1
|
Masimov R, Wasan EK. Chitosan non-particulate vaccine delivery systems. JOURNAL OF PHARMACY & PHARMACEUTICAL SCIENCES : A PUBLICATION OF THE CANADIAN SOCIETY FOR PHARMACEUTICAL SCIENCES, SOCIETE CANADIENNE DES SCIENCES PHARMACEUTIQUES 2024; 27:12921. [PMID: 39114808 PMCID: PMC11303186 DOI: 10.3389/jpps.2024.12921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 07/12/2024] [Indexed: 08/10/2024]
Abstract
Chitosan is an extensively used polymer for drug delivery applications in particulate and non-particulate carriers. Chitosan-based particulate, nano-, and microparticle, carriers have been the most extensively studied for the delivery of therapeutics and vaccines. However, chitosan has also been used in vaccine applications for its adjuvant properties in various hydrogels or as a carrier coating material. The focus of this review will be on the usage of chitosan as a vaccine adjuvant based on its intrinsic immunogenicity; the various forms of chitosan-based non-particulate delivery systems such as thermosensitive hydrogels, microneedles, and conjugates; and the advantages of its role as a coating material for vaccine carriers.
Collapse
Affiliation(s)
| | - Ellen K. Wasan
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
2
|
Yao Y, Zhang Z, Yang Z. The combination of vaccines and adjuvants to prevent the occurrence of high incidence of infectious diseases in bovine. Front Vet Sci 2023; 10:1243835. [PMID: 37885619 PMCID: PMC10598632 DOI: 10.3389/fvets.2023.1243835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 09/27/2023] [Indexed: 10/28/2023] Open
Abstract
As the global population grows, the demand for beef and dairy products is also increasing. The cattle industry is facing tremendous pressures and challenges. The expanding cattle industry has led to an increased risk of disease in cattle. These diseases not only cause economic losses but also pose threats to public health and safety. Hence, ensuring the health of cattle is crucial. Vaccination is one of the most economical and effective methods of preventing bovine infectious diseases. However, there are fewer comprehensive reviews of bovine vaccines available. In addition, the variable nature of bovine infectious diseases will result in weakened or even ineffective immune protection from existing vaccines. This shows that it is crucial to improve overall awareness of bovine vaccines. Adjuvants, which are crucial constituents of vaccines, have a significant role in enhancing vaccine response. This review aims to present the latest advances in bovine vaccines mainly including types of bovine vaccines, current status of development of commonly used vaccines, and vaccine adjuvants. In addition, this review highlights the main challenges and outstanding problems of bovine vaccines and adjuvants in the field of research and applications. This review provides a theoretical and practical basis for the eradication of global bovine infectious diseases.
Collapse
Affiliation(s)
- Yiyang Yao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Zhipeng Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Zhangping Yang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education, Yangzhou University, Yangzhou, China
| |
Collapse
|
3
|
Hanaki A, Ogawa K, Tagami T, Ozeki T. Fabrication and Characterization of Antibody-Loaded Cationic Porous PLGA Microparticles for Sustained Antibody Release. AAPS J 2023; 25:92. [PMID: 37740072 DOI: 10.1208/s12248-023-00859-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 08/30/2023] [Indexed: 09/24/2023] Open
Abstract
Poly lactic-co-glycolic acid (PLGA) microparticles have been formulated to allow the sustained release of numerous drugs, including antibodies. It is well-known that antibodies are susceptible to chemical and physical stress; therefore, it is necessary to be loaded on PLGA microparticles under mild conditions. In the present study, we constructed cationic porous PLGA microparticles that could be electrostatically adsorbed with infliximab as a model antibody. Cationic porous PLGA microparticles were prepared using the double emulsion method by adding polyethyleneimine and ammonium bicarbonate. After antibody loading, surface pores closure was achieved by mild heating. The size of the optimized formulation was approximately 5 μm, exhibiting a positive charge. The loaded antibody was gradually released from the formulation over 56 days. Based on a tumor necrosis factor (TNF)-α inhibition assay, the released infliximab maintained its pharmacological activity. Collectively, we successfully loaded antibodies into PLGA microparticles while maintaining activity and demonstrating long-acting properties.
Collapse
Affiliation(s)
- Ayaka Hanaki
- Drug Delivery and Nano Pharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi, 467-8603, Japan
| | - Koki Ogawa
- Drug Delivery and Nano Pharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi, 467-8603, Japan
| | - Tatsuaki Tagami
- Drug Delivery and Nano Pharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi, 467-8603, Japan
| | - Tetsuya Ozeki
- Drug Delivery and Nano Pharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi, 467-8603, Japan.
| |
Collapse
|
4
|
Khalid K, Poh CL. The development of DNA vaccines against SARS-CoV-2. Adv Med Sci 2023; 68:213-226. [PMID: 37364379 PMCID: PMC10290423 DOI: 10.1016/j.advms.2023.05.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 04/07/2023] [Accepted: 05/31/2023] [Indexed: 06/28/2023]
Abstract
BACKGROUND The COVID-19 pandemic exerted significant impacts on public health and global economy. Research efforts to develop vaccines at warp speed against SARS-CoV-2 led to novel mRNA, viral vectored, and inactivated vaccines being administered. The current COVID-19 vaccines incorporate the full S protein of the SARS-CoV-2 Wuhan strain but rapidly emerging variants of concern (VOCs) have led to significant reductions in protective efficacies. There is an urgent need to develop next-generation vaccines which could effectively prevent COVID-19. METHODS PubMed and Google Scholar were systematically reviewed for peer-reviewed papers up to January 2023. RESULTS A promising solution to the problem of emerging variants is a DNA vaccine platform since it can be easily modified. Besides expressing whole protein antigens, DNA vaccines can also be constructed to include specific nucleotide genes encoding highly conserved and immunogenic epitopes from the S protein as well as from other structural/non-structural proteins to develop effective vaccines against VOCs. DNA vaccines are associated with low transfection efficiencies which could be enhanced by chemical, genetic, and molecular adjuvants as well as delivery systems. CONCLUSIONS The DNA vaccine platform offers a promising solution to the design of effective vaccines. The challenge of limited immunogenicity in humans might be solved through the use of genetic modifications such as the addition of nuclear localization signal (NLS) peptide gene, strong promoters, MARs, introns, TLR agonists, CD40L, and the development of appropriate delivery systems utilizing nanoparticles to increase uptake by APCs in enhancing the induction of potent immune responses.
Collapse
Affiliation(s)
- Kanwal Khalid
- Centre for Virus and Vaccine Research, School of Medical and Life Sciences, Sunway University, Bandar Sunway, Malaysia
| | - Chit Laa Poh
- Centre for Virus and Vaccine Research, School of Medical and Life Sciences, Sunway University, Bandar Sunway, Malaysia.
| |
Collapse
|
5
|
Xu H, Zhu S, Govinden R, Chenia HY. Multiple Vaccines and Strategies for Pandemic Preparedness of Avian Influenza Virus. Viruses 2023; 15:1694. [PMID: 37632036 PMCID: PMC10459121 DOI: 10.3390/v15081694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/14/2023] [Accepted: 07/27/2023] [Indexed: 08/27/2023] Open
Abstract
Avian influenza viruses (AIV) are a continuous cause of concern due to their pandemic potential and devasting effects on poultry, birds, and human health. The low pathogenic avian influenza virus has the potential to evolve into a highly pathogenic avian influenza virus, resulting in its rapid spread and significant outbreaks in poultry. Over the years, a wide array of traditional and novel strategies has been implemented to prevent the transmission of AIV in poultry. Mass vaccination is still an economical and effective approach to establish immune protection against clinical virus infection. At present, some AIV vaccines have been licensed for large-scale production and use in the poultry industry; however, other new types of AIV vaccines are currently under research and development. In this review, we assess the recent progress surrounding the various types of AIV vaccines, which are based on the classical and next-generation platforms. Additionally, the delivery systems for nucleic acid vaccines are discussed, since these vaccines have attracted significant attention following their significant role in the fight against COVID-19. We also provide a general introduction to the dendritic targeting strategy, which can be used to enhance the immune efficiency of AIV vaccines. This review may be beneficial for the avian influenza research community, providing ideas for the design and development of new AIV vaccines.
Collapse
Affiliation(s)
- Hai Xu
- Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China;
- Discipline of Microbiology, School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal, Durban 4001, South Africa;
| | - Shanyuan Zhu
- Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China;
| | - Roshini Govinden
- Discipline of Microbiology, School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal, Durban 4001, South Africa;
| | - Hafizah Y. Chenia
- Discipline of Microbiology, School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal, Durban 4001, South Africa;
| |
Collapse
|
6
|
Bohmer M, Xue Y, Jankovic K, Dong Y. Advances in engineering and delivery strategies for cytokine immunotherapy. Expert Opin Drug Deliv 2023; 20:579-595. [PMID: 37104673 PMCID: PMC10330431 DOI: 10.1080/17425247.2023.2208344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 04/25/2023] [Indexed: 04/29/2023]
Abstract
INTRODUCTION Cytokine immunotherapy is a growing field for the treatment of cancer, infectious disease, autoimmunity, and other ailments. Therapeutic cytokines are a class of secreted, small proteins that play a pivotal role in regulating the innate and adaptive immune system by provoking or mitigating immune responses. In the clinic, cytokines are frequently combined with other treatments, such as small molecules and monoclonal antibodies. However, the clinical translation of cytokine therapies is hindered by their short half-life, pleiotropic nature, and off-target effects, which cause diminished efficacy and severe systemic toxicity. Such toxicity limits dosage, thus resulting in suboptimal doses. Accordingly, numerous efforts have been devoted to exploring strategies to promote cytokine therapies by improving their tissue specificity and pharmacokinetics. AREAS COVERED Preclinical and clinical research into bioengineering and delivery strategies for cytokines, consisting of bioconjugation, fusion proteins, nanoparticles, and scaffold-based systems. EXPERT OPINION These approaches pave the way for the development of next-generation cytokine treatments with greater clinical benefit and reduced toxicity, circumventing such issues currently associated with cytokine therapy.
Collapse
Affiliation(s)
- Margaret Bohmer
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Yonger Xue
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Katarina Jankovic
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Yizhou Dong
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
- Pelotonia Institute for Immune-Oncology, The Ohio State University, Columbus, OH, 43210, USA
- Center for Cancer Engineering, The Ohio State University, Columbus, OH, 43210, USA
- Center for Cancer Metabolism, Department of Radiation Oncology, The Ohio State University, Columbus, OH, 43210, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210, USA
- The Center for Clinical and Translational Science, The Ohio State University, Columbus, OH, 43210, USA
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
- Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University, Columbus, OH, 43210, USA
- Icahn Genomics Institute, Precision Immunology Institute, Department of Oncological Sciences, Tisch Cancer Institute, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| |
Collapse
|
7
|
Wilson B, Mukundan Geetha K. Nanomedicine to deliver biological macromolecules for treating COVID-19. Vaccine 2022; 40:3931-3941. [PMID: 35660038 PMCID: PMC9149150 DOI: 10.1016/j.vaccine.2022.05.068] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 03/21/2022] [Accepted: 05/19/2022] [Indexed: 12/15/2022]
Abstract
Coronavirus disease (COVID-19) was first reported in December 2019, China and later it was found that the causative microorganism is severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2). As on 3rd June 2021, SARS-CoV-2 has affected 171049741 people worldwide with 3549710 deaths. Nanomedicine such as nanoparticles, liposomes, lipid nanoparticles, virus-like nanoparticles offer tremendous hopes to treat viral infections including COVID-19. Most importantly target specific ligands can be attached on the surface of them and this makes them more target specific and the loaded drug can be delivered to cellular and molecular level. These properties of nanomedicines can be utilized to deliver drugs or vaccines to treat viral diseases including SARS-CoV-2 infection. This review discusses about SARS-CoV-2 and the potential application of nanomedicines for delivering biological macromolecules like vaccines and drugs for treating COVID-19.
Collapse
Affiliation(s)
- Barnabas Wilson
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Dayananda Sagar University, Kumaraswamy Layout, Bangalore, Karnataka 560078, India.
| | - Kannoth Mukundan Geetha
- Department of Pharmacology, College of Pharmaceutical Sciences, Dayananda Sagar University, Kumaraswamy Layout, Bangalore, Karnataka 560078, India
| |
Collapse
|
8
|
Souri M, Chiani M, Farhangi A, Mehrabi MR, Nourouzian D, Raahemifar K, Soltani M. Anti-COVID-19 Nanomaterials: Directions to Improve Prevention, Diagnosis, and Treatment. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:783. [PMID: 35269270 PMCID: PMC8912597 DOI: 10.3390/nano12050783] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/18/2022] [Accepted: 02/23/2022] [Indexed: 02/04/2023]
Abstract
Following the announcement of the outbreak of COVID-19 by the World Health Organization, unprecedented efforts were made by researchers around the world to combat the disease. So far, various methods have been developed to combat this "virus" nano enemy, in close collaboration with the clinical and scientific communities. Nanotechnology based on modifiable engineering materials and useful physicochemical properties has demonstrated several methods in the fight against SARS-CoV-2. Here, based on what has been clarified so far from the life cycle of SARS-CoV-2, through an interdisciplinary perspective based on computational science, engineering, pharmacology, medicine, biology, and virology, the role of nano-tools in the trio of prevention, diagnosis, and treatment is highlighted. The special properties of different nanomaterials have led to their widespread use in the development of personal protective equipment, anti-viral nano-coats, and disinfectants in the fight against SARS-CoV-2 out-body. The development of nano-based vaccines acts as a strong shield in-body. In addition, fast detection with high efficiency of SARS-CoV-2 by nanomaterial-based point-of-care devices is another nanotechnology capability. Finally, nanotechnology can play an effective role as an agents carrier, such as agents for blocking angiotensin-converting enzyme 2 (ACE2) receptors, gene editing agents, and therapeutic agents. As a general conclusion, it can be said that nanoparticles can be widely used in disinfection applications outside in vivo. However, in in vivo applications, although it has provided promising results, it still needs to be evaluated for possible unintended immunotoxicity. Reviews like these can be important documents for future unwanted pandemics.
Collapse
Affiliation(s)
- Mohammad Souri
- Department of NanoBiotechnology, Pasteur Institute of Iran, Tehran 13169-43551, Iran; (M.S.); (M.C.); (A.F.)
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran 19967-15433, Iran
| | - Mohsen Chiani
- Department of NanoBiotechnology, Pasteur Institute of Iran, Tehran 13169-43551, Iran; (M.S.); (M.C.); (A.F.)
| | - Ali Farhangi
- Department of NanoBiotechnology, Pasteur Institute of Iran, Tehran 13169-43551, Iran; (M.S.); (M.C.); (A.F.)
| | - Mohammad Reza Mehrabi
- Department of NanoBiotechnology, Pasteur Institute of Iran, Tehran 13169-43551, Iran; (M.S.); (M.C.); (A.F.)
| | - Dariush Nourouzian
- Department of NanoBiotechnology, Pasteur Institute of Iran, Tehran 13169-43551, Iran; (M.S.); (M.C.); (A.F.)
| | - Kaamran Raahemifar
- Data Science and Artificial Intelligence Program, College of Information Sciences and Technology (IST), Penn State University, State College, PA 16801, USA;
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L 3G1, Canada
- School of Optometry and Vision Science, Faculty of Science, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L 3G1, Canada
| | - M. Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran 19967-15433, Iran
- Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada
- Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, ON N2L 3G1, Canada
- Advanced Bioengineering Initiative Center, Multidisciplinary International Complex, K. N. Toosi University of Technology, Tehran 14176-14411, Iran
| |
Collapse
|
9
|
Tang J, Cai L, Xu C, Sun S, Liu Y, Rosenecker J, Guan S. Nanotechnologies in Delivery of DNA and mRNA Vaccines to the Nasal and Pulmonary Mucosa. NANOMATERIALS 2022; 12:nano12020226. [PMID: 35055244 PMCID: PMC8777913 DOI: 10.3390/nano12020226] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 01/03/2022] [Accepted: 01/05/2022] [Indexed: 02/07/2023]
Abstract
Recent advancements in the field of in vitro transcribed mRNA (IVT-mRNA) vaccination have attracted considerable attention to such vaccination as a cutting-edge technique against infectious diseases including COVID-19 caused by SARS-CoV-2. While numerous pathogens infect the host through the respiratory mucosa, conventional parenterally administered vaccines are unable to induce protective immunity at mucosal surfaces. Mucosal immunization enables the induction of both mucosal and systemic immunity, efficiently removing pathogens from the mucosa before an infection occurs. Although respiratory mucosal vaccination is highly appealing, successful nasal or pulmonary delivery of nucleic acid-based vaccines is challenging because of several physical and biological barriers at the airway mucosal site, such as a variety of protective enzymes and mucociliary clearance, which remove exogenously inhaled substances. Hence, advanced nanotechnologies enabling delivery of DNA and IVT-mRNA to the nasal and pulmonary mucosa are urgently needed. Ideal nanocarriers for nucleic acid vaccines should be able to efficiently load and protect genetic payloads, overcome physical and biological barriers at the airway mucosal site, facilitate transfection in targeted epithelial or antigen-presenting cells, and incorporate adjuvants. In this review, we discuss recent developments in nucleic acid delivery systems that target airway mucosa for vaccination purposes.
Collapse
Affiliation(s)
- Jie Tang
- Department of Pediatrics, Ludwig-Maximilians University of Munich, 80337 Munich, Germany;
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane 4072, Australia;
| | - Larry Cai
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane 4072, Australia;
| | - Chuanfei Xu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, Third Military Medical University, Chongqing 400038, China; (C.X.); (S.S.); (Y.L.)
| | - Si Sun
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, Third Military Medical University, Chongqing 400038, China; (C.X.); (S.S.); (Y.L.)
| | - Yuheng Liu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, Third Military Medical University, Chongqing 400038, China; (C.X.); (S.S.); (Y.L.)
| | - Joseph Rosenecker
- Department of Pediatrics, Ludwig-Maximilians University of Munich, 80337 Munich, Germany;
- Correspondence: (J.R.); (S.G.); Tel.: +49-89-440057713 (J.R.); +86-23-68771645 (S.G.)
| | - Shan Guan
- Department of Pediatrics, Ludwig-Maximilians University of Munich, 80337 Munich, Germany;
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, Third Military Medical University, Chongqing 400038, China; (C.X.); (S.S.); (Y.L.)
- Correspondence: (J.R.); (S.G.); Tel.: +49-89-440057713 (J.R.); +86-23-68771645 (S.G.)
| |
Collapse
|
10
|
Kenubih A. Foot and Mouth Disease Vaccine Development and Challenges in Inducing Long-Lasting Immunity: Trends and Current Perspectives. VETERINARY MEDICINE-RESEARCH AND REPORTS 2021; 12:205-215. [PMID: 34513635 PMCID: PMC8420785 DOI: 10.2147/vmrr.s319761] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 07/29/2021] [Indexed: 11/26/2022]
Abstract
Foot and mouth disease (FMD) is an extremely contagious viral disease of livestock caused by foot and mouse disease virus genus: Aphthovirus, which causes a serious economic impact on both individual farmers and the national economy. Many attempts to advance a vaccine for FMD have failed to induce sterile immunity. The classical methods of vaccine production were due to selective accumulation of mutations around antigenic and binding sites. Reversion of the agent by positive selection and quasi-species swarm, use of this method is inapplicable for use in non-endemic areas. Chemical attenuation using binary ethyleneimine (BEI) protected the capsid integrity and produced a pronounced immunity against the challenge strain. Viral antigens which have been chemically synthesized or expressed in viruses, plasmid, or plants were tried in the vaccination of animals. DNA vaccines expressing either structural or nonstructural protein antigens have been tried to immunize animals. Using interleukins as a genetic adjuvant for DNA vaccines have a promising effect. While the challenges of inducing sterile immunity lies on non-structural (NS) proteins of FMDV which are responsible for apoptosis of dendritic cells and have negative effects on lympho-proliferative responses which lead to transient immunosuppression. Furthermore, destruction of host protein trafficking by nonstructural proteins suppressed CD8+ T-cell proliferation. In this review, it tried to address multiple approaches for vaccine development trials and bottle necks of producing sterile immunity.
Collapse
Affiliation(s)
- Ambaye Kenubih
- University of Gondar, College of Veterinary Medicine and Animal Sciences, Para-Clinical Studies, Gondar, Ethiopia
| |
Collapse
|
11
|
Franck CO, Fanslau L, Bistrovic Popov A, Tyagi P, Fruk L. Biopolymer-based Carriers for DNA Vaccine Design. Angew Chem Int Ed Engl 2021; 60:13225-13243. [PMID: 32893932 PMCID: PMC8247987 DOI: 10.1002/anie.202010282] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Indexed: 12/16/2022]
Abstract
Over the last 30 years, genetically engineered DNA has been tested as novel vaccination strategy against various diseases, including human immunodeficiency virus (HIV), hepatitis B, several parasites, and cancers. However, the clinical breakthrough of the technique is confined by the low transfection efficacy and immunogenicity of the employed vaccines. Therefore, carrier materials were designed to prevent the rapid degradation and systemic clearance of DNA in the body. In this context, biopolymers are a particularly promising DNA vaccine carrier platform due to their beneficial biochemical and physical characteristics, including biocompatibility, stability, and low toxicity. This article reviews the applications, fabrication, and modification of biopolymers as carrier medium for genetic vaccines.
Collapse
Affiliation(s)
- Christoph O. Franck
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgePhillipa Fawcett DriveCambridgeCB3 0ASUK
| | - Luise Fanslau
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgePhillipa Fawcett DriveCambridgeCB3 0ASUK
| | - Andrea Bistrovic Popov
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgePhillipa Fawcett DriveCambridgeCB3 0ASUK
| | - Puneet Tyagi
- Dosage Form Design and DevelopmentBioPharmaceuticals DevelopmentR&DAstra ZenecaGaithersburgMD20878USA
| | - Ljiljana Fruk
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgePhillipa Fawcett DriveCambridgeCB3 0ASUK
| |
Collapse
|
12
|
Franck CO, Fanslau L, Bistrovic Popov A, Tyagi P, Fruk L. Biopolymer‐based Carriers for DNA Vaccine Design. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202010282] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Christoph O. Franck
- Department of Chemical Engineering and Biotechnology University of Cambridge Phillipa Fawcett Drive Cambridge CB3 0AS UK
| | - Luise Fanslau
- Department of Chemical Engineering and Biotechnology University of Cambridge Phillipa Fawcett Drive Cambridge CB3 0AS UK
| | - Andrea Bistrovic Popov
- Department of Chemical Engineering and Biotechnology University of Cambridge Phillipa Fawcett Drive Cambridge CB3 0AS UK
| | - Puneet Tyagi
- Dosage Form Design and Development BioPharmaceuticals Development R&D Astra Zeneca Gaithersburg MD 20878 USA
| | - Ljiljana Fruk
- Department of Chemical Engineering and Biotechnology University of Cambridge Phillipa Fawcett Drive Cambridge CB3 0AS UK
| |
Collapse
|
13
|
Sanchez J, Gonçalves E, Llano A, Gonzáles P, Fernández-Maldonado M, Vogt A, Soria A, Perez S, Cedeño S, Fernández MA, Nourikyan J, de Bernard S, Ganoza C, Pedruzzi E, Bonduelle O, Mothe B, Gòmez CE, Esteban M, Garcia F, Lama JR, Brander C, Combadiere B. Immune Profiles Identification by Vaccinomics After MVA Immunization in Randomized Clinical Study. Front Immunol 2020; 11:586124. [PMID: 33244316 PMCID: PMC7683801 DOI: 10.3389/fimmu.2020.586124] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 10/12/2020] [Indexed: 12/04/2022] Open
Abstract
Background Our previous work has demonstrated the benefits of transcutaneous immunization in targeting Langerhans cells and preferentially inducing CD8 T-cell responses. Methods In this randomized phase Ib clinical trial including 20 HIV uninfected volunteers, we compared the safety and immunogenicity of the MVA recombinant vaccine expressing HIV-B antigen (MVA-B) by transcutaneous and intramuscular routes. We hypothesized that the quality of innate and adaptive immunity differs according to the route of immunization and explored the quality of the vector vaccine-induced immune responses. We also investigated the early blood transcriptome and serum cytokine levels to identify innate events correlated with the strength and quality of adaptive immunity. Results We demonstrate that MVA-B vaccine is safe by both routes, but that the quality and intensity of both innate and adaptive immunity differ significantly. Transcutaneous vaccination promoted CD8 responses in the absence of antibodies and slightly affected gene expression, involving mainly genes associated with metabolic pathways. Intramuscular vaccination, on the other hand, drove robust changes in the expression of genes involved in IL-6 and interferon signalling pathways, mainly those associated with humoral responses, and also some levels of CD8 response. Conclusion Thus, vaccine delivery route perturbs early innate responses that shape the quality of adaptive immunity. Clinical Trial Registration http://ClinicalTrials.gov, identifier PER-073-13.
Collapse
Affiliation(s)
- Jorge Sanchez
- Centro de Investigaciones Tecnológicas, Biomedicas y Medioambientales, Universidad Nacional Mayor de San Marcos, Lima, Peru
| | - Elena Gonçalves
- Sorbonne Université, Inserm, Centre d'Immunologie et des Maladies Infectieuses (CIMIParis), Paris, France
| | - Anuska Llano
- IrsiCaixa AIDS Research Institute-HIVACAT, Hospital Universitari Germans Trias i Pujol, Barcelona, Spain
| | | | | | - Annika Vogt
- Clinical Research Center for Hair and Skin Science, Department of Dermatology, Venerology and Allergy, Charité-Universitatsmedizin Berlin, corporate member of Freie Universitaet Berlin, Humboldt-Universitaet zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | | | - Susana Perez
- Centro de Investigaciones Tecnológicas, Biomedicas y Medioambientales, Universidad Nacional Mayor de San Marcos, Lima, Peru
| | - Samandhy Cedeño
- IrsiCaixa AIDS Research Institute-HIVACAT, Hospital Universitari Germans Trias i Pujol, Barcelona, Spain
| | - Marco Antonio Fernández
- Flow Cytometry Facility, Germans Trias i Pujol Research Institute (IGTP), Hospital Universitari Germans Trias i Pujol, Barcelona, Spain
| | | | | | | | - Eric Pedruzzi
- Sorbonne Université, Inserm, Centre d'Immunologie et des Maladies Infectieuses (CIMIParis), Paris, France
| | - Olivia Bonduelle
- Sorbonne Université, Inserm, Centre d'Immunologie et des Maladies Infectieuses (CIMIParis), Paris, France
| | - Beatriz Mothe
- IrsiCaixa AIDS Research Institute-HIVACAT, Hospital Universitari Germans Trias i Pujol, Barcelona, Spain.,Fundació Lluita contra la Sida, Infectious Diseases Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Carmen E Gòmez
- Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Mariano Esteban
- Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Felipe Garcia
- Infectious Diseases Department, Hospital Clínic, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Javier R Lama
- Asociacion Civil Impacta Salud y Educacion, Lima, Peru
| | - Christian Brander
- IrsiCaixa AIDS Research Institute-HIVACAT, Hospital Universitari Germans Trias i Pujol, Barcelona, Spain.,Faculty of Medicine, Universitat de Vic-Central de Catalunya (UVic-UCC), Vic, Spain.,Institució Catalana de Recerca I Estudis Avançats (ICREA), Barcelona, Spain
| | - Behazine Combadiere
- Sorbonne Université, Inserm, Centre d'Immunologie et des Maladies Infectieuses (CIMIParis), Paris, France
| |
Collapse
|
14
|
Chauhan G, Madou MJ, Kalra S, Chopra V, Ghosh D, Martinez-Chapa SO. Nanotechnology for COVID-19: Therapeutics and Vaccine Research. ACS NANO 2020; 14:7760-7782. [PMID: 32571007 PMCID: PMC7325519 DOI: 10.1021/acsnano.0c04006] [Citation(s) in RCA: 207] [Impact Index Per Article: 51.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 06/22/2020] [Indexed: 05/04/2023]
Abstract
The current global health threat by the novel coronavirus disease 2019 (COVID-19) requires an urgent deployment of advanced therapeutic options available. The role of nanotechnology is highly relevant to counter this "virus" nano enemy. Nano intervention is discussed in terms of designing effective nanocarriers to counter the conventional limitations of antiviral and biological therapeutics. This strategy directs the safe and effective delivery of available therapeutic options using engineered nanocarriers, blocking the initial interactions of viral spike glycoprotein with host cell surface receptors, and disruption of virion construction. Controlling and eliminating the spread and reoccurrence of this pandemic demands a safe and effective vaccine strategy. Nanocarriers have potential to design risk-free and effective immunization strategies for severe acute respiratory syndrome coronavirus 2 vaccine candidates such as protein constructs and nucleic acids. We discuss recent as well as ongoing nanotechnology-based therapeutic and prophylactic strategies to fight against this pandemic, outlining the key areas for nanoscientists to step in.
Collapse
Affiliation(s)
- Gaurav Chauhan
- School of Engineering and Sciences,
Tecnologico de Monterrey, Av. Eugenio
Garza Sada 2501 Sur, 64849 Monterrey, Nuevo León,
Mexico
| | - Marc J. Madou
- School of Engineering and Sciences,
Tecnologico de Monterrey, Av. Eugenio
Garza Sada 2501 Sur, 64849 Monterrey, Nuevo León,
Mexico
- Department of Mechanical and Aerospace
Engineering, University of California
Irvine, Engineering Gateway 4200, Irvine,
California 92697, United States
| | - Sourav Kalra
- Department of Pharmaceutical Technology
(Process Chemistry), National Institute of Pharmaceutical
Education and Research, Sector 67, S.A.S. Nagar,
Punjab 160062, India
| | - Vianni Chopra
- Institute of Nano Science
and Technology, Habitat Centre, Phase 10 Mohali,
160062 Punjab, India
| | - Deepa Ghosh
- Institute of Nano Science
and Technology, Habitat Centre, Phase 10 Mohali,
160062 Punjab, India
| | - Sergio O. Martinez-Chapa
- School of Engineering and Sciences,
Tecnologico de Monterrey, Av. Eugenio
Garza Sada 2501 Sur, 64849 Monterrey, Nuevo León,
Mexico
| |
Collapse
|
15
|
Liu X, Qi L, Lv J, Zhang Z, Zhou P, Ma Z, Wang Y, Zhang Y, Pan L. The immune response to a recombinant Lactococcus lactis oral vaccine against foot-and-mouth disease virus in mice. Biotechnol Lett 2020; 42:1907-1917. [PMID: 32385744 PMCID: PMC7210100 DOI: 10.1007/s10529-020-02900-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 04/24/2020] [Indexed: 11/11/2022]
Abstract
Objective Development of an effective mucosal vaccine to induce specific immune responses against Foot-and-mouth disease virus (FMDV). Results For this purpose, the FMDV VP1 gene (SPVP1) was optimized and synthesized based on the codon bias of Lactococcus lactis (L. lactis), and then incorporated in the plasmid pNZ8148. L. lactis NZ9000 containing the pNZ8148-SPVP1 recombinant plasmid was used as an oral delivery vehicle to induce anti-FMDV mucosal and systemic immune responses in mice. After confirmation that the SPVP1 protein was expressed successfully in the recombinant L. latic, the mice were orally challenged with NZ9000-pNZ8148, NZ9000-pNZ8148-SPVP1, phosphate-buffered saline as a mock infection group, or with inactivated vaccine as a positive group. Mice immunized with NZ9000-pNZ8148-SPVP1 produced high levels of mucosal secretory IgA (sIgA), antigen-specific serum IgG, IgA, and neutralizing antibodies, and developed stronger cell-mediated immune reactions and significant T spleen lymphocyte proliferation. Furthermore, the recombinant group generated much higher levels of IFN-γ, IL-2, IL-4, IL-5, and IL-10 than the other groups. Conclusions Potent immune responses were successfully elicited in mice with FMDV VP1 delivered through L. lactis.
Collapse
Affiliation(s)
- Xinsheng Liu
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Linlin Qi
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Jianliang Lv
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Zhongwang Zhang
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China.
| | - Peng Zhou
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Zhongyuan Ma
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Yonglu Wang
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Yongguang Zhang
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, China
| | - Li Pan
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China. .,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, China.
| |
Collapse
|
16
|
Zheng HH, Wang LQ, Fu PF, Zheng LL, Chen HY, Liu F. Characterization of a recombinant pseudorabies virus expressing porcine parvovirus VP2 protein and porcine IL-6. Virol J 2020; 17:19. [PMID: 32014014 PMCID: PMC6998180 DOI: 10.1186/s12985-020-1292-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 01/23/2020] [Indexed: 12/18/2022] Open
Abstract
Background Porcine parvovirus (PPV) and pseudorabies virus (PRV) are the important etiological agents of swine infectious diseases, resulting in huge economic losses to the Chinese swine industry. Interleukin-6 (IL-6) has the roles to support host immune response to infections as a pleiotropic cytokine. It is essential to construct a live attenuated vaccine-based recombinant PRV that expresses PPV VP2 protein and porcine IL-6 for prevention and control of PRV and PPV. Methods The recombinant plasmid, pGVP2-IL6, was constructed by porcine IL-6 gene substituting for EGFP gene of the PRV transfer plasmid pGVP2-EGFP containing VP2 gene of PPV. Plasmid pGVP2-IL6 was transfected into swine testicle cells pre-infected with the virus rPRV-VP2-EGFP strain through homologous recombination and plaque purification to generate a recombinant virus rPRV-VP2-IL6. The recombinant PRV was further identified by PCR and DNA sequencing, and the expression of the VP2 protein and porcine IL-6 was analyzed by reverse transcription-PCR (RT-PCR) and Western blot. The virus titer was calculated according to Reed and Muench method. The immunogenicity of the recombinant virus was preliminarily evaluated in mice by intramuscular administration twice with the rPRV-VP2-IL6 at 4-week intervals. Results A recombinant virus rPRV-VP2-IL6 was successfully constructed and confirmed in this study. The properties of rPRV-VP2-IL6 were similar to the parental virus HB98 in terms of growth curve, morphogenesis and virus plaque sizes, and rPRV-VP2-IL6 was proliferated in different cell types. It induced specific antibodies against PPV as well as a strong increase of PPV-specific lymphocyte proliferation responses in mice immunized with rPRV-VP2-IL6, and provided partial protection against the virulent PPV challenge. rPRV-VP2-IL6 also induced a high level of neutralizing antibodies against PRV, and significantly reduced the mortality rate of (1 of 10) following virulent PRV challenge compared with the control (10 of 10). Conclusions The recombinant rPRV-VP2-IL6 might be a potential candidate vaccine against PRV and PPV infections in pigs.
Collapse
Affiliation(s)
- Hui-Hua Zheng
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengdong New District Longzi Lake#15, 450046, Zhengzhou, Henan Province, People's Republic of China
| | - Lin-Qing Wang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengdong New District Longzi Lake#15, 450046, Zhengzhou, Henan Province, People's Republic of China.,Department of Life Science, Zhengzhou Normal University, Zhengzhou, 450044, Henan Province, People's Republic of China
| | - Peng-Fei Fu
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengdong New District Longzi Lake#15, 450046, Zhengzhou, Henan Province, People's Republic of China
| | - Lan-Lan Zheng
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengdong New District Longzi Lake#15, 450046, Zhengzhou, Henan Province, People's Republic of China
| | - Hong-Ying Chen
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengdong New District Longzi Lake#15, 450046, Zhengzhou, Henan Province, People's Republic of China.
| | - Fang Liu
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengdong New District Longzi Lake#15, 450046, Zhengzhou, Henan Province, People's Republic of China.
| |
Collapse
|
17
|
Lim M, Badruddoza AZM, Firdous J, Azad M, Mannan A, Al-Hilal TA, Cho CS, Islam MA. Engineered Nanodelivery Systems to Improve DNA Vaccine Technologies. Pharmaceutics 2020; 12:E30. [PMID: 31906277 PMCID: PMC7022884 DOI: 10.3390/pharmaceutics12010030] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/16/2019] [Accepted: 12/21/2019] [Indexed: 12/18/2022] Open
Abstract
DNA vaccines offer a flexible and versatile platform to treat innumerable diseases due to the ease of manipulating vaccine targets simply by altering the gene sequences encoded in the plasmid DNA delivered. The DNA vaccines elicit potent humoral and cell-mediated responses and provide a promising method for treating rapidly mutating and evasive diseases such as cancer and human immunodeficiency viruses. Although this vaccine technology has been available for decades, there is no DNA vaccine that has been used in bed-side application to date. The main challenge that hinders the progress of DNA vaccines and limits their clinical application is the delivery hurdles to targeted immune cells, which obstructs the stimulation of robust antigen-specific immune responses in humans. In this updated review, we discuss various nanodelivery systems that improve DNA vaccine technologies to enhance the immunological response against target diseases. We also provide possible perspectives on how we can bring this exciting vaccine technology to bedside applications.
Collapse
Affiliation(s)
- Michael Lim
- Nanotechnology Engineering Program, University of Waterloo, Waterloo, ON N2L 3G1, Canada;
| | - Abu Zayed Md Badruddoza
- Department of Chemical and Life Sciences Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA;
| | - Jannatul Firdous
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
| | - Mohammad Azad
- Department of Chemical, Biological and Bioengineering, North Carolina A&T State University, Greensboro, NC 27411, USA;
| | - Adnan Mannan
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chittagong 4331, Bangladesh;
| | - Taslim Ahmed Al-Hilal
- Department of Pharmaceutical Sciences, University of Texas El Paso, El Paso, TX 79968, USA;
| | - Chong-Su Cho
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Technology, Seoul National University, Gwanak-gu, Seoul 08826, Korea
| | | |
Collapse
|
18
|
Abstract
Infectious diseases are the tip of the iceberg in the economic burden of the developing countries, due to the resistance of the pathogens to antibiotics and the lack of vaccines. The vaccines have become a big challenge in the last decades, where the attention has been focused on scientific challenges such as new vaccine development and adjuvants or delivery systems. The classical vaccines were developed from live-attenuated or killed organisms, such as influenza, smallpox, and BCG, as well as subunits such as Hepatitis B. The attenuated vaccines carry the risk of regaining their pathogenicity under immunosuppression conditions. The development of subunit vaccines without risk are considered as an essential need in combination with adequate delivery systems to obtain desired cell and humoral immune responses against infectious diseases. In the last decades, the use of nanoparticles as a delivery system in vaccines has received special attention to improve vaccine efficacy. These nanoparticles could be composed of lipids, metal and nonmetal inorganics, several polymers, and virus-like particles, which have been tested in research; some of them have already been approved for human and animal use. The characteristics of the nanoparticles have allowed targeting desired antigen-presenting cells to improve immunization strategies to induce protection. The main characteristics of the nanoparticles are to protect the antigens from early proteolytic degradation, control antigen release, and help antigen uptake and processing by antigen-presenting cells, and they should be safe for human and veterinary use. In addition, the nanoparticles could be modified in their physicochemical properties to target specific cells and improve vaccine efficacy. This chapter focuses on the nanoparticle-based vaccine formulations and the approaches used to realize efficient delivery of vaccines in order to induce host protective immunity against infectious diseases.
Collapse
Affiliation(s)
- Diana Diaz-Arévalo
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia-FIDIC, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, DC, Colombia
| | - Mingtao Zeng
- Center of Emphasis in Infectious Diseases, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, United States
| |
Collapse
|
19
|
Immunogenicity and protective efficacy of recombinant proteins consisting of multiple epitopes of foot-and-mouth disease virus fused with flagellin. Appl Microbiol Biotechnol 2019; 103:3367-3379. [PMID: 30888465 DOI: 10.1007/s00253-019-09691-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 02/07/2019] [Accepted: 02/08/2019] [Indexed: 10/27/2022]
Abstract
Many recent studies have shown that flagellin fused to heterologous antigens can induce significantly enhanced humoral and cellular immune responses through its adjuvant activity. Therefore, in this study, two key B cell epitopes and a truncated VP1 (ΔVP1) protein from foot-and-mouth disease virus (FMDV) were expressed as flagellin fusion proteins in different patterns. Specifically, ΔVP1 and two duplicates of two key B cell epitopes (2×B1B2) were fused separately to the C-terminus of flagellin with a universal exogenous T cell epitope to construct FT (Flagellin-Truncated VP1) and FME (Flagellin-Multiple Epitopes). In addition, the D3 domain of flagellin was replaced by ΔVP1 in FME, yielding FTME (Flagellin-Truncated VP1-Multiple Epitopes). The immunogenicity and protective efficacy of the three fusion proteins as novel FMDV vaccine candidates were evaluated. The results showed that FT, FME, and FTME elicited significant FMDV-specific IgG responses at 10 μg/dose compared with the mock group (P < 0.05), with FTME producing the highest response. No significant differences in the antibody response to FTME were observed between different immunization routes or among adjuvants (ISA-206, poly(I·C), MPLA, and CpG-ODN) in mice. In addition, at 30 μg/dose, all three fusion proteins significantly induced neutralizing antibody production and upregulated the levels of some cytokines, including TNF-α, IFN-γ, and IL-12, in guinea pigs. Importantly, all three fusion proteins provided effective protective immunity against FMDV challenge in guinea pigs, though different protection rates were found. The results presented in this study indicate that the FTME fusion protein is a promising novel vaccine candidate for the future prevention and control of foot-and-mouth disease.
Collapse
|
20
|
Gu C, He X, Zheng W, Liu D, Ma X, Jin N. WITHDRAWN: Developing of recombinant bivalent DNA vaccine deliveried by fowlpox virus vector and detecting of immunological activities to foot-and-mouth disease virus (FMDV) in animals. Vet Anim Sci 2019. [DOI: 10.1016/j.vas.2019.100053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
21
|
McKiernan PJ, Lynch P, Ramsey JM, Cryan SA, Greene CM. Knockdown of Gene Expression in Macrophages by microRNA Mimic-Containing Poly (Lactic- co-glycolic Acid) Microparticles. MEDICINES (BASEL, SWITZERLAND) 2018; 5:E133. [PMID: 30558310 PMCID: PMC6313440 DOI: 10.3390/medicines5040133] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 12/10/2018] [Accepted: 12/14/2018] [Indexed: 01/05/2023]
Abstract
Background: microRNA (miRNA) regulate target gene expression through translational repression and/or mRNA degradation and are involved in the regulation of inflammation. Macrophages are key inflammatory cells that are important in chronic inflammatory lung diseases such as cystic fibrosis (CF). Macrophage-expressed miRNA represent therapeutic drug targets, yet delivery of nucleic acids to macrophages has proved challenging. Methods: miRNAs were encapsulated in poly (lactic-co-glycolic acid) (PLGA)-based microparticles using double emulsion solvent evaporation and characterised for physicochemical features. Phorbol myristic acetate (PMA)-differentiated U937 macrophages were transfected with empty PLGA microparticles or those encapsulating a premiR-19b-3p or scrambled control miRNA mimic. miRNA internalisation and knockdown of a miR-19b-3p target gene, secretory leucoprotease inhibitor (SLPI), were determined by qRT-PCR. Results: Microparticle formulations were consistently found to be 2⁻3μm and all had a negative ζ potential (-5 mV to -14 mV). Encapsulation efficiency of premiR-19b-3p was 37.6 ± 13.4%. Levels of mature miR-19b-3p were higher in macrophages after delivery of premiR-19b-3p microparticles compared to empty or scrambled control miRNA-containing microparticles. Significant SLPI knockdown was achieved 72 hours post-delivery of premiR-19b-3p microparticles compared to controls. Conclusions: miRNA-encapsulating PLGA microparticles offer a new treatment paradigm for delivery to macrophages that could potentially be administered to CF lungs via inhalation.
Collapse
Affiliation(s)
- Paul J McKiernan
- Department of Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland.
| | - Patrick Lynch
- Department of Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland.
- Drug Delivery and Advanced Materials Team, School of Pharmacy, Royal College of Surgeons in Ireland, Dublin 2, Ireland.
| | - Joanne M Ramsey
- Drug Delivery and Advanced Materials Team, School of Pharmacy, Royal College of Surgeons in Ireland, Dublin 2, Ireland.
- Centre for Research in Medical Devices (CURAM), RCSI, Dublin and National University of Ireland, Galway H91 HE94, Ireland.
- Trinity Centre for Bioengineering, Trinity College Dublin, Dublin 2, Ireland.
- Tissue Engineering Research Group, Royal College of Surgeons in Ireland, 123 St Stephens Green, Dublin 2, Ireland.
| | - Sally Ann Cryan
- Drug Delivery and Advanced Materials Team, School of Pharmacy, Royal College of Surgeons in Ireland, Dublin 2, Ireland.
- Centre for Research in Medical Devices (CURAM), RCSI, Dublin and National University of Ireland, Galway H91 HE94, Ireland.
- Trinity Centre for Bioengineering, Trinity College Dublin, Dublin 2, Ireland.
- Tissue Engineering Research Group, Royal College of Surgeons in Ireland, 123 St Stephens Green, Dublin 2, Ireland.
| | - Catherine M Greene
- Lung Biology Group, Department of Clinical Microbiology, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland.
| |
Collapse
|
22
|
Lee LYY, Izzard L, Hurt AC. A Review of DNA Vaccines Against Influenza. Front Immunol 2018; 9:1568. [PMID: 30038621 PMCID: PMC6046547 DOI: 10.3389/fimmu.2018.01568] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 06/25/2018] [Indexed: 01/07/2023] Open
Abstract
The challenges of effective vaccination against influenza are gaining more mainstream attention, as recent influenza seasons have reported low efficacy in annual vaccination programs worldwide. Combined with the potential emergence of novel influenza viruses resulting in a pandemic, the need for effective alternatives to egg-produced conventional vaccines has been made increasingly clear. DNA vaccines against influenza have been in development since the 1990s, but the initial excitement over success in murine model trials has been tempered by comparatively poor performance in larger animal models. In the intervening years, much progress has been made to refine the DNA vaccine platform-the rational design of antigens and expression vectors, the development of novel vaccine adjuvants, and the employment of innovative gene delivery methods. This review discusses how these advances have been applied in recent efforts to develop an effective influenza DNA vaccine.
Collapse
|
23
|
Riitho V, Walters AA, Somavarapu S, Lamp B, Rümenapf T, Krey T, Rey FA, Oviedo-Orta E, Stewart GR, Locker N, Steinbach F, Graham SP. Design and evaluation of the immunogenicity and efficacy of a biomimetic particulate formulation of viral antigens. Sci Rep 2017; 7:13743. [PMID: 29062078 PMCID: PMC5653838 DOI: 10.1038/s41598-017-13915-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 09/19/2017] [Indexed: 11/17/2022] Open
Abstract
Subunit viral vaccines are typically not as efficient as live attenuated or inactivated vaccines at inducing protective immune responses. This paper describes an alternative ‘biomimetic’ technology; whereby viral antigens were formulated around a polymeric shell in a rationally arranged fashion with a surface glycoprotein coated on to the surface and non-structural antigen and adjuvant encapsulated. We evaluated this model using BVDV E2 and NS3 proteins formulated in poly-(D, L-lactic-co-glycolic acid) (PLGA) nanoparticles adjuvanted with polyinosinic:polycytidylic acid (poly(I:C) as an adjuvant (Vaccine-NP). This Vaccine-NP was compared to ovalbumin and poly(I:C) formulated in a similar manner (Control-NP) and a commercial adjuvanted inactivated BVDV vaccine (IAV), all inoculated subcutaneously and boosted prior to BVDV-1 challenge. Significant virus-neutralizing activity, and E2 and NS3 specific antibodies were observed in both Vaccine-NP and IAV groups following the booster immunisation. IFN-γ responses were observed in ex vivo PBMC stimulated with E2 and NS3 proteins in both vaccinated groups. We observed that the protection afforded by the particulate vaccine was comparable to the licenced IAV formulation. In conclusion, the biomimetic particulates showed a promising immunogenicity and efficacy profile that may be improved by virtue of being a customisable mode of delivery.
Collapse
Affiliation(s)
- Victor Riitho
- Virology Department, Animal and Plant Health Agency, Woodham Lane, Addlestone, KT15 3NB, United Kingdom.,Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, United Kingdom.,International Livestock Research Institute, P.O. Box 30709, Nairobi, 00100, Kenya
| | - Adam A Walters
- Virology Department, Animal and Plant Health Agency, Woodham Lane, Addlestone, KT15 3NB, United Kingdom.,Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, United Kingdom.,The Jenner Institute, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, United Kingdom
| | | | - Benjamin Lamp
- Institute for Virology, University of Veterinary Medicine Vienna, Veterinaerplatz 1, 1210, Vienna, Austria
| | - Till Rümenapf
- Institute for Virology, University of Veterinary Medicine Vienna, Veterinaerplatz 1, 1210, Vienna, Austria
| | - Thomas Krey
- Institut Pasteur, Unité de Virologie Structurale, Department Virologie, Paris CNRS UMR, 3569, Paris, France.,Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.,German Center for Infection Research (DZIF), 30625, Hannover, Germany
| | - Felix A Rey
- Institut Pasteur, Unité de Virologie Structurale, Department Virologie, Paris CNRS UMR, 3569, Paris, France
| | - Ernesto Oviedo-Orta
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, United Kingdom.,Sanofi Pasteur, 1541, Avenue Marcel Merieux - Campus Merieux, 69280, Marcy, L'Etoile, France
| | - Graham R Stewart
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, United Kingdom
| | - Nicolas Locker
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, United Kingdom
| | - Falko Steinbach
- Virology Department, Animal and Plant Health Agency, Woodham Lane, Addlestone, KT15 3NB, United Kingdom.,Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, United Kingdom
| | - Simon P Graham
- Virology Department, Animal and Plant Health Agency, Woodham Lane, Addlestone, KT15 3NB, United Kingdom. .,Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, United Kingdom. .,The Pirbright Institute, Ash Road, Pirbright, Woking, GU24 0NF, United Kingdom.
| |
Collapse
|
24
|
Ma YP, Ke H, Liang ZL, Ma JY, Hao L, Liu ZX. Protective efficacy of cationic-PLGA microspheres loaded with DNA vaccine encoding the sip gene of Streptococcus agalactiae in tilapia. FISH & SHELLFISH IMMUNOLOGY 2017; 66:345-353. [PMID: 28476676 DOI: 10.1016/j.fsi.2017.05.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 04/21/2017] [Accepted: 05/01/2017] [Indexed: 05/21/2023]
Abstract
Streptococcus agalactiae (S. agalactiae) is an important fish pathogen, which has received more attention in the past decade due to the increasing economic losses in the tilapia industry worldwide. As existing effective vaccines of S. agalactiae in fish have obvious disadvantage, to select immunoprotective antigens and package materials would undoubtedly contribute to the development of novel oral vaccines. In the present study, surface immunogenic protein (sip) was selected from the S. agalactiae serovar I a genomes as immunogenic protein in DNA vaccine form with cationic chitosan and biodegradable and biocompatible PLGA. The pcSip plasmid in cationic-PLGA was successfully expressed in tissues of immunized tilapia and the immunogenicity was assessed in tilapia challenge model. A significant increase was observed in the cytokine levels of IL-1β, TNF-α, CC1, CC2 in spleen and kidney tissues. Furthermore, immunized tilapia conferred different levels of protection against challenge with a lethal dose of highly virulent serovar I a S. agalactiae. Our results indicated that the pcSip plasmid in cationic-PLGA induced high level of antibodies and protection against S. agalactiae infection, could be effective oral DNA vaccine candidates.
Collapse
Affiliation(s)
- Yan-Ping Ma
- Guangdong Provincial Key Laboratory of Livestock Disease Prevention; Guangdong Open Laboratory of Veterinary Public Health; Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
| | - Hao Ke
- Guangdong Provincial Key Laboratory of Livestock Disease Prevention; Guangdong Open Laboratory of Veterinary Public Health; Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China.
| | - Zhi-Ling Liang
- Guangdong Provincial Key Laboratory of Livestock Disease Prevention; Guangdong Open Laboratory of Veterinary Public Health; Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
| | - Jiang-Yao Ma
- Guangdong Provincial Key Laboratory of Livestock Disease Prevention; Guangdong Open Laboratory of Veterinary Public Health; Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
| | - Le Hao
- Guangdong Provincial Key Laboratory of Livestock Disease Prevention; Guangdong Open Laboratory of Veterinary Public Health; Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
| | - Zhen-Xing Liu
- Guangdong Provincial Key Laboratory of Livestock Disease Prevention; Guangdong Open Laboratory of Veterinary Public Health; Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China.
| |
Collapse
|
25
|
Kammona O, Bourganis V, Karamanidou T, Kiparissides C. Recent developments in nanocarrier-aided mucosal vaccination. Nanomedicine (Lond) 2017; 12:1057-1074. [PMID: 28440707 DOI: 10.2217/nnm-2017-0015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
To date, most of the licensed vaccines for mucosal delivery are based on live-attenuated viruses which carry the risk of regaining their pathogenicity. Therefore, the development of efficient nonviral vectors allowing the induction of potent humoral and cell-mediated immunity is regarded as an imperative scientific challenge as well as a commercial breakthrough for the pharma industries. For a successful translation to the clinic, such nanocarriers should protect the antigens from mucosal enzymes, facilitate antigen uptake by microfold cells and allow the copresentation of robust, safe for human use, mucosal adjuvants to antigen-presenting cells. Finally, the developed formulations should exhibit accuracy regarding the administered dose, a major drawback of mucosal vaccines in comparison with parenteral ones.
Collapse
Affiliation(s)
- Olga Kammona
- Chemical Process & Energy Resources Institute, Centre for Research & Technology Hellas, PO Box 60361, 57001 Thessaloniki, Greece
| | - Vassilis Bourganis
- Department of Chemical Engineering, Aristotle University of Thessaloniki, PO Box 472, 54124 Thessaloniki, Greece
| | - Theodora Karamanidou
- Department of Chemical Engineering, Aristotle University of Thessaloniki, PO Box 472, 54124 Thessaloniki, Greece
| | - Costas Kiparissides
- Department of Chemical Engineering, Aristotle University of Thessaloniki, PO Box 472, 54124 Thessaloniki, Greece.,Chemical Process & Energy Resources Institute, Centre for Research & Technology Hellas, PO Box 60361, 57001 Thessaloniki, Greece
| |
Collapse
|
26
|
Liu X, Fang Y, Zhou P, Lu Y, Zhang Q, Xiao S, Dong Z, Pan L, Lv J, Zhang Z, Zhang Y, Wang Y. Chimeric virus-like particles elicit protective immunity against serotype O foot-and-mouth disease virus in guinea pigs. Appl Microbiol Biotechnol 2017; 101:4905-4914. [PMID: 28365796 DOI: 10.1007/s00253-017-8246-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 03/06/2017] [Accepted: 03/09/2017] [Indexed: 10/19/2022]
Abstract
Foot-and-mouth disease (FMD) is an acute and highly contagious disease caused by foot-and-mouth disease virus (FMDV) that can affect cloven-hoofed animal species, leading to severe economic losses worldwide. Therefore, the development of a safe and effective new vaccine to prevent and control FMD is both urgent and necessary. In this study, we developed a chimeric virus-like particle (VLP) vaccine candidate for serotype O FMDV and evaluated its protective immunity in guinea pigs. Chimeric VLPs were formed by the antigenic structural protein VP1 from serotype O and segments of the viral capsid proteins (VP2, VP3, and VP4) from serotype A. The chimeric VLPs elicited significant humoral and cellular immune responses with a higher level of anti-FMDV antibodies and cytokines than the control group. Furthermore, four of the five guinea pigs vaccinated with the chimeric VLPs were completely protected against challenge with 100 50% guinea pig infectious doses (GPID50) of the virulent FMDV strain O/MAY98. These data suggest that chimeric VLPs are potential candidates for the development of new vaccines against FMDV.
Collapse
Affiliation(s)
- Xinsheng Liu
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| | - Yuzhen Fang
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| | - Peng Zhou
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| | - Yanzhen Lu
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.,College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, 730070, China
| | - Qiaoling Zhang
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| | - Shuai Xiao
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.,College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, 730070, China
| | - Zhaoliang Dong
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| | - Li Pan
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| | - Jianliang Lv
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| | - Zhongwang Zhang
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| | - Yongguang Zhang
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China. .,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
| | - Yonglu Wang
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China. .,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
| |
Collapse
|
27
|
Angelos JA, Chigerwe M, Edman JM, Hess JF. Systemic and ocular immune responses in cattle following intranasal vaccination with precipitated or partially solubilized recombinant Moraxella bovis cytotoxin adjuvanted with polyacrylic acid. Am J Vet Res 2017; 77:1411-1418. [PMID: 27901388 DOI: 10.2460/ajvr.77.12.1411] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To evaluate changes in systemic and ocular antibody responses of steers following intranasal vaccination with precipitated or partially solubilized recombinant Moraxella bovis cytotoxin (MbxA). ANIMALS 13 Angus steers with ages ranging from 318 to 389 days and weights ranging from 352 to 437 kg. PROCEDURES Steers were assigned to receive 500 μg of a precipitated (MbxA-P; n = 5) or partially solubilized (MbxA-S; 5) recombinant MbxA subunit adjuvanted with polyacrylic acid. A control group (n = 3) received the adjuvant alone. Each steer received the assigned treatment (1 mL/nostril) on days 0 and 28. Serum and tear samples were collected on days 0 (before vaccination), 14, 28, 42, and 55. Changes in MbxA-neutralizing antibody titers and MbxA-specific IgG concentrations in serum and tears and changes in MbxA-specific IgA concentrations in tears were measured. RESULTS Mean fold changes in MbxA-specific IgG concentration in serum and tears and MbxA-neutralizing antibody titer in tears for the MbxA-P group were significantly greater than those for the MbxA-S and control groups. Mean serum MbxA-neutralizing antibody titer did not differ among the 3 groups. Although the mean fold change in tear MbxA-specific IgA concentration differed significantly among the groups in the overall analysis, post hoc comparisons failed to identify any significant pairwise differences. CONCLUSIONS AND CLINICAL RELEVANCE Systemic and ocular immune responses induced by intranasal administration of the MbxA-P vaccine were superior to those induced by the MbxA-S vaccine. Additional research is necessary to determine whether the MbxA-P vaccine can prevent naturally occurring infectious bovine keratoconjunctivitis.
Collapse
|
28
|
Lyons NA, Lyoo YS, King DP, Paton DJ. Challenges of Generating and Maintaining Protective Vaccine-Induced Immune Responses for Foot-and-Mouth Disease Virus in Pigs. Front Vet Sci 2016; 3:102. [PMID: 27965966 PMCID: PMC5127833 DOI: 10.3389/fvets.2016.00102] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 11/01/2016] [Indexed: 12/31/2022] Open
Abstract
Vaccination can play a central role in the control of outbreaks of foot-and-mouth disease (FMD) by reducing both the impact of clinical disease and the extent of virus transmission between susceptible animals. Recent incursions of exotic FMD virus lineages into several East Asian countries have highlighted the difficulties of generating and maintaining an adequate immune response in vaccinated pigs. Factors that impact vaccine performance include (i) the potency, antigenic payload, and formulation of a vaccine; (ii) the antigenic match between the vaccine and the heterologous circulating field strain; and (iii) the regime (timing, frequency, and herd-level coverage) used to administer the vaccine. This review collates data from studies that have evaluated the performance of foot-and-mouth disease virus vaccines at the individual and population level in pigs and identifies research priorities that could provide new insights to improve vaccination in the future.
Collapse
Affiliation(s)
- Nicholas A. Lyons
- The Pirbright Institute, Pirbright, UK
- European Commission for the Control of Foot-and-Mouth Disease, Food and Agriculture Organization of the United Nations, Rome, Italy
| | - Young S. Lyoo
- College of Veterinary Medicine, Konkuk University, Seoul, South Korea
| | | | | |
Collapse
|
29
|
Shakya AK, Chowdhury MYE, Tao W, Gill HS. Mucosal vaccine delivery: Current state and a pediatric perspective. J Control Release 2016; 240:394-413. [PMID: 26860287 PMCID: PMC5381653 DOI: 10.1016/j.jconrel.2016.02.014] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 01/21/2016] [Accepted: 02/05/2016] [Indexed: 12/30/2022]
Abstract
Most childhood infections occur via the mucosal surfaces, however, parenterally delivered vaccines are unable to induce protective immunity at these surfaces. In contrast, delivery of vaccines via the mucosal routes can allow antigens to interact with the mucosa-associated lymphoid tissue (MALT) to induce both mucosal and systemic immunity. The induced mucosal immunity can neutralize the pathogen on the mucosal surface before it can cause infection. In addition to reinforcing the defense at mucosal surfaces, mucosal vaccination is also expected to be needle-free, which can eliminate pain and the fear of vaccination. Thus, mucosal vaccination is highly appealing, especially for the pediatric population. However, vaccine delivery across mucosal surfaces is challenging because of the different barriers that naturally exist at the various mucosal surfaces to keep the pathogens out. There have been significant developments in delivery systems for mucosal vaccination. In this review we provide an introduction to the MALT, highlight barriers to vaccine delivery at different mucosal surfaces, discuss different approaches that have been investigated for vaccine delivery across mucosal surfaces, and conclude with an assessment of perspectives for mucosal vaccination in the context of the pediatric population.
Collapse
Affiliation(s)
| | | | - Wenqian Tao
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, USA
| | - Harvinder Singh Gill
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, USA.
| |
Collapse
|
30
|
Nabi H, Rashid I, Ahmad N, Durrani A, Akbar H, Islam S, Bajwa AA, Shehzad W, Ashraf K, Imran N. Induction of specific humoral immune response in mice immunized with ROP18 nanospheres from Toxoplasma gondii. Parasitol Res 2016; 116:359-370. [DOI: 10.1007/s00436-016-5298-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 10/13/2016] [Indexed: 01/05/2023]
|
31
|
Affiliation(s)
- Mingming Zhang
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, 236 Baidi Road, Nankai District, Tianjin 300192, China
| | - Yanhang Hong
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, 236 Baidi Road, Nankai District, Tianjin 300192, China
| | - Wenjuan Chen
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, 236 Baidi Road, Nankai District, Tianjin 300192, China
| | - Chun Wang
- Department
of Biomedical Engineering, University of Minnesota, 7-105 Hasselmo
Hall, 312 Church Street S. E., Minneapolis, Minnesota 55455, United States
| |
Collapse
|
32
|
Kaikabo AA, AbdulKarim SM, Abas F. Evaluation of the efficacy of chitosan nanoparticles loaded ΦKAZ14 bacteriophage in the biological control of colibacillosis in chickens. Poult Sci 2016; 96:295-302. [PMID: 27702916 PMCID: PMC5266084 DOI: 10.3382/ps/pew255] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Revised: 03/12/2016] [Accepted: 06/14/2016] [Indexed: 12/12/2022] Open
Abstract
Disease inflicted by avian pathogenic Escherichia coli (APEC) causes economic losses and burden to the poultry industry worldwide. In this study, the efficacy of chitosan nanoparticles loaded ΦKAZ14 (C-ΦKAZ14 NPs) as an oral biological therapy for Colibacillosis was evaluated. C-ΦKAZ14 NPs containing 107 PFU/ml of ΦKAZ14 (Myoviridae; T4-like coliphage) bacteriophage were used to treat experimentally APEC-infected COBB 500 broiler chicks. C-ΦKAZ14 NPs and ΦKAZ14 bacteriophage were administered orally in a single dose. The clinical symptoms, mortality, and pathology in the infected birds were recorded and compared with those of control birds that did not receive C-ΦKAZ14 NPs or naked ΦKAZ14 bacteriophage. The results showed that C-ΦKAZ14 NP intervention decreased mortality from 58.33 to 16.7% with an increase in the protection rate from 42.00 to 83.33%. The bacterial colonization of the intestines of infected birds was significantly higher in the untreated control than in the C-ΦKAZ14 NP-treated group (2.30×109 ± 0.02 and 0.79×103 ± 0.10 CFU/mL, respectively) (P ≤ 0.05). Similarly, a significant difference in the fecal shedding of Escherichia coli was observed on d 7 post challenge between the untreated control and the C-ΦKAZ14 NP-treated group (2.35×109 ± 0.05 and 1.58×103 ± 0.06 CFU/mL, respectively) (P ≤ 0.05). Similar trends were observed from d 14 until d 21 when the experiment was terminated. Treatment with C-ΦKAZ14 NPs improved the body weights of the infected chicks. A difference in body weight on d 7 post challenge was observed between the untreated control and the C-ΦKAZ14 NP-treated group (140 ± 20 g and 160 ± 20 g, respectively). The increase was significant (P ≤ 0.05) on d 21 between the 2 groups (240 ± 30 g and 600 ± 80 g, respectively). Consequently, the clinical signs and symptoms were ameliorated upon treatment with C-ΦKAZ14 NPs compared with infected untreated birds. In all, based on the results, it can be concluded that the encapsulation of bacteriophage could enhance bacteriophage therapy and is a valuable approach for controlling APEC infections in poultry.
Collapse
Affiliation(s)
- A A Kaikabo
- Faculty of Food Science and Technology, University Putra Malaysia, 43300 UPM Serdang, Selangor, Malaysia.,Bacteriology Research Department, National Veterinary Research Institute, P.M.B 01, Vom, Nigeria
| | - S M AbdulKarim
- Faculty of Food Science and Technology, University Putra Malaysia, 43300 UPM Serdang, Selangor, Malaysia
| | - F Abas
- Faculty of Food Science and Technology, University Putra Malaysia, 43300 UPM Serdang, Selangor, Malaysia
| |
Collapse
|
33
|
Carneiro C, Correia A, Lima T, Vilanova M, Pais C, Gomes AC, Real Oliveira MEC, Sampaio P. Protective effect of antigen delivery using monoolein-based liposomes in experimental hematogenously disseminated candidiasis. Acta Biomater 2016; 39:133-145. [PMID: 27150234 DOI: 10.1016/j.actbio.2016.05.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 04/22/2016] [Accepted: 05/01/2016] [Indexed: 01/08/2023]
Abstract
UNLABELLED We evaluated the potential of a liposomal antigen delivery system (ADS) containing Candida albicans cell wall surface proteins (CWSP) in mediating protection against systemic candidiasis. Treatment of bone-marrow-derived dendritic cells with CWSP-loaded dioctadecyldimethylammonium bromide:monoolein (DODAB:MO) liposomes enhanced and prolonged their activation comparatively to free antigen, indicating that liposome-entrapped CWSP were released more sustainable. Therefore, we immunized mice with CWSP either in a free form or loaded into two different DODAB:MO liposome formulations, respectively designated as ADS1 and ADS2, prior to intravenous C. albicans infection. Immunization with ADS1, but not with ADS2, conferred significant protection to infected mice, comparatively to immunization with CWSP or empty liposomes as control. ADS1-immunized mice presented significantly higher serum levels of C. albicans-specific antibodies that enhanced phagocytosis of this fungus. In these mice, a mixed cytokine production profile was observed encompassing IFN-γ, IL-4, IL-17A and IL-10. Nevertheless, only production of IL-4, IL-17 and IL-10 was higher than in controls. In this study we demonstrated that DODAB:MO liposomes enhance the immunogenicity of C. albicans antigens and host protection in a murine model of systemic candidiasis. Therefore, this liposomal adjuvant could be a promising candidate to assess in vaccination against this pathogenic fungus. STATEMENT OF SIGNIFICANCE This work describes the immunomodulation capacity of the previously validated antigen delivery system (ADS) composed by dioctadecyldimethylammonium bromide (DODAB) and monoolein (MO) lipids incorporating the cell wall surface proteins (CWSP) from C. albicans. Here, we not only present the ability of this system in facilitating antigen uptake by DCs in vitro, but also that this system induces higher levels of pro-inflammatory cytokines and opsonizing specific IgG antibodies in serum of mice immunized subcutaneously. We show that the ADS are efficient nanocarrier and modulate the immune response against intravenous C. albicans infection favoring mouse protection. In sum, we show that the incorporation of C. albicans antigens in DODAB:MO nanocarries are a promising vaccine strategy against C. albicans fungal infection.
Collapse
|
34
|
Xiao Y, Chen HY, Wang Y, Yin B, Lv C, Mo X, Yan H, Xuan Y, Huang Y, Pang W, Li X, Yuan YA, Tian K. Large-scale production of foot-and-mouth disease virus (serotype Asia1) VLP vaccine in Escherichia coli and protection potency evaluation in cattle. BMC Biotechnol 2016; 16:56. [PMID: 27371162 PMCID: PMC4930597 DOI: 10.1186/s12896-016-0285-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 06/21/2016] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Foot-and-mouth disease (FMD) is an acute, highly contagious disease that infects cloven-hoofed animals. Vaccination is an effective means of preventing and controlling FMD. Compared to conventional inactivated FMDV vaccines, the format of FMDV virus-like particles (VLPs) as a non-replicating particulate vaccine candidate is a promising alternative. RESULTS In this study, we have developed a co-expression system in E. coli, which drove the expression of FMDV capsid proteins (VP0, VP1, and VP3) in tandem by a single plasmid. The co-expressed FMDV capsid proteins (VP0, VP1, and VP3) were produced in large scale by fermentation at 10 L scale and the chromatographic purified capsid proteins were auto-assembled as VLPs in vitro. Cattle vaccinated with a single dose of the subunit vaccine, comprising in vitro assembled FMDV VLP and adjuvant, developed FMDV-specific antibody response (ELISA antibodies and neutralizing antibodies) with the persistent period of 6 months. Moreover, cattle vaccinated with the subunit vaccine showed the high protection potency with the 50 % bovine protective dose (PD50) reaching 11.75 PD50 per dose. CONCLUSIONS Our data strongly suggest that in vitro assembled recombinant FMDV VLPs produced from E. coli could function as a potent FMDV vaccine candidate against FMDV Asia1 infection. Furthermore, the robust protein expression and purification approaches described here could lead to the development of industrial level large-scale production of E. coli-based VLPs against FMDV infections with different serotypes.
Collapse
Affiliation(s)
- Yan Xiao
- National Research Center for Veterinary Medicine, Road Cuiwei, High-Tech District, Luoyang, 471003, People's Republic of China
| | - Hong-Ying Chen
- National University of Singapore (Suzhou) Research Institute, 377 Lin Quan Street, Suzhou Industrial Park, Jiangsu, 215123, China
| | - Yuzhou Wang
- National Research Center for Veterinary Medicine, Road Cuiwei, High-Tech District, Luoyang, 471003, People's Republic of China
| | - Bo Yin
- National University of Singapore (Suzhou) Research Institute, 377 Lin Quan Street, Suzhou Industrial Park, Jiangsu, 215123, China
| | - Chaochao Lv
- National Research Center for Veterinary Medicine, Road Cuiwei, High-Tech District, Luoyang, 471003, People's Republic of China
| | - Xiaobing Mo
- National University of Singapore (Suzhou) Research Institute, 377 Lin Quan Street, Suzhou Industrial Park, Jiangsu, 215123, China.,Department of Biological Sciences and Centre for Bioimaging Sciences, National University of Singapore, 14 Science Drive 4, Singapore, 117543, Singapore
| | - He Yan
- National Research Center for Veterinary Medicine, Road Cuiwei, High-Tech District, Luoyang, 471003, People's Republic of China
| | - Yajie Xuan
- National Research Center for Veterinary Medicine, Road Cuiwei, High-Tech District, Luoyang, 471003, People's Republic of China
| | - Yuxin Huang
- National Research Center for Veterinary Medicine, Road Cuiwei, High-Tech District, Luoyang, 471003, People's Republic of China
| | - Wenqiang Pang
- National Research Center for Veterinary Medicine, Road Cuiwei, High-Tech District, Luoyang, 471003, People's Republic of China
| | - Xiangdong Li
- National Research Center for Veterinary Medicine, Road Cuiwei, High-Tech District, Luoyang, 471003, People's Republic of China
| | - Y Adam Yuan
- National University of Singapore (Suzhou) Research Institute, 377 Lin Quan Street, Suzhou Industrial Park, Jiangsu, 215123, China. .,Department of Biological Sciences and Centre for Bioimaging Sciences, National University of Singapore, 14 Science Drive 4, Singapore, 117543, Singapore.
| | - Kegong Tian
- National Research Center for Veterinary Medicine, Road Cuiwei, High-Tech District, Luoyang, 471003, People's Republic of China. .,College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, 450002, People's Republic of China.
| |
Collapse
|
35
|
Fischer S, Diers S, Bauerfeind R, Czerny CP, Neumann S. Dynamics of salivary immunoglobulin A and serum interleukin 6 levels in newborn calves. Livest Sci 2016. [DOI: 10.1016/j.livsci.2016.05.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
36
|
Lactic acid bacteria as mucosal delivery vehicles: a realistic therapeutic option. Appl Microbiol Biotechnol 2016; 100:5691-701. [DOI: 10.1007/s00253-016-7557-x] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 04/12/2016] [Accepted: 04/14/2016] [Indexed: 12/11/2022]
|
37
|
Kaur A, Jyoti K, Rai S, Sidhu R, Pandey RS, Jain UK, Katyal A, Madan J. Tetanus toxoid-loaded cationic non-aggregated nanostructured lipid particles triggered strong humoral and cellular immune responses. J Microencapsul 2016; 33:263-73. [DOI: 10.3109/02652048.2016.1169324] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Amandeep Kaur
- Department of Pharmaceutics, Chandigarh College of Pharmacy, Mohali, Punjab, India
| | - Kiran Jyoti
- Department of Pharmaceutics, Chandigarh College of Pharmacy, Mohali, Punjab, India
| | - Shweta Rai
- Dr. B.R Ambedkar Centre for Biomedical Research, University of Delhi, Delhi, India
| | - Rupinder Sidhu
- Department of Pharmaceutics, Chandigarh College of Pharmacy, Mohali, Punjab, India
| | - Ravi Shankar Pandey
- SLT Institute of Pharmaceutical Sciences, Guru Ghasidas University, Bilaspur, Chhattisgarh, India
| | - Upendra Kumar Jain
- Department of Pharmaceutics, Chandigarh College of Pharmacy, Mohali, Punjab, India
| | - Anju Katyal
- Dr. B.R Ambedkar Centre for Biomedical Research, University of Delhi, Delhi, India
| | - Jitender Madan
- Department of Pharmaceutics, Chandigarh College of Pharmacy, Mohali, Punjab, India
| |
Collapse
|
38
|
Protection against Foot-and-Mouth Disease Virus in Guinea Pigs via Oral Administration of Recombinant Lactobacillus plantarum Expressing VP1. PLoS One 2015; 10:e0143750. [PMID: 26629822 PMCID: PMC4667879 DOI: 10.1371/journal.pone.0143750] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 11/09/2015] [Indexed: 11/19/2022] Open
Abstract
Mucosal vaccination is an effective strategy for generating antigen-specific immune responses against mucosal infections of foot-and-mouth disease virus (FMDV). In this study, Lactobacillus plantarum strains NC8 and WCFS1 were used as oral delivery vehicles containing a pSIP411-VP1 recombinant plasmid to initiate mucosal and systemic immune responses in guinea pigs. Guinea pigs were orally vaccinated (three doses) with NC8-pSIP411, NC8-pSIP411-VP1, WCFS1-pSIP411, WCFS1-pSIP411-VP1 or milk. Animals immunized with NC8-pSIP411-VP1 and WCFS1-pSIP411-VP1 developed high levels of antigen-specific serum IgG, IgA, IgM, mucosal secretory IgA (sIgA) and neutralizing antibodies, and revealed stronger cell-mediated immune responses and enhanced protection against FMDV challenge compared with control groups. The recombinant pSIP411-VP1 effectively improved immunoprotection against FMDV in guinea pigs.
Collapse
|
39
|
Pan L, Zhang Z, Lv J, Zhou P, Hu W, Fang Y, Chen H, Liu X, Shao J, Zhao F, Ding Y, Lin T, Chang H, Zhang J, Zhang Y, Wang Y. Induction of mucosal immune responses and protection of cattle against direct-contact challenge by intranasal delivery with foot-and-mouth disease virus antigen mediated by nanoparticles. Int J Nanomedicine 2014; 9:5603-18. [PMID: 25506214 PMCID: PMC4260661 DOI: 10.2147/ijn.s72318] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The aim of this study was to enhance specific mucosal, systemic, and cell-mediated immunity and to induce earlier onset of protection against direct-contact challenge in cattle by intranasal delivery of a nanoparticle-based nasal vaccine against type A foot-and-mouth disease (FMD). In this study, two kinds of nanoparticle-based nasal vaccines against type A FMD were designed: (1) chitosan-coated poly(lactic-co-glycolic acid) (PLGA) loaded with plasmid DNA (Chi-PLGA-DNA) and (2) chitosan-trehalose and inactivated foot-and-mouth disease virus (FMDV) (Chi-Tre-Inactivated). Cattle were immunized by an intranasal route with nanoparticles and then challenged for 48 hours by direct contact with two infected donor cattle per pen. Donors were inoculated intradermally in the tongue 48 hours before challenge, with 0.2 mL cattle-passaged FMDV. Serological and mucosal antibody responses were evaluated, and virus excretion and the number of contact infections were quantified. FMDV-specific secretory immunoglobulin (Ig)A (sIgA) antibodies in nasal washes were initially detected at 4 days postvaccination (dpv) with two kinds of nanoparticles. The highest levels of sIgA expression were observed in nasal washes, at 10 dpv, from animals with Chi-PLGA-DNA nanoparticles, followed by animals immunized once by intranasal route with a double dose of Chi-Tre-Inactivated nanoparticles and animals immunized by intranasal route three times with Chi-Tre-Inactivated nanoparticles (P<0.05). FMDV-specific IgA antibodies in serum showed a similar pattern. All animals immunized by intranasal route developed low levels of detectable IgG in serum at 10 dpv. Following stimulation with FMDV, the highest levels of proliferation were observed in splenocytes harvested from Chi-PLGA-DNA-immunized animals, followed by proliferation of cells harvested from Chi-Tre-Inactivated nanoparticle-immunized animals (P<0.05). Higher protection rates were associated with the highest sIgA antibody responses induced in the Chi-PLGA-DNA nanoparticle-immunized group. Only one animal was clinically affected with mild signs after 7 days of contact challenge, after a delay of 2–3 days compared with the clinically affected negative-control group. Of the five animals directly challenged that were vaccinated by intranasal route with a double dose of Chi-Tre-Inactivated, four were clinically infected; however, the degree of severity of disease in this group was lower than in control cattle. The number of viral RNA copies in nasal swabs from the vaccinated, severely infected group was significantly higher than in swabs from the vaccinated, clinically protected group. These data suggested that intranasal delivery of Chi-PLGA-DNA nanoparticles resulted in higher levels of mucosal, systemic, and cell-mediated immunity than did of Chi-Tre-Inactivated nanoparticles. In conclusion, although intranasal delivery with FMDV antigen mediated by nanoparticles did not provide complete clinical protection, it reduced disease severity and virus excretion and delayed clinical symptoms. Chi-PLGA-DNA nanoparticle vaccines have potential as a nasal delivery system for vaccines.
Collapse
Affiliation(s)
- Li Pan
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou, Gansu, People's Republic of China ; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, People's Republic of China
| | - Zhongwang Zhang
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou, Gansu, People's Republic of China ; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, People's Republic of China
| | - Jianliang Lv
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou, Gansu, People's Republic of China ; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, People's Republic of China
| | - Peng Zhou
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou, Gansu, People's Republic of China ; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, People's Republic of China
| | - Wenfa Hu
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou, Gansu, People's Republic of China ; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, People's Republic of China
| | - Yuzhen Fang
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou, Gansu, People's Republic of China ; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, People's Republic of China
| | - Haotai Chen
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou, Gansu, People's Republic of China ; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, People's Republic of China
| | - Xinsheng Liu
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou, Gansu, People's Republic of China ; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, People's Republic of China
| | - Junjun Shao
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou, Gansu, People's Republic of China ; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, People's Republic of China
| | - Furong Zhao
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou, Gansu, People's Republic of China ; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, People's Republic of China
| | - Yaozhong Ding
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou, Gansu, People's Republic of China ; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, People's Republic of China
| | - Tong Lin
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou, Gansu, People's Republic of China ; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, People's Republic of China
| | - Huiyun Chang
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou, Gansu, People's Republic of China ; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, People's Republic of China
| | - Jie Zhang
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou, Gansu, People's Republic of China ; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, People's Republic of China
| | - Yongguang Zhang
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou, Gansu, People's Republic of China ; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, People's Republic of China
| | - Yonglu Wang
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou, Gansu, People's Republic of China ; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, People's Republic of China
| |
Collapse
|
40
|
Xu Y, Yuen PW, Lam JKW. Intranasal DNA Vaccine for Protection against Respiratory Infectious Diseases: The Delivery Perspectives. Pharmaceutics 2014; 6:378-415. [PMID: 25014738 PMCID: PMC4190526 DOI: 10.3390/pharmaceutics6030378] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 06/20/2014] [Accepted: 06/24/2014] [Indexed: 11/16/2022] Open
Abstract
Intranasal delivery of DNA vaccines has become a popular research area recently. It offers some distinguished advantages over parenteral and other routes of vaccine administration. Nasal mucosa as site of vaccine administration can stimulate respiratory mucosal immunity by interacting with the nasopharyngeal-associated lymphoid tissues (NALT). Different kinds of DNA vaccines are investigated to provide protection against respiratory infectious diseases including tuberculosis, coronavirus, influenza and respiratory syncytial virus (RSV) etc. DNA vaccines have several attractive development potential, such as producing cross-protection towards different virus subtypes, enabling the possibility of mass manufacture in a relatively short time and a better safety profile. The biggest obstacle to DNA vaccines is low immunogenicity. One of the approaches to enhance the efficacy of DNA vaccine is to improve DNA delivery efficiency. This review provides insight on the development of intranasal DNA vaccine for respiratory infections, with special attention paid to the strategies to improve the delivery of DNA vaccines using non-viral delivery agents.
Collapse
Affiliation(s)
- Yingying Xu
- Department of Pharmacology & Pharmacy, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Pokfulam, 21 Sassoon Road, Hong Kong, China.
| | - Pak-Wai Yuen
- Department of Pharmacology & Pharmacy, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Pokfulam, 21 Sassoon Road, Hong Kong, China.
| | - Jenny Ka-Wing Lam
- Department of Pharmacology & Pharmacy, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Pokfulam, 21 Sassoon Road, Hong Kong, China.
| |
Collapse
|
41
|
Su LK, Yu F, Li ZF, Zeng C, Xu QA, Fan MW. Intranasal co-delivery of IL-6 gene enhances the immunogenicity of anti-caries DNA vaccine. Acta Pharmacol Sin 2014; 35:592-8. [PMID: 24705100 DOI: 10.1038/aps.2013.184] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 11/26/2013] [Indexed: 11/09/2022] Open
Abstract
AIM To investigate the effects of co-delivering IL-6 expressing plasmid pCI-IL-6 on the immunogenicity of the anti-caries DNA vaccine pCIA-P, which encodes the surface protein antigen PAc of Streptococcus mutans. METHODS Plasmid pCI-IL-6 was constructed by inserting the murine IL-6 gene into the pCI vector. Expression of IL-6 in vitro was assessed using Western blot analysis. BALB/c mice were intranasally co-immunized with pCIA-P plus pCI-IL-6 on d 0 and 14. Anti-PAc IgG and secretory IgA (sIgA) were assessed by ELISA. Splenocytes from the mice were re-stimulated with the PAc protein, and IFN-γ and IL-4 production was measured using ELISA. Splenocyte proliferation was analyzed with flow cytometry. Rats were similarly immunized, and dental caries scores were determined using the Keyes method. RESULTS Marked expression of IL-6 was found in COS-7 cells transfected with pCI-IL-6. In the pCI-IL-6 co-immunized mice, the specific IgG antibodies in serum and sIgA antibodies in saliva were significantly higher than those in the control mice at weeks 4 and 8. Moreover, the secretion of IFN-γ from splenocytes in response to re-stimulation with PAc protein was significantly higher in the pCI-IL-6 co-immunized mice than that in the control mice, whereas the secretion of IL-4 had no significant difference. The proliferation of splenocytes from the pCI-IL-6 co-immunized mice was significantly higher than that from the mice immunized with pCIA-P and pCI vector. In the rat caries model, the pCI-IL-6 co-immunization rats displayed lower caries scores than the control rats. CONCLUSION Intranasal co-delivery of IL-6 gene significantly enhances the immunogenicity of the anti-caries DNA vaccine.
Collapse
|
42
|
Costa D, Valente AJM, Miguel MG, Queiroz J. Plasmid DNA hydrogels for biomedical applications. Adv Colloid Interface Sci 2014; 205:257-64. [PMID: 24011472 DOI: 10.1016/j.cis.2013.08.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 08/05/2013] [Accepted: 08/05/2013] [Indexed: 01/05/2023]
Abstract
In the last few years, our research group has focused on the design and development of plasmid DNA (pDNA) based systems as devices to be used therapeutically in the biomedical field. Biocompatible macro and micro plasmid DNA gels were prepared by a cross-linking reaction. For the first time, the pDNA gels have been investigated with respect to their swelling in aqueous solution containing different additives. Furthermore, we clarified the fundamental and basic aspects of the solute release mechanism from pDNA hydrogels and the significance of this information is enormous as a basic tool for the formulation of pDNA carriers for drug/gene delivery applications. The co-delivery of a specific gene and anticancer drugs, combining chemical and gene therapies in the treatment of cancer was the main challenge of our research. Significant progresses have been made with a new p53 encoding pDNA microgel that is suitable for the loading and release of pDNA and doxorubicin. This represents a strong valuable finding in the strategic development of systems to improve cancer cure through the synergetic effect of chemical and gene therapy.
Collapse
Affiliation(s)
- Diana Costa
- CICS - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6201-001 Covilhã, Portugal.
| | | | - M Graça Miguel
- Department of Chemistry, University of Coimbra, Coimbra, Portugal
| | - João Queiroz
- CICS - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6201-001 Covilhã, Portugal
| |
Collapse
|
43
|
Cheon IS, Park SM, Lee HJ, Hong JE, Ji SY, Shim BS, Kim KH, Heo PS, Kim YY, Jung HJ, Ka H, Han SH, Song M, Yun CH. Functional characteristics of porcine peripheral T cells stimulated with IL-2 or IL-2 and PMA. Res Vet Sci 2013; 96:54-61. [PMID: 24374118 DOI: 10.1016/j.rvsc.2013.11.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2013] [Revised: 10/29/2013] [Accepted: 11/30/2013] [Indexed: 12/17/2022]
Abstract
In human or mouse, mature T cells express either CD4 or CD8, resulting in different functions in the periphery. Interestingly, porcine CD4 and CD8 double positive (DP) T cells are present in the blood, and their proportions change from youth to adulthood. However, the features of these cells in swine are poorly understood. We investigated the fate of porcine peripheral T cells based on their functional characteristics, including proliferation and the expression of CD4 and CD8 co-receptors. The results showed that all the populations changed their CD8 expression in a time-dependent manner and porcine T cells had different proliferative pattern from human T cells. The results further revealed that Th2 cytokines were increased later in porcine T cells compared to human T cells upon stimulation with IL-2+PMA. Collectively, we found that the fate of porcine peripheral T cells is different from that of human T cells, and the changes occur in a time- and stimulation-dependent manner.
Collapse
Affiliation(s)
- In Su Cheon
- Department of Agricultural Biotechnology, and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea; Laboratory Sciences Division, International Vaccine Institute, Seoul, Republic of Korea
| | - Sung-Moo Park
- Department of Agricultural Biotechnology, and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea; Laboratory Sciences Division, International Vaccine Institute, Seoul, Republic of Korea; Center for Food and Bioconvergence, Seoul National University, Seoul, Republic of Korea
| | - Hye Jin Lee
- Department of Agricultural Biotechnology, and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Ji Eun Hong
- Department of Agricultural Biotechnology, and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Sang Yun Ji
- Department of Agricultural Biotechnology, and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea; National Institute of Animal Science, RDA, Republic of Korea
| | - Byoung-Shik Shim
- Department of Agricultural Biotechnology, and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea; Laboratory Sciences Division, International Vaccine Institute, Seoul, Republic of Korea
| | - Kwang Ho Kim
- Department of Agricultural Biotechnology, and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Pil Seung Heo
- Department of Agricultural Biotechnology, and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Yoo Yong Kim
- Department of Agricultural Biotechnology, and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Hyun Jung Jung
- National Institute of Animal Science, RDA, Republic of Korea
| | - Hakhyun Ka
- Department of Biological Science and Technology, Yonsei University, Wonju, Republic of Korea
| | - Seung Hyun Han
- Department of Oral Microbiology & Immunology, Dental Research Institute, BK21 Program, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Manki Song
- Laboratory Sciences Division, International Vaccine Institute, Seoul, Republic of Korea.
| | - Cheol-Heui Yun
- Department of Agricultural Biotechnology, and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea; Center for Food and Bioconvergence, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
44
|
Wang J, Zhu R, Gao B, Wu B, Li K, Sun X, Liu H, Wang S. The enhanced immune response of hepatitis B virus DNA vaccine using SiO2@LDH nanoparticles as an adjuvant. Biomaterials 2013; 35:466-78. [PMID: 24099705 DOI: 10.1016/j.biomaterials.2013.09.060] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 09/17/2013] [Indexed: 02/06/2023]
Abstract
Various approaches have been used to improve systemic immune response to infectious disease or virus, and DNA vaccination has been demonstrated to be one of these effective ways to elicit protective immunity against pathogens. Our previous studies showed that layered double hydroxides (LDH) nanoparticles could be efficiently taken up by the MDDCs and had an adjuvant activity for DC maturation. To further enhance the immune adjuvant activity of LDH, core-shell structure SiO2@LDH nanoparticles were synthesized with an average diameter of about 210 nm. And its high transfection efficiency in vitro was demonstrated by using GFP expression plasmid as model DNA. Exposing SiO2@LDH nanoparticles to macrophages caused a higher dose-dependent expression of IFN-γ, IL-6, CD86 and MHC II, compared with SiO2 and LDH respectively. Furthermore, in vivo immunization of BALB/c mice indicated that, DNA vaccine loaded-SiO2@LDH nanoparticles not only induced much higher serum antibody response than naked DNA vaccine and plain nanoparticles, but also obviously promoted T-cell proliferation and skewed T helper to Th1 polarization. Additionally, it was proved that the caveolae-mediated uptake of SiO2@LDH nanoparticles by macrophage lead to macrophages activation via NF-κB signaling pathway. Our results indicate that SiO2@LDH nanoparticles could serve as a potential non-viral gene delivery system.
Collapse
Affiliation(s)
- Jin Wang
- Tenth People's Hospital, School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, PR China
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Mucosal application of cationic poly(D,L-lactide-co-glycolide) microparticles as carriers of DNA vaccine and adjuvants to protect chickens against infectious bursal disease. Vaccine 2013; 31:3656-62. [PMID: 23777953 DOI: 10.1016/j.vaccine.2013.06.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 05/28/2013] [Accepted: 06/04/2013] [Indexed: 11/21/2022]
Abstract
Infectious bursal disease virus (IBDV) is an immunosuppressive virus of chickens. The virus protein (VP) 2 induces neutralizing antibodies, which protect chickens against the disease. The aim of this study was to develop a cationic poly(d,l-lactide-co-glycolide) (PLGA) microparticle (MP) based IBDV-VP2 DNA vaccine (MP-IBDV-DNA) for chickens to be delivered orally and by eye drop route. The tested IBDV-VP2 DNA vaccines were immunogenic for specific-pathogen-free chickens and induced an antibody response after intramuscular application. Co-inoculation with a plasmid encoding chicken IL-2 (chIL-2) or CpG-ODN did not significantly improve protection against IBDV challenge. However, the application of a MP-IBDV-DNA vaccine alone or in combination with a delayed oral and eye drop application of cationic MP loaded with CpG-ODN or chIL-2 improved protection against challenge. The MP-IBDV-DNA-vaccinated chickens showed less pathological and histopathological bursal lesions, a reduced IBDV antigen load as well as T-cell influx into the bursa of Fabricius (BF) compared to the other groups (p<0.05). The addition of chIL-2 loaded MP improved challenge virus clearance from the BF as demonstrated by lower neutralizing antibody titers and reduced IL-4 and IFN-α mRNA expression in the bursa at 7 days postchallenge compared to the other challenged groups. Overall, the efficacy of the IBDV-DNA vaccine was improved by adsorption of the DNA vaccine onto cationic PLGA-MP, which also allowed mucosal application of the DNA vaccine.
Collapse
|
46
|
Gerdts V, Mutwiri G, Richards J, van Drunen Littel-van den Hurk S, Potter AA. Carrier molecules for use in veterinary vaccines. Vaccine 2012; 31:596-602. [PMID: 23219438 DOI: 10.1016/j.vaccine.2012.11.067] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Revised: 11/16/2012] [Accepted: 11/25/2012] [Indexed: 11/27/2022]
Abstract
The practice of immunization of animals and humans has been carried out for centuries and is generally accepted as the most cost effective and sustainable method of infectious disease control. Over the past 20 years there have been significant changes in our ability to produce antigens by conventional extraction and purification, recombinant DNA and synthesis. However, many of these products need to be combined with carrier molecules to generate optimal immune responses. This review covers selected topics in the development of carrier technologies for use in the veterinary vaccine field, including glycoconjugate and peptide vaccines, microparticle and nanoparticle formulations, and finally virus-like particles.
Collapse
Affiliation(s)
- Volker Gerdts
- Vaccine and Infectious Disease Organization - International Vaccine Centre, University of Saskatchewan, Saskatoon, SK, Canada
| | | | | | | | | |
Collapse
|
47
|
Rodrigues S, Dionísio M, López CR, Grenha A. Biocompatibility of chitosan carriers with application in drug delivery. J Funct Biomater 2012; 3:615-41. [PMID: 24955636 PMCID: PMC4030999 DOI: 10.3390/jfb3030615] [Citation(s) in RCA: 201] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Revised: 08/03/2012] [Accepted: 08/21/2012] [Indexed: 01/30/2023] Open
Abstract
Chitosan is one of the most used polysaccharides in the design of drug delivery strategies for administration of either biomacromolecules or low molecular weight drugs. For these purposes, it is frequently used as matrix forming material in both nano and micron-sized particles. In addition to its interesting physicochemical and biopharmaceutical properties, which include high mucoadhesion and a great capacity to produce drug delivery systems, ensuring the biocompatibility of the drug delivery vehicles is a highly relevant issue. Nevertheless, this subject is not addressed as frequently as desired and even though the application of chitosan carriers has been widely explored, the demonstration of systems biocompatibility is still in its infancy. In this review, addressing the biocompatibility of chitosan carriers with application in drug delivery is discussed and the methods used in vitro and in vivo, exploring the effect of different variables, are described. We further provide a discussion on the pros and cons of used methodologies, as well as on the difficulties arising from the absence of standardization of procedures.
Collapse
Affiliation(s)
- Susana Rodrigues
- Centre for Molecular and Structural Biomedicine (CBME), Institute for Biotechnology and Bioengineering (IBB), Faculty of Sciences and Technology, University of Algarve, Campus de Gambelas, Faro 8005-139, Portugal.
| | - Marita Dionísio
- Centre for Molecular and Structural Biomedicine (CBME), Institute for Biotechnology and Bioengineering (IBB), Faculty of Sciences and Technology, University of Algarve, Campus de Gambelas, Faro 8005-139, Portugal.
| | - Carmen Remuñán López
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Santiago de Compostela, Campus Vida, Santiago de Compostela 15782, Spain.
| | - Ana Grenha
- Centre for Molecular and Structural Biomedicine (CBME), Institute for Biotechnology and Bioengineering (IBB), Faculty of Sciences and Technology, University of Algarve, Campus de Gambelas, Faro 8005-139, Portugal.
| |
Collapse
|
48
|
Abstract
The vast majority of human pathogens colonize and invade at the mucosal surfaces. Preventing infection at these sites via mucosally active vaccines is a promising and rational approach for vaccine development. However, it is only recently that the stimulation of local immunity at the mucosal surfaces has become a primary objective in addition to inducing systemic immunity. This review describes vaccine formulations designed for mucosal delivery to the nasal-associated lymphoid tissue, via intranasal administration. The association of antigens with mucosal adjuvants and delivery systems is emphasised.
Collapse
Affiliation(s)
- Mehfuz Zaman
- School of Chemistry and Molecular Biosciences (SCMB), The University of Queensland, St. Lucia, 4072 QLD Australia
| | - Saranya Chandrudu
- School of Chemistry and Molecular Biosciences (SCMB), The University of Queensland, St. Lucia, 4072 QLD Australia
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences (SCMB), The University of Queensland, St. Lucia, 4072 QLD Australia
- School of Pharmacy, The University of Queensland, Brisbane, QLD Australia
| |
Collapse
|
49
|
Nanomedicine and veterinary science: the reality and the practicality. Vet J 2012; 193:12-23. [PMID: 22365842 DOI: 10.1016/j.tvjl.2012.01.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Revised: 12/20/2011] [Accepted: 01/03/2012] [Indexed: 01/04/2023]
Abstract
Nanomedicine is a rapidly expanding field with a promising future that is already permeating veterinary science. This review summarises the current applications for nanoparticles in human medicine and explores their potential applicability for veterinary use. The principles underlying the use of nanoparticles in drug delivery, imaging and as vaccine adjuvants are explored along with the unique issues surrounding nanoparticle toxicity and regulatory approval. A brief overview of the properties of different nanoparticle systems including, liposomes, micelles, emulsions and inorganic nanoparticles, is provided, along with a description of their current and potential future applications in veterinary medicine.
Collapse
|