111770. [PMID: 34834185 PMCID: PMC8620904 DOI: 10.3390/pharmaceutics13111770] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/13/2021] [Accepted: 10/19/2021] [Indexed: 12/24/2022] Open
Abstract
Swift vaccination is necessary as a response to disease outbreaks and pandemics; otherwise, the species under attack is at risk of a high fatality rate or even mass extinction. Statistics suggest that at least 16 billion injections are administered worldwide every year. Such a high rate of needle/syringe injection administration worldwide is alarming due to the risk of needle-stick injuries, disease spread due to cross-contamination and the reuse of needles, and the misuse of needles. In addition, there are production, handling, and disposal costs. Needle phobia is an additional issue faced by many recipients of injections with needles. In addition to a detailed literature review highlighting the need for needle-free injection systems, a compressed air-driven needle-free jet injection system with a hydro-pneumatic mechanism was designed and developed by employing an axiomatic design approach. The proposed injection system has higher flexibility, uninterrupted force generation, and provides the possibility of delivering repeated injections at different tissue depths from the dermis to the muscle (depending on the drug delivery requirements) by controlling the inlet compressed air pressure. The designed needle-free jet injector consists of two primary circuits: the pneumatic and the hydraulic circuit. The pneumatic circuit is responsible for driving, pressurizing, and repeatability. The hydraulic circuit precisely injects and contains the liquid jet, allowing us to control the volume of the liquid jet at elevated pressure by offering flexibility in the dose volume per injection. Finally, in this paper we report on the successful design and working model of an air-driven needle-free jet injector for 0.2–0.5 mL drug delivery by ex vivo experimental validation.
Collapse
|
15
|
Li H, Wang S, Hu G, Zhang L, Liu S, Lu S. DNA priming immunization is more effective than recombinant protein vaccine in eliciting antigen-specific B cell responses. Emerg Microbes Infect 2021; 10:833-841. [PMID: 33853515 PMCID: PMC8812797 DOI: 10.1080/22221751.2021.1918026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
While DNA prime-protein boost vaccination approach has been widely used in preclinical and clinical studies especially in the field of HIV vaccine development, the exact role of DNA immunization has not been fully identified. Our previous work demonstrated that DNA immunization was able to elicit T follicular helper (Tfh) cell responses and germinal center (GC) B cell development in a mouse model. In the current report, a mouse immunogenicity study was conducted to further ask whether DNA immunization is able to elicit antigen-specific B cell responses. Using HIV-1 Env as model antigen delivered in the form of DNA prime-protein boost, our data demonstrated that DNA prime was able to enhance the antigen-specific B cell responses for both Env-specific antibody secreting cells (ASC) and memory B cells. Furthermore, the DNA priming can greatly reduce the need of including an adjuvant as part of the recombinant protein vaccine boost formulation. Our findings revealed one mechanism that supports the value of DNA priming in assisting the inductin of high affinity and long lasting antigen specific antibody responses.
Collapse
Affiliation(s)
- Haiying Li
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Shixia Wang
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Guangnan Hu
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Lu Zhang
- Department of General Surgery, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | | | - Shan Lu
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
16
|
Coelho-Rocha ND, Barroso FAL, Tavares LM, Dos Santos ESS, Azevedo V, Drumond MM, Mancha-Agresti P. Main Features of DNA-Based Vectors for Use in Lactic Acid Bacteria and Update Protocols. Methods Mol Biol 2021; 2197:285-304. [PMID: 32827144 DOI: 10.1007/978-1-0716-0872-2_16] [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] [Indexed: 06/11/2023]
Abstract
DNA vaccines have been used as a promising strategy for delivery of immunogenic and immunomodulatory molecules into the host cells. Although, there are some obstacles involving the capability of the plasmid vector to reach the cell nucleus in great number to promote the expected benefits. In order to improve the delivery and, consequently, increase the expression levels of the target proteins carried by DNA vaccines, alternative methodologies have been explored, including the use of non-pathogenic bacteria as delivery vectors to carry, deliver, and protect the DNA from degradation, enhancing plasmid expression.
Collapse
Affiliation(s)
- Nina D Coelho-Rocha
- Laboratory of Cellular and Molecular Genetics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Fernanda A L Barroso
- Laboratory of Cellular and Molecular Genetics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Laísa M Tavares
- Laboratory of Cellular and Molecular Genetics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Ester S S Dos Santos
- Laboratory of Cellular and Molecular Genetics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Vasco Azevedo
- Laboratory of Cellular and Molecular Genetics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Mariana M Drumond
- Laboratory of Cellular and Molecular Genetics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Center of Federal Education of Minas Gerais (CEFET-MG), Belo Horizonte, Minas Gerais, Brazil
| | - Pamela Mancha-Agresti
- Laboratory of Cellular and Molecular Genetics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
17
|
Hettinga J, Carlisle R. Vaccination into the Dermal Compartment: Techniques, Challenges, and Prospects. Vaccines (Basel) 2020; 8:E534. [PMID: 32947966 PMCID: PMC7564253 DOI: 10.3390/vaccines8030534] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/08/2020] [Accepted: 09/09/2020] [Indexed: 01/06/2023] Open
Abstract
In 2019, an 'influenza pandemic' and 'vaccine hesitancy' were listed as two of the top 10 challenges to global health by the WHO. The skin is a unique vaccination site, due to its immune-rich milieu, which is evolutionarily primed to respond to challenge, and its ability to induce both humoral and cellular immunity. Vaccination into this dermal compartment offers a way of addressing both of the challenges presented by the WHO, as well as opening up avenues for novel vaccine formulation and dose-sparing strategies to enter the clinic. This review will provide an overview of the diverse range of vaccination techniques available to target the dermal compartment, as well as their current state, challenges, and prospects, and touch upon the formulations that have been developed to maximally benefit from these new techniques. These include needle and syringe techniques, microneedles, DNA tattooing, jet and ballistic delivery, and skin permeabilization techniques, including thermal ablation, chemical enhancers, ablation, electroporation, iontophoresis, and sonophoresis.
Collapse
Affiliation(s)
| | - Robert Carlisle
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford OX3 7DQ, UK;
| |
Collapse
|
18
|
Benson HAE, Grice JE, Mohammed Y, Namjoshi S, Roberts MS. Topical and Transdermal Drug Delivery: From Simple Potions to Smart Technologies. Curr Drug Deliv 2019; 16:444-460. [PMID: 30714524 PMCID: PMC6637104 DOI: 10.2174/1567201816666190201143457] [Citation(s) in RCA: 165] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 01/16/2019] [Accepted: 01/25/2019] [Indexed: 01/02/2023]
Abstract
This overview on skin delivery considers the evolution of the principles of percutaneous ab-sorption and skin products from ancient times to today. Over the ages, it has been recognised that products may be applied to the skin for either local or systemic effects. As our understanding of the anatomy and physiology of the skin has improved, this has facilitated the development of technologies to effectively and quantitatively deliver solutes across this barrier to specific target sites in the skin and beyond. We focus on these technologies and their role in skin delivery today and in the future.
Collapse
Affiliation(s)
- Heather A E Benson
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University of Technology, Perth, Australia
| | - Jeffrey E Grice
- Diamantina Institute, The University of Queensland, Translational Research Institute, QLD, 4102, Australia
| | - Yousuf Mohammed
- Diamantina Institute, The University of Queensland, Translational Research Institute, QLD, 4102, Australia
| | - Sarika Namjoshi
- Diamantina Institute, The University of Queensland, Translational Research Institute, QLD, 4102, Australia
| | - Michael S Roberts
- Diamantina Institute, The University of Queensland, Translational Research Institute, QLD, 4102, Australia.,School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| |
Collapse
|
19
|
DNA vaccine based on conserved HA-peptides induces strong immune response and rapidly clears influenza virus infection from vaccinated pigs. PLoS One 2019; 14:e0222201. [PMID: 31553755 PMCID: PMC6760788 DOI: 10.1371/journal.pone.0222201] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 08/24/2019] [Indexed: 01/08/2023] Open
Abstract
Swine influenza virus (SIVs) infections cause a significant economic impact to the pork industry. Moreover, pigs may act as mixing vessel favoring genome reassortment of diverse influenza viruses. Such an example is the pandemic H1N1 (pH1N1) virus that appeared in 2009, harboring a combination of gene segments from avian, pig and human lineages, which rapidly reached pandemic proportions. In order to confront and prevent these possible emergences as well as antigenic drift phenomena, vaccination remains of vital importance. The present work aimed to evaluate a new DNA influenza vaccine based on distinct conserved HA-peptides fused with flagellin and applied together with Diluvac Forte as adjuvant using a needle-free device (IntraDermal Application of Liquids, IDAL®). Two experimental pig studies were performed to test DNA-vaccine efficacy against SIVs in pigs. In the first experiment, SIV-seronegative pigs were vaccinated with VC4-flagellin DNA and intranasally challenged with a pH1N1. In the second study, VC4-flagellin DNA vaccine was employed in SIV-seropositive animals and challenged intranasally with an H3N2 SIV-isolate. Both experiments demonstrated a reduction in the viral shedding after challenge, suggesting vaccine efficacy against both the H1 and H3 influenza virus subtypes. In addition, the results proved that maternally derived antibodies (MDA) did not constitute an obstacle to the vaccine approach used. Moreover, elevated titers in antibodies both against H1 and H3 proteins in serum and in bronchoalveolar lavage fluids (BALFs) was detected in the vaccinated animals along with a markedly increased mucosal IgA response. Additionally, vaccinated animals developed stronger neutralizing antibodies in BALFs and higher inhibiting hemagglutination titers in sera against both the pH1N1 and H3N2 influenza viruses compared to unvaccinated, challenged-pigs. It is proposed that the described DNA-vaccine formulation could potentially be used as a multivalent vaccine against SIV infections.
Collapse
|
20
|
Yong CY, Ong HK, Tang HC, Yeap SK, Omar AR, Ho KL, Tan WS. Infectious hematopoietic necrosis virus: advances in diagnosis and vaccine development. PeerJ 2019; 7:e7151. [PMID: 31341728 PMCID: PMC6640626 DOI: 10.7717/peerj.7151] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 05/20/2019] [Indexed: 12/16/2022] Open
Abstract
The aquaculture of salmonid fishes is a multi-billion dollar industry with production over 3 million tons annually. However, infectious hematopoietic necrosis virus (IHNV), which infects and kills salmon and trout, significantly reduces the revenue of the salmon farming industry. Currently, there is no effective treatment for IHNV infected fishes; therefore, early detection and depopulation of the infected fishes remain the most common practices to contain the spread of IHNV. Apart from hygiene practices in aquaculture and isolation of infected fishes, loss of fishes due to IHNV infection can also be significantly reduced through vaccination programs. In the current review, some of the diagnostic methods for IHNV, spanning from clinical diagnosis to cell culture, serological and molecular methods are discussed in detail. In addition, some of the most significant candidate vaccines for IHNV are also extensively discussed, particularly the DNA vaccines.
Collapse
Affiliation(s)
- Chean Yeah Yong
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia.,Laboratory of Vaccines and Immunotherapeutics, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Hui Kian Ong
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Hooi Chia Tang
- Laboratory of Vaccines and Immunotherapeutics, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Swee Keong Yeap
- China ASEAN College of Marine Sciences, Xiamen University Malaysia, Sepang, Selangor, Malaysia
| | - Abdul Rahman Omar
- Laboratory of Vaccines and Immunotherapeutics, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Kok Lian Ho
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Wen Siang Tan
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia.,Laboratory of Vaccines and Immunotherapeutics, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| |
Collapse
|
21
|
Liu S, Wang S, Lu S. Using DNA Immunization to Elicit Monoclonal Antibodies in Mice, Rabbits, and Humans. Hum Gene Ther 2019; 29:997-1003. [PMID: 30027758 DOI: 10.1089/hum.2018.077] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
In recent vaccine studies, DNA immunization was found to effectively stimulate both innate and adaptive immunities to elicit high levels of antigen-specific antibody responses. The DNA molecule itself can activate multiple pathways of innate immunity. The in vivo production of antigens allows for presentation by major histocompatibility complexes, so that T-cell responses are generated to help in the development of antigen-specific B cells, leading to high-affinity antibody response. By using the same process, DNA immunization should also be effective at producing functionally potent monoclonal antibodies (mAbs). Furthermore, the in vivo expressed proteins can maximally maintain the native structures and go through appropriate post-transcriptional modifications. By combining such advantages, DNA immunization can be expected to play more important roles in the future to elicit mAbs against difficult targets from a wide range of host systems. The current report shares our group's experience in using DNA immunization to elicit mAbs in the mouse, rabbit, and human models.
Collapse
Affiliation(s)
| | - Shixia Wang
- 2 Department of Medicine, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Shan Lu
- 2 Department of Medicine, University of Massachusetts Medical School , Worcester, Massachusetts
| |
Collapse
|
22
|
Dingman R, Balu-Iyer SV. Immunogenicity of Protein Pharmaceuticals. J Pharm Sci 2019; 108:1637-1654. [PMID: 30599169 PMCID: PMC6720129 DOI: 10.1016/j.xphs.2018.12.014] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 12/19/2018] [Accepted: 12/20/2018] [Indexed: 02/07/2023]
Abstract
Protein therapeutics have drastically changed the landscape of treatment for many diseases by providing a regimen that is highly specific and lacks many off-target toxicities. The clinical utility of many therapeutic proteins has been undermined by the potential development of unwanted immune responses against the protein, limiting their efficacy and negatively impacting its safety profile. This review attempts to provide an overview of immunogenicity of therapeutic proteins, including immune mechanisms and factors influencing immunogenicity, impact of immunogenicity, preclinical screening methods, and strategies to mitigate immunogenicity.
Collapse
Affiliation(s)
- Robert Dingman
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214
| | - Sathy V Balu-Iyer
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214.
| |
Collapse
|
23
|
Abstract
Non-viral gene delivery to skeletal muscle was one of the first applications of gene therapy that went into the clinic, mainly because skeletal muscle is an easily accessible tissue for local gene transfer and non-viral vectors have a relatively safe and low immunogenic track record. However, plasmid DNA, naked or complexed to the various chemistries, turn out to be moderately efficient in humans when injected locally and very inefficient (and very toxic in some cases) when injected systemically. A number of clinical applications have been initiated however, based on transgenes that were adapted to good local impact and/or to a wide physiological outcome (i.e., strong humoral and cellular immune responses following the introduction of DNA vaccines). Neuromuscular diseases seem more challenging for non-viral vectors. Nevertheless, the local production of therapeutic proteins that may act distantly from the injected site and/or the hydrodynamic perfusion of safe plasmids remains a viable basis for the non-viral gene therapy of muscle disorders, cachexia, as well as peripheral neuropathies.
Collapse
|
24
|
Sisteré-Oró M, Vergara-Alert J, Stratmann T, López-Serrano S, Pina-Pedrero S, Córdoba L, Pérez-Maillo M, Pleguezuelos P, Vidal E, Veljkovic V, Segalés J, Nielsen J, Fomsgaard A, Darji A. Conserved HA-peptide NG34 formulated in pCMV-CTLA4-Ig reduces viral shedding in pigs after a heterosubtypic influenza virus SwH3N2 challenge. PLoS One 2019; 14:e0212431. [PMID: 30822308 PMCID: PMC6396909 DOI: 10.1371/journal.pone.0212431] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 02/01/2019] [Indexed: 11/19/2022] Open
Abstract
Swine influenza viruses (SIVs), the causal agents of swine influenza, are not only important to control due to the economic losses in the swine industry, but also can be pandemic pathogens. Vaccination is one of the most relevant strategies to control and prevent influenza infection. Current human vaccines against influenza induce strain-specific immunity and annual update is required due to the virus antigenic shift phenomena. Previously, our group has reported the use of conserved hemagglutinin peptides (HA-peptides) derived from H1-influenza virus as a potential multivalent vaccine candidate. Immunization of swine with these HA-peptides elicited antibodies that recognized and neutralized heterologous influenza viruses in vitro and demonstrated strong hemagglutination-inhibiting activity. In the present work, we cloned one HA-peptide (named NG34) into a plasmid fused with cytotoxic T lymphocyte-associated antigen (CTLA4) which is a molecule that modifies T cell activation and with an adjuvant activity interfering with the adaptive immune response. The resulting plasmid, named pCMV-CTLA4-Ig-NG34, was administered twice to animals employing a needle-free delivery approach. Two studies were carried out to test the efficacy of pCMV-CTLA4-Ig-NG34 as a potential swine influenza vaccine, one in seronegative and another in seropositive pigs against SIV. The second one was aimed to evaluate whether pCMV-CTLA4-Ig-NG34 vaccination would overcome maternally derived antibodies (MDA). After immunization, all animals were intranasally challenged with an H3N2 influenza strain. A complete elimination or significant reduction in the viral shedding was observed within the first week after the challenge in the vaccinated animals from both studies. In addition, no challenged heterologous virus load was detected in the airways of vaccinated pigs. Overall, it is suggested that the pCMV-CTLA4-Ig-NG34 vaccine formulation could potentially be used as a multivalent vaccine against influenza viruses.
Collapse
Affiliation(s)
- Marta Sisteré-Oró
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Júlia Vergara-Alert
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Thomas Stratmann
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Sergi López-Serrano
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Sonia Pina-Pedrero
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Lorena Córdoba
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Mónica Pérez-Maillo
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Patrícia Pleguezuelos
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Enric Vidal
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Veljko Veljkovic
- Centre for Multidisciplinary Research, Institute of Nuclear Sciences VINCA, Belgrade, Serbia
| | - Joaquim Segalés
- UAB, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- Departament de Sanitat i Anatomia Animals, Facultat de Veterinària, UAB, Bellaterra (Cerdanyola del Vallès), Barcelona, Spain
| | - Jens Nielsen
- Virus Research and Development Laboratory, Department of Virus and Microbiological Special Diagnostics, Statens Serum Institut, Copenhagen S, Denmark
| | - Anders Fomsgaard
- Virus Research and Development Laboratory, Department of Virus and Microbiological Special Diagnostics, Statens Serum Institut, Copenhagen S, Denmark
| | - Ayub Darji
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- * E-mail:
| |
Collapse
|
25
|
Khalili S, Rasaee MJ, Bamdad T, Mard-Soltani M, Asadi Ghalehni M, Jahangiri A, Pouriayevali MH, Aghasadeghi MR, Malaei F. A Novel Molecular Design for a Hybrid Phage-DNA Construct Against DKK1. Mol Biotechnol 2018; 60:833-842. [PMID: 30182325 DOI: 10.1007/s12033-018-0115-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Nucleic acid immunization has recently exhibited a great promise for immunotherapy of various diseases. However, it is now clear that powerful strategies are imminently needed to improve their efficiency. In this regard, whole bacteriophage particles have been described as efficient DNA vaccine delivery vehicles, capable of circumventing the limitations of naked DNA immunization. Moreover, phage particles could be engineered to display specific peptides on their surfaces. Given these inherent characteristics of phages, we have designed a novel hybrid phage-DNA immunization vector using both M13 and pAAV plasmid elements. Following the construction and in vitro confirmation of the designed vectors, they were used for comparative mice immunization, carrying the same DNA sequence. The results indicated the efficacy of the designed hybrid phage particles, to elicit higher humoral immunity, in comparison to conventional DNA-immunization vectors (pCI). In light of these findings, it could be concluded that using adeno-associated virus (AAV) expression cassette along with displaying TAT peptide on the surface of the phage particle could be deemed as an appealing strategy to enhance the DNA-immunization and vaccination efficacy.
Collapse
Affiliation(s)
- Saeed Khalili
- Department of Biology Sciences, Shahid Rajaee Teacher Training University, Tehran, Iran
- Department of Medical Biotechnology, Faculty of Medical Sciences, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohamad Javad Rasaee
- Department of Medical Biotechnology, Faculty of Medical Sciences, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Taravat Bamdad
- Department of Virology, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Maysam Mard-Soltani
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Dezful University of Medical Sciences, Dezful, Iran
| | - Majid Asadi Ghalehni
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Abolfazl Jahangiri
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | | | | - Fatemeh Malaei
- Department of Medical Biotechnology, Faculty of Medical Sciences, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
26
|
Zhang S, Zhao S, Jin X, Wang B, Zhao G. Microneedles Improve the Immunogenicity of DNA Vaccines. Hum Gene Ther 2018; 29:1004-1010. [PMID: 29968486 DOI: 10.1089/hum.2018.073] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
DNA vaccines can elicit both humoral and cellular immune responses in mice. However, their poor immunogenicity is a major obstacle toward clinical applications. Improving the efficiency of delivery of DNA vaccines has become a key issue. Vaccination via microneedles penetrating the epidermis can dramatically enhance the stimulation of immune responses. This study showed that by using microneedles to deliver DNA vaccines, gene expression and corresponding immune responses were greatly improved compared to conventional needle injection. The quantitative analysis of gene expressions was made at 6, 24, 72, and 144 h after the DNA delivery. DNA expression levels increased in a time-dependent manner and were substantially greater than with syringe injection after 6 h and 24 h. This elevated expression was followed by markedly enhanced immune responses, with 6-10 times higher levels of antibody and T-cell responses.
Collapse
Affiliation(s)
- Shijie Zhang
- Key Laboratory of Medical Molecular Virology of the Ministry of Health and Ministry of Education, School of Basic Medical Sciences, Fudan University , Shanghai, People's Republic of China
| | - Shushu Zhao
- Key Laboratory of Medical Molecular Virology of the Ministry of Health and Ministry of Education, School of Basic Medical Sciences, Fudan University , Shanghai, People's Republic of China
| | - Xiang Jin
- Key Laboratory of Medical Molecular Virology of the Ministry of Health and Ministry of Education, School of Basic Medical Sciences, Fudan University , Shanghai, People's Republic of China
| | - Bin Wang
- Key Laboratory of Medical Molecular Virology of the Ministry of Health and Ministry of Education, School of Basic Medical Sciences, Fudan University , Shanghai, People's Republic of China
| | - Gan Zhao
- Key Laboratory of Medical Molecular Virology of the Ministry of Health and Ministry of Education, School of Basic Medical Sciences, Fudan University , Shanghai, People's Republic of China
| |
Collapse
|
27
|
Chen WC, Murawsky CM. Strategies for Generating Diverse Antibody Repertoires Using Transgenic Animals Expressing Human Antibodies. Front Immunol 2018; 9:460. [PMID: 29563917 PMCID: PMC5845867 DOI: 10.3389/fimmu.2018.00460] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 02/21/2018] [Indexed: 01/14/2023] Open
Abstract
Therapeutic molecules derived from antibodies have become a dominant class of drugs used to treat human disease. Increasingly, therapeutic antibodies are discovered using transgenic animal systems that have been engineered to express human antibodies. While the engineering details differ, these platforms share the ability to raise an immune response that is comprised of antibodies with fully human idiotypes. Although the predominant transgenic host species has been mouse, the genomes of rats, rabbits, chickens, and cows have also been modified to express human antibodies. The creation of transgenic animal platforms expressing human antibody repertoires has revolutionized therapeutic antibody drug discovery. The observation that the immune systems of these animals are able to recognize and respond to a wide range of therapeutically relevant human targets has led to a surge in antibody-derived drugs in current development. While the clinical success of fully human monoclonal antibodies derived from transgenic animals is well established, recent trends have seen increasingly stringent functional design goals and a shift in difficulty as the industry attempts to tackle the next generation of disease-associated targets. These challenges have been met with a number of novel approaches focused on the generation of large, high-quality, and diverse antibody repertoires. In this perspective, we describe some of the strategies and considerations we use for manipulating the immune systems of transgenic animal platforms (such as XenoMouse®) with a focus on maximizing the diversity of the primary response and steering the ensuing antibody repertoire toward a desired outcome.
Collapse
Affiliation(s)
- Weihsu C Chen
- Biologics Discovery, Department of Therapeutic Discovery, Amgen British Columbia Inc., Burnaby, BC, Canada
| | - Christopher M Murawsky
- Biologics Discovery, Department of Therapeutic Discovery, Amgen British Columbia Inc., Burnaby, BC, Canada
| |
Collapse
|
28
|
Tian M, Zhou Z, Tan S, Fan X, Li L, Ullah N. Formulation in DDA-MPLA-TDB Liposome Enhances the Immunogenicity and Protective Efficacy of a DNA Vaccine against Mycobacterium tuberculosis Infection. Front Immunol 2018. [PMID: 29535714 PMCID: PMC5835323 DOI: 10.3389/fimmu.2018.00310] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Despite the vaccine Mycobacterium bovis Bacillus Calmette–Guérin is used worldwide, tuberculosis (TB) remains the first killer among infectious diseases. An effective vaccine is urgently required. DNA vaccine has shown prophylactic as well as therapeutic effects against TB, while its weak immunogenicity hinders the application. As a strong inducer of Th1-biased immune response, DMT, consisting of dimethyldioctadecylammonium (DDA) and two pattern recognition receptor agonists monophosphoryl lipid A and trehalose 6,6′-dibehenate (TDB), was a newly developed liposomal adjuvant. To elucidate the action mechanism of DMT and improve immunological effects induced by DNA vaccine, a new recombinant eukaryotic expression plasmid pCMFO that secretes the fusion of four multistage antigens (Rv2875, Rv3044, Rv2073c, and Rv0577) of Mycobacterium tuberculosis was constructed. pCMFO/DDA and pCMFO/DMT complexes were then prepared and their physicochemical properties were analyzed. The immunogenicity and protection against M. tuberculosis infection in vaccinated C57BL/6 mice were compared. Formulation of DNA and two agonists into the DDA liposome decreased zeta potential but increased the stability of storage, which resulted in a slower and longer-lasting release of DNA from the DNA–DMT complex than the DNA–DDA liposome. Besides Th1-biased responses, pCMFO/DMT vaccinated mice elicited more significantly CFMO-specific IL2+ TCM cell responses in the spleen and provided an enhanced and persistent protection against M. tuberculosis aerosol infection, compared to pCMFO/DDA and pCMFO groups. Therefore, the adjuvant DMT can release DNA and agonists slowly, which might attribute to the improved protection of DMT adjuvanted vaccines. pCMFO/DMT, a very promising TB vaccine, warrants for further preclinical and clinical trials.
Collapse
Affiliation(s)
- Maopeng Tian
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zijie Zhou
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Songwei Tan
- Tongji School of Pharmacy, National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan, China
| | - Xionglin Fan
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Longmeng Li
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Nadeem Ullah
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
29
|
Hansen DT, Craciunescu FM, Fromme P, Johnston SA, Sykes KF. Generation of High-Specificity Antibodies against Membrane Proteins Using DNA-Gold Micronanoplexes for Gene Gun Immunization. ACTA ACUST UNITED AC 2018. [PMID: 29516482 DOI: 10.1002/cpps.50] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Membrane proteins are the molecular interface of the cell and its environs; however, studies of membrane proteins are highly technically challenging, mainly due to instability of the isolated protein. Towards the production of antibodies that recognize properly folded and stabilized forms of membrane protein antigen, we describe a DNA-based immunization method for mice that expresses the antigen in the membranes of dendritic cells, thus allowing direct presentation to the immune system. This genetic immunization approach employs a highly efficient method of biolistic delivery based on DNA-gold micronanoplexes, which are complexes of micron-sized gold particles that allow dermal penetration and nanometer-sized gold particles that provide a higher surface area for DNA binding than micron gold alone. In contrast to antibodies derived from immunizations with detergent-solubilized protein or with protein fragments, antibodies from genetic immunization are expected to have a high capacity for binding conformational epitopes and for modulating membrane protein activity. © 2018 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Debra T Hansen
- Biodesign Center for Applied Structural Discovery, Arizona State University, Tempe, Arizona
| | - Felicia M Craciunescu
- Biodesign Center for Applied Structural Discovery, Arizona State University, Tempe, Arizona
| | - Petra Fromme
- Biodesign Center for Applied Structural Discovery, Arizona State University, Tempe, Arizona
| | - Stephen A Johnston
- Biodesign Center for Innovations in Medicine, Arizona State University, Tempe, Arizona
| | - Kathryn F Sykes
- Biodesign Center for Innovations in Medicine, Arizona State University, Tempe, Arizona.,Current address: HealthTell, Inc, Chandler, Arizona
| |
Collapse
|
30
|
Peng B, Peng N, Zhang Y, Zhang F, Li X, Chang H, Fang F, Wang F, Lu F, Chen Z. Comparison of the Protective Efficacy of Neutralizing Epitopes of 2009 Pandemic H1N1 Influenza Hemagglutinin. Front Immunol 2017; 8:1070. [PMID: 28912784 PMCID: PMC5583165 DOI: 10.3389/fimmu.2017.01070] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 08/16/2017] [Indexed: 12/03/2022] Open
Abstract
The 2009 H1N1 influenza (Pdm09) pandemic has been referred to as the first influenza pandemic of the twenty-first century. There is a marked difference in antigenicity between the pandemic H1N1 virus and past seasonal H1N1 viruses, which allowed the pandemic virus to spread rapidly in humans. Antibodies (Abs) against hemagglutinin (HA), especially neutralizing Abs against epitopes in the head of HA, play critical roles in defending the host against the virus. Some preexisting neutralizing Abs that recognize neutralizing epitopes of Pdm09 HA, thereby affording cross-protection, have been reported. To better understand the protective effects of epitopes in Pdm09 HA, we constructed a series of plasmid DNAs (DNA vaccines) by cloning various combinations of Pdm09 neutralizing epitopes into the HA backbone derived from A/PR/8/1934 (H1N1). We subsequently compared the protective immune responses induced by these various forms of HA in a mouse model. We found that the plasmid DNAs with epitope substitutions provided better protection against lethal virus challenge and induced higher strain-specific antibody titers, with epitope Sa being the most effective. Moreover, the combination of epitopes Sa and Sb provided almost complete protection in mice. These findings provide new insights into the protective efficacy of neutralizing epitopes of influenza HA.
Collapse
Affiliation(s)
- Bo Peng
- College of Life Science, Hunan Normal University, Changsha, China
| | - Na Peng
- College of Life Science, Hunan Normal University, Changsha, China
| | - Yanan Zhang
- College of Life Science, Hunan Normal University, Changsha, China
| | - Fenghua Zhang
- College of Life Science, Hunan Normal University, Changsha, China
| | - Xuguang Li
- Centre for Biologics Evaluation, Biologics and Genetic Therapies Directorate, Health Products and Food Branch, Health Canada, Ottawa, ON, Canada
| | - Haiyan Chang
- College of Life Science, Hunan Normal University, Changsha, China
| | - Fang Fang
- College of Life Science, Hunan Normal University, Changsha, China
| | - Fuyan Wang
- Department of Immunology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Fangguo Lu
- School of Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Ze Chen
- College of Life Science, Hunan Normal University, Changsha, China.,Shanghai Institute of Biological Products, Shanghai, China
| |
Collapse
|
31
|
Karlsson I, Borggren M, Nielsen J, Christensen D, Williams J, Fomsgaard A. Increased humoral immunity by DNA vaccination using an α-tocopherol-based adjuvant. Hum Vaccin Immunother 2017; 13:1823-1830. [PMID: 28613978 DOI: 10.1080/21645515.2017.1321183] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
DNA vaccines induce broad immunity, which involves both humoral and strong cellular immunity, and can be rapidly designed for novel or evolving pathogens such as influenza. However, the humoral immunogenicity in humans and higher animals has been suboptimal compared with that of traditional vaccine approaches. We tested whether the emulsion-based and α-tocopherol containing adjuvant Diluvac Forte® has the ability to enhance the immunogenicity of a naked DNA vaccine (i.e., plasmid DNA). As a model vaccine, we used plasmids encoding both a surface-exposed viral glycoprotein (hemagglutinin) and an internal non-glycosylated nucleoprotein in the Th1/Th2 balanced CB6F1 mouse model. The naked DNA (50 µg) was premixed at a 1:1 volume/volume ratio with Diluvac Forte®, an emulsion containing different concentrations of α-tocopherol, the emulsion alone or endotoxin-free phosphate-buffered saline (PBS). The animals received 2 intracutaneous immunizations spaced 3 weeks apart. When combined with Diluvac Forte® or the emulsion containing α-tocopherol, the DNA vaccine induced a more potent and balanced immunoglobulin G (IgG)1 and IgG2c response, and both IgG subclass responses were significantly enhanced by the adjuvant. The DNA vaccine also induced CD4+ and CD8+ vaccine-specific T cells; however, the adjuvant did not exert a significant impact. We concluded that the emulsion-based adjuvant Diluvac Forte® enhanced the immunogenicity of a naked DNA vaccine encoding influenza proteins and that the adjuvant constituent α-tocopherol plays an important role in this immunogenicity. This induction of a potent and balanced humoral response without impairment of cellular immunity constitutes an important advancement toward effective DNA vaccines.
Collapse
Affiliation(s)
- Ingrid Karlsson
- a Virus Research and Development Laboratory, Department of Virus and Microbiological Special Diagnostics , Statens Serum Institut , Copenhagen , Denmark
| | - Marie Borggren
- a Virus Research and Development Laboratory, Department of Virus and Microbiological Special Diagnostics , Statens Serum Institut , Copenhagen , Denmark
| | - Jens Nielsen
- a Virus Research and Development Laboratory, Department of Virus and Microbiological Special Diagnostics , Statens Serum Institut , Copenhagen , Denmark
| | - Dennis Christensen
- b Department of Infectious Disease Immunology, Vaccine Adjuvant Research , Statens Serum Institut , Copenhagen , Denmark
| | - Jim Williams
- c Nature Technology Corporation , Lincoln , NE , USA
| | - Anders Fomsgaard
- a Virus Research and Development Laboratory, Department of Virus and Microbiological Special Diagnostics , Statens Serum Institut , Copenhagen , Denmark.,d Infectious Disease Research Unit, Clinical Institute , University of Southern Denmark , Odense , Denmark
| |
Collapse
|
32
|
Bertran K, Balzli C, Lee DH, Suarez DL, Kapczynski DR, Swayne DE. Protection of White Leghorn chickens by U.S. emergency H5 vaccination against clade 2.3.4.4 H5N2 high pathogenicity avian influenza virus. Vaccine 2017; 35:6336-6344. [PMID: 28554502 DOI: 10.1016/j.vaccine.2017.05.051] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 05/01/2017] [Accepted: 05/16/2017] [Indexed: 10/19/2022]
Abstract
During December 2014-June 2015, the U.S. experienced a high pathogenicity avian influenza (HPAI) outbreak caused by clade 2.3.4.4 H5Nx Goose/Guangdong lineage viruses with devastating consequences for the poultry industry. Three vaccines, developed based on updating existing registered vaccines or currently licensed technologies, were evaluated for possible use: an inactivated reverse genetics H5N1 vaccine (rgH5N1) and an RNA particle vaccine (RP-H5), both containing the hemagglutinin gene of clade 2.3.4.4 strain, and a recombinant herpesvirus turkey vectored vaccine (rHVT-H5) containing the hemagglutinin gene of clade 2.2 strain. The efficacy of the three vaccines, alone or in combination, was assessed in White Leghorn chickens against clade 2.3.4.4 H5N2 HPAI virus challenge. In Study 1, single (rHVT-H5) and prime-boost (rHVT-H5+rgH5N1 or rHVT-H5+RP-H5) vaccination strategies protected chickens with high levels of protective immunity and significantly reduced virus shedding. In Study 2, single vaccination with either rgH5N1 or RP-H5 vaccines provided clinical protection in adult chickens and significantly reduced virus shedding. In Study 3, double rgH5N1 vaccination protected adult chickens from clinical signs and mortality when challenged 20weeks post-boost, with high levels of long-lasting protective immunity and significantly reduced virus shedding. These studies support the use of genetically related vaccines, possibly in combination with a broad protective priming vaccine, for emergency vaccination programs against clade 2.3.4.4 H5Nx HPAI virus in young and adult layer chickens.
Collapse
Affiliation(s)
- Kateri Bertran
- Exotic and Emerging Avian Viral Diseases Research Unit, Southeast Poultry Research Laboratory, US National Poultry Research Center, Agricultural Research Service, US Department of Agriculture, 934, College Station Rd, Athens, GA 30605, United States.
| | - Charles Balzli
- Exotic and Emerging Avian Viral Diseases Research Unit, Southeast Poultry Research Laboratory, US National Poultry Research Center, Agricultural Research Service, US Department of Agriculture, 934, College Station Rd, Athens, GA 30605, United States.
| | - Dong-Hun Lee
- Exotic and Emerging Avian Viral Diseases Research Unit, Southeast Poultry Research Laboratory, US National Poultry Research Center, Agricultural Research Service, US Department of Agriculture, 934, College Station Rd, Athens, GA 30605, United States.
| | - David L Suarez
- Exotic and Emerging Avian Viral Diseases Research Unit, Southeast Poultry Research Laboratory, US National Poultry Research Center, Agricultural Research Service, US Department of Agriculture, 934, College Station Rd, Athens, GA 30605, United States.
| | - Darrell R Kapczynski
- Exotic and Emerging Avian Viral Diseases Research Unit, Southeast Poultry Research Laboratory, US National Poultry Research Center, Agricultural Research Service, US Department of Agriculture, 934, College Station Rd, Athens, GA 30605, United States.
| | - David E Swayne
- Exotic and Emerging Avian Viral Diseases Research Unit, Southeast Poultry Research Laboratory, US National Poultry Research Center, Agricultural Research Service, US Department of Agriculture, 934, College Station Rd, Athens, GA 30605, United States.
| |
Collapse
|
33
|
Bagley KC, Schwartz JA, Andersen H, Eldridge JH, Xu R, Ota-Setlik A, Geltz JJ, Halford WP, Fouts TR. An Interleukin 12 Adjuvanted Herpes Simplex Virus 2 DNA Vaccine Is More Protective Than a Glycoprotein D Subunit Vaccine in a High-Dose Murine Challenge Model. Viral Immunol 2017; 30:178-195. [PMID: 28085634 DOI: 10.1089/vim.2016.0136] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Vaccination is a proven intervention against human viral diseases; however, success against Herpes Simplex Virus 2 (HSV-2) remains elusive. Most HSV-2 vaccines tested in humans to date contained just one or two immunogens, such as the virion attachment receptor glycoprotein D (gD) and/or the envelope fusion protein, glycoprotein B (gB). At least three factors may have contributed to the failures of subunit-based HSV-2 vaccines. First, immune responses directed against one or two viral antigens may lack sufficient antigenic breadth for efficacy. Second, the antibody responses elicited by these vaccines may have lacked necessary Fc-mediated effector functions. Third, these subunit vaccines may not have generated necessary protective cellular immune responses. We hypothesized that a polyvalent combination of HSV-2 antigens expressed from a DNA vaccine with an adjuvant that polarizes immune responses toward a T helper 1 (Th1) phenotype would compose a more effective vaccine. We demonstrate that delivery of DNA expressing full-length HSV-2 glycoprotein immunogens by electroporation with the adjuvant interleukin 12 (IL-12) generates substantially greater protection against a high-dose HSV-2 vaginal challenge than a recombinant gD subunit vaccine adjuvanted with alum and monophosphoryl lipid A (MPL). Our results further show that DNA vaccines targeting optimal combinations of surface glycoproteins provide better protection than gD alone and provide similar survival benefits and disease symptom reductions compared with a potent live attenuated HSV-2 0ΔNLS vaccine, but that mice vaccinated with HSV-2 0ΔNLS clear the virus much faster. Together, our data indicate that adjuvanted multivalent DNA vaccines hold promise for an effective HSV-2 vaccine, but that further improvements may be required.
Collapse
Affiliation(s)
| | | | | | | | - Rong Xu
- 3 Profectus Biosciences , Tarrytown, New York
| | | | - Joshua J Geltz
- 4 Department of Microbiology and Immunology, Southern Illinois University School of Medicine , Springfield, Illinois
| | - William P Halford
- 4 Department of Microbiology and Immunology, Southern Illinois University School of Medicine , Springfield, Illinois
| | | |
Collapse
|
34
|
Neutrophils are Essential in Short Hairpin RNA of Indoleamine 2,3- Dioxygenase Mediated-antitumor Efficiency. MOLECULAR THERAPY-NUCLEIC ACIDS 2016; 5:e397. [PMID: 27922590 PMCID: PMC5159481 DOI: 10.1038/mtna.2016.105] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 11/01/2016] [Indexed: 02/08/2023]
Abstract
Indoleamine 2,3-dioxygenase (IDO) is a rate limiting enzyme in tryptophan-degrading pathways and IDO activity results in immune suppression. Targeting IDO is a strategy of cancer immunotherapies. Our previous studies demonstrate that delivery of short hairpin against IDO (IDO shRNA) suppresses tumor growth and increases neutrophils infiltration into tumor. Neutrophils reveal antitumorigenic “N1” or protumorigenic “N2” phenotype in tumor microenvironment. However, the function of IDO shRNA-induced neutrophils is not clear. The LLC1 lung cancer model was used to investigate the role of these neutrophils. Intramuscular injection of IDO shRNA or IDO inhibitor treatment delayed tumor growth and both treatments increased neutrophil infiltration in tumor. Enriched tumor-infiltrating neutrophils expressed both high level of tumor necrosis factor-α and tumor necrosis factor-β (N1 and N2 associated molecules, respectively). In addition, IDO shRNA treatment induced interferon-γ and tryptophan transfer RNA expression in splenocytes. Systematic depletion of neutrophils abolished the IDO shRNA-induced therapeutic effect but did not affect the effect of IDO inhibitor. The levels of interferon-γ and tumor necrosis factor-α were suppressed in IDO shRNA treatment splenocytes after neutrophils depletion. In conclusion, these tumor-infiltrating neutrophils show antitumorigenic phenotype in spleen after IDO shRNA treatment in a murine lung cancer model.
Collapse
|
35
|
Bagley K, Xu R, Ota-Setlik A, Egan M, Schwartz J, Fouts T. The catalytic A1 domains of cholera toxin and heat-labile enterotoxin are potent DNA adjuvants that evoke mixed Th1/Th17 cellular immune responses. Hum Vaccin Immunother 2016; 11:2228-40. [PMID: 26042527 PMCID: PMC4635876 DOI: 10.1080/21645515.2015.1026498] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
DNA encoded adjuvants are well known for increasing the magnitude of cellular and/or humoral immune responses directed against vaccine antigens. DNA adjuvants can also tune immune responses directed against vaccine antigens to better protect against infection of the target organism. Two potent DNA adjuvants that have unique abilities to tune immune responses are the catalytic A1 domains of Cholera Toxin (CTA1) and Heat-Labile Enterotoxin (LTA1). Here, we have characterized the adjuvant activities of CTA1 and LTA1 using HIV and SIV genes as model antigens. Both of these adjuvants enhanced the magnitude of antigen-specific cellular immune responses on par with those induced by the well-characterized cytokine adjuvants IL-12 and GM-CSF. CTA1 and LTA1 preferentially enhanced cellular responses to the intracellular antigen SIVmac239-gag over those for the secreted HIVBaL-gp120 antigen. IL-12, GM-CSF and electroporation did the opposite suggesting differences in the mechanisms of actions of these diverse adjuvants. Combinations of CTA1 or LTA1 with IL-12 or GM-CSF generated additive and better balanced cellular responses to both of these antigens. Consistent with observations made with the holotoxin and the CTA1-DD adjuvant, CTA1 and LTA1 evoked mixed Th1/Th17 cellular immune responses. Together, these results show that CTA1 and LTA1 are potent DNA vaccine adjuvants that favor the intracellular antigen gag over the secreted antigen gp120 and evoke mixed Th1/Th17 responses against both of these antigens. The results also indicate that achieving a balanced immune response to multiple intracellular and extracellular antigens delivered via DNA vaccination may require combining adjuvants that have different and complementary mechanisms of action.
Collapse
|
36
|
Borggren M, Nielsen J, Bragstad K, Karlsson I, Krog JS, Williams JA, Fomsgaard A. Vector optimization and needle-free intradermal application of a broadly protective polyvalent influenza A DNA vaccine for pigs and humans. Hum Vaccin Immunother 2016; 11:1983-90. [PMID: 25746201 DOI: 10.1080/21645515.2015.1011987] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The threat posed by the 2009 pandemic H1N1 virus emphasized the need for new influenza A virus vaccines inducing a broad cross-protective immune response for use in both humans and pigs. An effective and broad influenza vaccine for pigs would greatly benefit the pork industry and contribute to public health by diminishing the risk of emerging highly pathogenic reassortants. Current inactivated protein vaccines against swine influenza produce only short-lived immunity and have no efficacy against heterologous strains. DNA vaccines are a potential alternative with advantages such as the induction of cellular and humoral immunity, inherent safety and rapid production time. We have previously developed a DNA vaccine encoding selected influenza proteins of pandemic origin and demonstrated broad protective immune responses in ferrets and pigs. In this study, we evaluated our DNA vaccine expressed by next-generation vectors. These new vectors can improve gene expression, but they are also efficiently produced on large scales and comply with regulatory guidelines by avoiding antibiotic resistance genes. In addition, a new needle-free delivery of the vaccine, convenient for mass vaccinations, was compared with intradermal needle injection followed by electroporation. We report that when our DNA vaccine is expressed by the new vectors and delivered to the skin with the needle-free device in the rabbit model, it can elicit an antibody response with the same titers as a conventional vector with intradermal electroporation. The needle-free delivery is already in use for traditional protein vaccines in pigs but should be considered as a practical alternative for the mass administration of broadly protective influenza DNA vaccines.
Collapse
Key Words
- BSA, bovine serum albumin
- DK, Denmark
- DNA vaccine
- DNA, DeoxyriboNucleic Acid
- ELISA, Enzyme Linked Immunosorbent Assay
- EP, electroporation
- FCS, fetal calf serum
- HA, hemagglutinin
- HAI, hemagglutination inhibition assay
- HAU, hemagglutination units
- HI, hemagglutination inhibition
- IDAL®, IntraDermal Application of Liquids®
- IgG, immunoglobulin G
- M, matrix protein
- MDCK cells, Madin-Darby Canine Kidney epithelial cells
- NA, neuraminidase
- NP, nucleoprotein
- NTC8385-VA1
- NTC9385R
- NZW, New Zealand White
- PBS, phosphate buffered saline
- RDE, receptor destroying enzyme
- SEM, standard error mean
- TMB, tetramethylbenzidine
- US, the United States
- WHO, world health organization
- bp, base pair
- i.d., intra-dermal
- influenza
- needle-free
- polyvalent
- tPA, tissue plasminogen activator
Collapse
Affiliation(s)
- Marie Borggren
- a Virus Research and Development Laboratory ; Department of Microbiological Diagnostic and Virology; Statens Serum Institut ; Copenhagen , Denmark
| | | | | | | | | | | | | |
Collapse
|
37
|
Liu S, Wang S, Lu S. DNA immunization as a technology platform for monoclonal antibody induction. Emerg Microbes Infect 2016; 5:e33. [PMID: 27048742 PMCID: PMC4855071 DOI: 10.1038/emi.2016.27] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Revised: 12/06/2015] [Accepted: 12/15/2015] [Indexed: 01/17/2023]
Abstract
To combat the threat of many emerging infectious diseases, DNA immunization offers a unique and powerful approach to the production of high-quality monoclonal antibodies (mAbs) against various pathogens. Compared with traditional protein-based immunization approaches, DNA immunization is efficient for testing novel immunogen designs, does not require the production or purification of proteins from a pathogen or the use of recombinant protein technology and is effective at generating mAbs against conformation-sensitive targets. Although significant progress in the use of DNA immunization to generate mAbs has been made over the last two decades, the literature does not contain an updated summary of this experience. The current review provides a comprehensive analysis of the literature, including our own work, describing the use of DNA immunization to produce highly functional mAbs, in particular, those against emerging infectious diseases. Critical factors such as immunogen design, delivery approach, immunization schedule, use of immune modulators and the role of final boost immunization are discussed in detail.
Collapse
Affiliation(s)
| | - Shixia Wang
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Shan Lu
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA
| |
Collapse
|
38
|
Zhang L, Jia N, Li J, Han Y, Cao W, Wang S, Huang Z, Lu S. Optimal designs of an HA-based DNA vaccine against H7 subtype influenza viruses. Hum Vaccin Immunother 2016; 10:1949-58. [PMID: 25424804 DOI: 10.4161/hv.28795] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The outbreak of a novel H7N9 influenza virus in 2013 has raised serious concerns for the potential of another avian-source pandemic influenza. Effective vaccines against H7N9 virus are important in the prevention and control of any major outbreak. Novel vaccination technologies are useful additions to existing approaches. In the current report, DNA vaccine studies were conducted to identify the optimal design of an H7 HA antigen using the HA gene from a previously reported H7N7 virus that is lethal in humans as the model antigen. New Zealand White rabbits were immunized with DNA vaccines expressing 1 of 3 forms of H7 HA antigen inserts encoding the HA gene from the same H7N7 virus. High-level H7 HA-specific IgG was detected by ELISA, and functional antibodies were confirmed by hemagglutination inhibition assay and pseudotyped virus-based neutralization assay against viruses expressing HA antigens from either the previous H7N7 virus or the novel H7N9 virus. HA antigen design under the tissue plasminogen activator leader (tPA) was the most immunogenic. The data presented in the current report confirm the immunogenicity of the H7 HA antigen and provide useful guidance to prepare for an optimized H7 HA DNA vaccine to help to control the emerging H7N9 virus if and when it is needed.
Collapse
Affiliation(s)
- Lu Zhang
- a Department of Infectious Diseases; The First Affiliated Hospital with Nanjing Medical University; Nanjing, PR China
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Liu H, Wang S, Jia Y, Li J, Huang Z, Lu S, Xing Y. N-Linked Glycosylation at an Appropriate Position in the Pre-S2 Domain Is Critical for Cellular and Humoral Immunity against Middle HBV Surface Antigen. TOHOKU J EXP MED 2016; 236:131-8. [PMID: 26062906 DOI: 10.1620/tjem.236.131] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Infection with hepatitis B virus (HBV) remains a worldwide health problem, and DNA-based vaccines against HBV have been tested for therapeutic applications. HBV possesses three envelope lipoproteins that are translated from a single reading-frame: large, middle, and small HBV surface antigens. Among these envelope proteins, the middle HBV surface antigen (MHBs) contains a constitutive N-linked glycosylation site at position 4 (Asn4) in the amino-terminal portion (MQWNSTTFHQ) of pre-S2 domain. Asn4 (shown in bold) is essential for secretion of viral particles and conserved among all serotypes of HBV, but its influence on the immunogenicity of MHBs remains unknown. Here, we constructed four MHBs genes carrying mutations, underlined, in the amino-terminal portion of pre-S2 domain. One mutant protein contains Q at position 4 (MQWQSTTFHQ). In addition, each of three mutant MHBs proteins contains a N-linked glycosylation site (N-X-S/T), relocated to position 5 (MQWQNTTFHQ), 6 (MQWQSNTSHQ) or 7 (MQWQSTNFTQ) in pre-S2 domain. The expression and immunogenic properties of mutant DNA vaccines were examined in 293T human renal epithelial cells and in BALB/c mice, respectively. We showed that Asn4 was critical for secretion and immunogenicity of MHBs. Moreover, the MHBs protein that carries a N-linked glycosylation site at position 5 or 7 retained the properties similar to wild-type MHBs. In contrast, the secretion-defective mutant protein carrying Asn at position 6 induced only marginal humoral and cellular immune responses in mice, despite the N-linked glycosylation. In conclusion, N-linked glycosylation at an appropriate position in pre-S2 domain is an essential requirement for DNA vaccine expressing MHBs.
Collapse
Affiliation(s)
- Hao Liu
- Department of Infectious Diseases, The First Affiliated Hospital of Nanjing Medical University, 2) Department of Infectious Diseases, Nanjing Children's Hospital, Affiliated to Nanjing Medical University, Nanjing, China
| | | | | | | | | | | | | |
Collapse
|
40
|
Zhang L, Wang W, Wang S. Effect of vaccine administration modality on immunogenicity and efficacy. Expert Rev Vaccines 2015; 14:1509-23. [PMID: 26313239 DOI: 10.1586/14760584.2015.1081067] [Citation(s) in RCA: 171] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The many factors impacting the efficacy of a vaccine can be broadly divided into three categories: features of the vaccine itself, including immunogen design, vaccine type, formulation, adjuvant and dosing; individual variations among vaccine recipients and vaccine administration-related parameters. While much literature exists related to vaccines, and recently systems biology has started to dissect the impact of individual subject variation on vaccine efficacy, few studies have focused on the role of vaccine administration-related parameters on vaccine efficacy. Parenteral and mucosal vaccinations are traditional approaches for licensed vaccines; novel vaccine delivery approaches, including needless injection and adjuvant formulations, are being developed to further improve vaccine safety and efficacy. This review provides a brief summary of vaccine administration-related factors, including vaccination approach, delivery route and method of administration, to gain a better understanding of their potential impact on the safety and immunogenicity of candidate vaccines.
Collapse
Affiliation(s)
- Lu Zhang
- a 1 Department of Infectious Diseases, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China.,b 2 China-US Vaccine Research Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Wei Wang
- c 3 Wang Biologics, LLC, Chesterfield, MO 63017, USA ; Current affiliation: Bayer HealthCare, Berkeley, CA 94710, USA
| | - Shixia Wang
- d 4 Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| |
Collapse
|
41
|
Tang DCC. A trail blazed through DNA vaccine, noninvasive vaccine, and innate-adaptive immunity duo. Hum Vaccin Immunother 2015; 10:2143-6. [PMID: 25424917 PMCID: PMC4896786 DOI: 10.4161/hv.29044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
42
|
Gonçalves de Assis NR, Batistoni de Morais S, Figueiredo BCP, Ricci ND, de Almeida LA, da Silva Pinheiro C, Martins VDP, Oliveira SC. DNA Vaccine Encoding the Chimeric Form of Schistosoma mansoni Sm-TSP2 and Sm29 Confers Partial Protection against Challenge Infection. PLoS One 2015; 10:e0125075. [PMID: 25942636 PMCID: PMC4420270 DOI: 10.1371/journal.pone.0125075] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 03/19/2015] [Indexed: 12/23/2022] Open
Abstract
Schistosomiasis is an important parasitic disease worldwide that affects more than 207 million people in 76 countries and causes approximately 250,000 deaths per year. The best long-term strategy to control schistosomiasis is through immunization combined with drug treatment. Due to the ability of DNA vaccines to generate humoral and cellular immune responses, such vaccines are considered a promising approach against schistosomiasis. Sm29 and tetraspanin-2 (Sm-TSP2) are two proteins that are located in the S. mansoni tegument of adult worms and schistosomula and induce high levels of protection through recombinant protein immunization. In this study, we transfected BHK-21 cells with plasmids encoding Sm29, Sm-TSP2 or a chimera containing both genes. Using RT-PCR analysis and western blot, we confirmed that the DNA vaccine constructs were transcribed and translated, respectively, in BHK-21 cells. After immunization of mice, we evaluated the reduction in worm burden. We observed worm burden reductions of 17-22%, 22%, 31-32% and 24-32% in animals immunized with the pUMVC3/Sm29, pUMVC3/SmTSP-2, pUMVC3/Chimera and pUMVC3/Sm29 + pUMVC3/SmTSP-2 plasmids, respectively. We evaluated the humoral response elicited by DNA vaccines, and animals immunized with pUMVC3/Sm29 and pUMVC3/Sm29 + pUMVC3/SmTSP-2 showed higher titers of anti-Sm29 antibodies. The cytokine profile produced by the spleen cells of immunized mice was then evaluated. We observed higher production of Th1 cytokines, such as TNF-α and IFN-γ, in vaccinated mice and no significant production of IL-4 and IL-5. The DNA vaccines tested in this study showed the ability to generate a protective immune response against schistosomiasis, probably through the production of Th1 cytokines. However, future strategies aiming to optimize the protective response induced by a chimeric DNA construct need to be developed.
Collapse
Affiliation(s)
- Natan Raimundo Gonçalves de Assis
- Departamento de Bioquímica e Imunologia do Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270–901, Belo Horizonte, MG, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), CNPq MCT, 31270–901, Belo Horizonte, MG, Brazil
| | - Suellen Batistoni de Morais
- Departamento de Bioquímica e Imunologia do Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270–901, Belo Horizonte, MG, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), CNPq MCT, 31270–901, Belo Horizonte, MG, Brazil
| | - Bárbara Castro Pimentel Figueiredo
- Departamento de Bioquímica e Imunologia do Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270–901, Belo Horizonte, MG, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), CNPq MCT, 31270–901, Belo Horizonte, MG, Brazil
| | - Natasha Delaqua Ricci
- Departamento de Bioquímica e Imunologia do Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270–901, Belo Horizonte, MG, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), CNPq MCT, 31270–901, Belo Horizonte, MG, Brazil
| | - Leonardo Augusto de Almeida
- Departamento de Bioquímica e Imunologia do Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270–901, Belo Horizonte, MG, Brazil
| | - Carina da Silva Pinheiro
- Departamento de Biointeração do Instituto de Ciências da Saúde, Universidade Federal da Bahia, 40110–100, Salvador, BA, Brazil
| | | | - Sergio Costa Oliveira
- Departamento de Bioquímica e Imunologia do Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270–901, Belo Horizonte, MG, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), CNPq MCT, 31270–901, Belo Horizonte, MG, Brazil
- * E-mail:
| |
Collapse
|
43
|
Abstract
The key impediment to the successful application of gene therapy in clinics is not the paucity of therapeutic genes. It is rather the lack of nontoxic and efficient strategies to transfer therapeutic genes into target cells. Over the past few decades, considerable progress has been made in gene transfer technologies, and thus far, three different delivery systems have been developed with merits and demerits characterizing each system. Viral and chemical methods of gene transfer utilize specialized carrier to overcome membrane barrier and facilitate gene transfer into cells. Physical methods, on the other hand, utilize various forms of mechanical forces to enforce gene entry into cells. Starting in 1980s, physical methods have been introduced as alternatives to viral and chemical methods to overcome various extra- and intracellular barriers that limit the amount of DNA reaching the intended cells. Accumulating evidence suggests that it is quite feasible to directly translocate genes into cytoplasm or even nuclei of target cells by means of mechanical force, bypassing endocytosis, a common pathway for viral and nonviral vectors. Indeed, several methods have been developed, and the majority of them share the same underlying mechanism of gene transfer, i.e., physically created transient pores in cell membrane through which genes get into cells. Here, we provide an overview of the current status and future research directions in the field of physical methods of gene transfer.
Collapse
|
44
|
Suschak JJ, Wang S, Fitzgerald KA, Lu S. Identification of Aim2 as a sensor for DNA vaccines. THE JOURNAL OF IMMUNOLOGY 2014; 194:630-6. [PMID: 25488991 DOI: 10.4049/jimmunol.1402530] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Recent human study data have re-established the value of DNA vaccines, especially in priming high-level Ag-specific Ab responses, but also raised questions about the mechanisms responsible for such effects. Whereas previous reports have shown involvement of downstream signaling molecules in the innate immune system, the current study investigated the role of absent in melanoma 2 (Aim2) as a sensor for DNA vaccines. The Aim2 inflammasome directs maturation of the proinflammatory cytokines IL-1β and IL-18 and an inflammatory form of cell death called pyroptosis. Both the humoral and cellular Ag-specific adaptive responses were significantly reduced in Aim2-deficient mice in an IL-1β/IL-18-independent manner after DNA vaccination. Surprisingly, Aim2-deficient mice also exhibited significantly lower levels of IFN-α/β at the site of injection. These results indicate a previously unreported link between DNA vaccine-induced pyroptotic cell death and vaccine immunogenicity that is instrumental in shaping the Ag-specific immune response to DNA vaccines.
Collapse
Affiliation(s)
- John J Suschak
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655; and
| | - Shixia Wang
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655; and
| | - Katherine A Fitzgerald
- Program in Innate Immunity, Division of Infectious Diseases, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655
| | - Shan Lu
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655; and
| |
Collapse
|
45
|
Eliciting specific humoral immunity from a plasmid DNA encoding infectious bursal disease virus polyprotein gene fused with avian influenza virus hemagglutinin gene. J Virol Methods 2014; 211:36-42. [PMID: 25445883 DOI: 10.1016/j.jviromet.2014.10.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Revised: 10/16/2014] [Accepted: 10/21/2014] [Indexed: 11/22/2022]
Abstract
DNA vaccine coding for infectious bursal disease virus (IBDV) polyprotein gene and that for avian influenza virus (AIV) hemagglutinin (HA) gene have been shown to induce immunity and provide protection against the respective disease. The present study was carried out to determine whether an IBDV polyprotein gene-based DNA fused with AIV HA gene could trigger immune response to both IBDV and AIV. After transfection, VP2 and HA were detected in the cytoplasm and at cell membrane, respectively, by immunofluorescent antibody double staining method, suggesting the fusion strategy did not affect the location of protein expression. VP4 cleavage between VP2 and HA was confirmed by Western blot, indicating the fusion strategy did not affect VP4 function in transfected cells. After vaccination in chickens, the DNA construct VP24-HA/pcDNA induced ELISA and virus neutralizing antibodies against VP2 and hemagglutination inhibition antibody against the HA subtype. The results indicated that a single plasmid construct carrying IBDV VP243 gene-based DNA fused with AIV HA gene can elicit specific antibody responses to both IBDV and AIV by DNA vaccination.
Collapse
|
46
|
Attaran H, Nili H, Tebianian M. Immunogenicity and protective efficacy of recombinant M2e.Hsp70c (Hsp70(359-610)) fusion protein against influenza virus infection in mice. Virol Sin 2014; 29:218-27. [PMID: 25160757 DOI: 10.1007/s12250-014-3428-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 08/05/2014] [Indexed: 11/30/2022] Open
Abstract
New strategies in vaccine development are urgently needed to combat emerging influenza viruses and to reduce the risk of pandemic disease surfacing. Being conserved, the M2e protein, is a potential candidate for universal vaccine development against influenza A viruses. Mycobacterium tuberculosis Hsp70 (mHsp70) is known to cultivate the function of immunogenic antigenpresenting cells, stimulate a strong cytotoxic T lymphocyte (CTL) response, and stop the induction of tolerance. Thus, in this study, a recombinant protein from the extracellular domain of influenza A virus matrix protein 2 (M2e), was fused to the C-terminus of Mycobacterium tuberculosis Hsp70 (Hsp70c), to generate a vaccine candidate. Humoral immune responses, IFN-γ-producing lymphocyte, and strong CTL activity were all induced to confirm the immunogenicity of M2e.Hsp70c (Hsp70(359-610)). And challenge tests showed protection against H1N1 and H9N2 strains in vaccinated groups. Finally these results demonstrates M2e.Hsp70c fusion protein can be a candidate for a universal influenza A vaccine.
Collapse
Affiliation(s)
- Hamidreza Attaran
- Avian Diseases Research Center, Faculty of Veterinary Medicine, University of Shiraz, Shiraz, 71345-1731, Iran,
| | | | | |
Collapse
|
47
|
Abstract
INTRODUCTION Tuberculosis (TB) remains a major health problem and novel vaccination regimens are urgently needed. AREAS COVERED DNA vaccines against TB have been tested in various preclinical models and strategies have been developed to increase their immunogenicity in large animal species. DNA vaccines are able to induce a wide variety of immune responses, including CD8(+) T-cell-mediated cytolytic and IFN-γ responses. DNA vaccination may be valuable in heterologous prime-boost strategies with the currently used bacillus Calmette-Guérin (BCG) vaccine. This approach could broaden the antigenic repertoire of BCG and enhance its weak induction of MHC class I-restricted immune responses. EXPERT OPINION DNA vaccines offer a number of advantages over certain other types of vaccines, such as the induction of robust MHC class I-restricted cytotoxic T lymphocyte (CTL), their generic manufacturing platform and their relatively low manufacturing costs. Because of their strong potential for inducing memory responses, DNA vaccines are particularly suited for priming immune responses. Furthermore, DNA vaccine technology may help antigen discovery by facilitating screening of candidate vaccines. Co-administration of BCG with plasmid DNA coding for immunodominant, subdominant and phase-specific antigens, poorly expressed by BCG, may lead to the development of improved TB vaccines.
Collapse
Affiliation(s)
- Nicolas Bruffaerts
- Scientific Institute of Public Health, O.D. CID-Immunology , Engelandstraat 642, Brussels, B1180 , Belgium
| | | | | |
Collapse
|
48
|
Ondondo BO. The influence of delivery vectors on HIV vaccine efficacy. Front Microbiol 2014; 5:439. [PMID: 25202303 PMCID: PMC4141443 DOI: 10.3389/fmicb.2014.00439] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 08/03/2014] [Indexed: 12/31/2022] Open
Abstract
Development of an effective HIV/AIDS vaccine remains a big challenge, largely due to the enormous HIV diversity which propels immune escape. Thus novel vaccine strategies are targeting multiple variants of conserved antibody and T cell epitopic regions which would incur a huge fitness cost to the virus in the event of mutational escape. Besides immunogen design, the delivery modality is critical for vaccine potency and efficacy, and should be carefully selected in order to not only maximize transgene expression, but to also enhance the immuno-stimulatory potential to activate innate and adaptive immune systems. To date, five HIV vaccine candidates have been evaluated for efficacy and protection from acquisition was only achieved in a small proportion of vaccinees in the RV144 study which used a canarypox vector for delivery. Conversely, in the STEP study (HVTN 502) where human adenovirus serotype 5 (Ad5) was used, strong immune responses were induced but vaccination was more associated with increased risk of HIV acquisition than protection in vaccinees with pre-existing Ad5 immunity. The possibility that pre-existing immunity to a highly promising delivery vector may alter the natural course of HIV to increase acquisition risk is quite worrisome and a huge setback for HIV vaccine development. Thus, HIV vaccine development efforts are now geared toward delivery platforms which attain superior immunogenicity while concurrently limiting potential catastrophic effects likely to arise from pre-existing immunity or vector-related immuno-modulation. However, it still remains unclear whether it is poor immunogenicity of HIV antigens or substandard immunological potency of the safer delivery vectors that has limited the success of HIV vaccines. This article discusses some of the promising delivery vectors to be harnessed for improved HIV vaccine efficacy.
Collapse
Affiliation(s)
- Beatrice O Ondondo
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford Oxford, UK
| |
Collapse
|
49
|
Weng TY, Yen MC, Huang CT, Hung JJ, Chen YL, Chen WC, Wang CY, Chang JY, Lai MD. DNA vaccine elicits an efficient antitumor response by targeting the mutant Kras in a transgenic mouse lung cancer model. Gene Ther 2014; 21:888-96. [PMID: 25077772 DOI: 10.1038/gt.2014.67] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 04/09/2014] [Accepted: 06/17/2014] [Indexed: 12/22/2022]
Abstract
Mutant Kras (V-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog) is observed in more than 20% of non-small-cell lung cancers; however, no effective Kras target therapy is available at present. The Kras DNA vaccine may represent as a novel immunotherapeutic agent in lung cancer. In this study, we investigated the antitumor efficacy of the Kras DNA vaccine in a genetically engineered inducible mouse lung tumor model driven by Kras(G12D). Lung tumors were induced by doxycycline, and the therapeutic effects of Kras DNA vaccine were evaluated with delivery of Kras(G12D) plasmids. Mutant Kras(G12D) DNA vaccine significantly decreased the tumor nodules. A dominant-negative mutant Kras(G12D)N17, devoid of oncogenic activity, achieved similar therapeutic effects. The T-helper 1 immune response was enhanced in mice treated with Kras DNA vaccine. Splenocytes from mice receiving Kras DNA vaccine presented an antigen-specific response by treatment with peptides of Kras but not Hras or OVA. The number of tumor-infiltrating CD8(+) T cells increased after Kras vaccination. In contrast, Kras DNA vaccine was not effective in the lung tumor in transgenic mice, which was induced by mutant L858R epidermal growth factor receptor. Overall, these results indicate that Kras DNA vaccine produces an effective antitumor response in transgenic mice, and may be useful in treating lung cancer-carrying Ras mutation.
Collapse
Affiliation(s)
- T-Y Weng
- 1] Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC [2] Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - M-C Yen
- 1] Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC [2] Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC [3] Center for Infectious Diseases and Signal Research, National Cheng Kung University, Tainan, Taiwan, ROC
| | - C-T Huang
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - J-J Hung
- Institute of Bioinformatics and Biosignal Transduction, College of Life Science, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Y-L Chen
- Department of Senior Citizen Services Management, Chia Nan University of Pharmacy and Science, Tainan, Taiwan, ROC
| | - W-C Chen
- 1] Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC [2] Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - C-Y Wang
- 1] Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC [2] Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - J-Y Chang
- National Institute of Cancer Research, National Health Research Institute, Tainan, Taiwan, ROC
| | - M-D Lai
- 1] Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC [2] Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC [3] Center for Infectious Diseases and Signal Research, National Cheng Kung University, Tainan, Taiwan, ROC
| |
Collapse
|
50
|
Chen Y, Wang S, Lu S. DNA Immunization for HIV Vaccine Development. Vaccines (Basel) 2014; 2:138-159. [PMID: 26344472 PMCID: PMC4494200 DOI: 10.3390/vaccines2010138] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 02/08/2014] [Accepted: 02/10/2014] [Indexed: 01/10/2023] Open
Abstract
DNA vaccination has been studied in the last 20 years for HIV vaccine research. Significant experience has been accumulated in vector design, antigen optimization, delivery approaches and the use of DNA immunization as part of a prime-boost HIV vaccination strategy. Key historical data and future outlook are presented. With better understanding on the potential of DNA immunization and recent progress in HIV vaccine research, it is anticipated that DNA immunization will play a more significant role in the future of HIV vaccine development.
Collapse
Affiliation(s)
- Yuxin Chen
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| | - Shixia Wang
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| | - Shan Lu
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|