1
|
Dehghan M, Askari H, Tohidfar M, Siadat S, Fatemi F. Improvement of RBD-FC Immunogenicity by Using Alum-Sodium Alginate Adjuvant Against SARS-COV-2. Influenza Other Respir Viruses 2024; 18:e70018. [PMID: 39478310 PMCID: PMC11525037 DOI: 10.1111/irv.70018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 08/31/2024] [Accepted: 09/04/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND Adjuvants use several mechanisms to boost immunogenicity and to modulate immune response. The strength of adsorption of antigen by adjuvants can be a determinant factor for significant improvement of immunopotentiation. METHODS We expressed recombinant RBD-FC in PichiaPink Strain 4 and examined the vaccination of mice by vaccine formulation with different adjuvants (sodium alginate and aluminum hydroxide, alone and together). RESULTS Sodium alginate significantly increased the immunogenicity and stability of RBD-FC antigen, so RBD-FC formulated with combined alginate and alum (AlSa) and sodium alginate alone showed higher antibody titer and stability. Immunogenicity of RBD-FC:AlSa was determined by serological assays including direct enzyme-linked immunosorbent assay (ELISA) and surrogate virus neutralization test (sVNT). High levels of IgGs and neutralizing antibodies were measured in serum of mice immunized with the RBD-FC:AlSa formulation. On the other hand, cytokines IL-10 and INF-γ were severely accumulated in response to RBD-FC:AlSa, and after 10 days, their accumulation was significantly declined, whereas IL-4 showed the highest and the lowest accumulation in response to alum and alginate, respectively. CONCLUSIONS Our data may suggest that combination of alum and sodium alginate has a better compatibility with RBD-FC in vaccine formulation.
Collapse
MESH Headings
- Alginates/chemistry
- Animals
- Mice
- Alum Compounds/administration & dosage
- Adjuvants, Immunologic/administration & dosage
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- SARS-CoV-2/immunology
- Mice, Inbred BALB C
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- Female
- Adjuvants, Vaccine
- COVID-19/prevention & control
- COVID-19/immunology
- Immunogenicity, Vaccine
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/chemistry
- Aluminum Hydroxide/administration & dosage
- Aluminum Hydroxide/immunology
- Aluminum Hydroxide/chemistry
- Humans
- Immunoglobulin G/blood
- Cytokines
- Immunoglobulin Fc Fragments/immunology
Collapse
Affiliation(s)
- Mahboobeh Dehghan
- Department of Cellular and Molecular Biology, Faculty of Life Sciences and BiotechnologyShahid Beheshti UniversityTehranIran
| | - Hossein Askari
- Department of Cellular and Molecular Biology, Faculty of Life Sciences and BiotechnologyShahid Beheshti UniversityTehranIran
| | - Masoud Tohidfar
- Department of Cellular and Molecular Biology, Faculty of Life Sciences and BiotechnologyShahid Beheshti UniversityTehranIran
| | | | - Fataneh Fatemi
- Protein Research CenterShahid Beheshti UniversityTehranIran
| |
Collapse
|
2
|
Tian X, Ai J, Tian X, Wei X. cGAS-STING pathway agonists are promising vaccine adjuvants. Med Res Rev 2024; 44:1768-1799. [PMID: 38323921 DOI: 10.1002/med.22016] [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: 10/17/2023] [Revised: 12/10/2023] [Accepted: 01/09/2024] [Indexed: 02/08/2024]
Abstract
Adjuvants are of critical value in vaccine development as they act on enhancing immunogenicity of antigen and inducing long-lasting immunity. However, there are only a few adjuvants that have been approved for clinical use, which highlights the need for exploring and developing new adjuvants to meet the growing demand for vaccination. Recently, emerging evidence demonstrates that the cGAS-STING pathway orchestrates innate and adaptive immunity by generating type I interferon responses. Many cGAS-STING pathway agonists have been developed and tested in preclinical research for the treatment of cancer or infectious diseases with promising results. As adjuvants, cGAS-STING agonists have demonstrated their potential to activate robust defense immunity in various diseases, including COVID-19 infection. This review summarized the current developments in the field of cGAS-STING agonists with a special focus on the latest applications of cGAS-STING agonists as adjuvants in vaccination. Potential challenges were also discussed in the hope of sparking future research interests to further the development of cGAS-STING as vaccine adjuvants.
Collapse
Affiliation(s)
- Xinyu Tian
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Centre for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Jiayuan Ai
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Centre for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Xiaohe Tian
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Centre for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Centre for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| |
Collapse
|
3
|
Yu X, Cai L, Yao J, Li C, Wang X. Agonists and Inhibitors of the cGAS-STING Pathway. Molecules 2024; 29:3121. [PMID: 38999073 PMCID: PMC11243509 DOI: 10.3390/molecules29133121] [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: 05/31/2024] [Revised: 06/24/2024] [Accepted: 06/24/2024] [Indexed: 07/14/2024] Open
Abstract
The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway is pivotal in immunotherapy. Several agonists and inhibitors of the cGAS-STING pathway have been developed and evaluated for the treatment of various diseases. The agonists aim to activate STING, with cyclic dinucleotides (CDNs) being the most common, while the inhibitors aim to block the enzymatic activity or DNA binding ability of cGAS. Meanwhile, non-CDN compounds and cGAS agonists are also gaining attention. The omnipresence of the cGAS-STING pathway in vivo indicates that its overactivation could lead to undesired inflammatory responses and autoimmune diseases, which underscores the necessity of developing both agonists and inhibitors of the cGAS-STING pathway. This review describes the molecular traits and roles of the cGAS-STING pathway and summarizes the development of cGAS-STING agonists and inhibitors. The information is supposed to be conducive to the design of novel drugs for targeting the cGAS-STING pathway.
Collapse
Affiliation(s)
- Xiaoxuan Yu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Linxiang Cai
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Jingyue Yao
- Department of Pharmacy, Fourth Military Medical University, Xi’an 710032, China;
| | - Cenming Li
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xiaoyong Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| |
Collapse
|
4
|
Silva LA, Souza MF, Carvalho TP, Santana CH, Guedes AC, Oliveira JBS, de Lima PA, Nogueira PRA, de Mello Brandão H, da Paixão TA, Santos RL. Comparative study on alginate/chitosan microcapsules and Montanide ISA 61 as vaccine adjuvants in mice. PLoS One 2024; 19:e0298117. [PMID: 38573916 PMCID: PMC10994407 DOI: 10.1371/journal.pone.0298117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 01/16/2024] [Indexed: 04/06/2024] Open
Abstract
Selection of adjuvant to be combined with the antigen is an extremely important point for formulating effective vaccines. The aim of this study was to evaluate reactogenicity, levels of IgM, IgG and subclasses (IgG1, IgG2b and IgG3), and protection elicited by vaccine formulations with association of chitosan coated alginate or Montanide ISA 61 with γ-irradiated Brucella ovis. The alginate/chitosan biopolymers as well as the Montanide ISA 61 emulsion elicited intense and long-lasting local response, especially when associated with the antigen. However, Montanide ISA 61 induced less intense reactogenicity when compared to alginate/chitosan. Furthermore, γ-irradiated B. ovis with Montanide ISA 61 induced higher levels of IgG2b an important marker of cellular immune response. In conclusion, Montanide ISA 61 resulted in milder reactogenicity when compared to the alginate/chitosan, while it induced a high IgG2b/IgG1 ratio compatible with a Th1 profile response.
Collapse
Affiliation(s)
- Laice A. Silva
- Departamento de Clínica e Cirurgia Veterinária, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Monique F. Souza
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Thaynara P. Carvalho
- Departamento de Clínica e Cirurgia Veterinária, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Clarissa H. Santana
- Departamento de Clínica e Cirurgia Veterinária, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Andressa C. Guedes
- Departamento de Clínica e Cirurgia Veterinária, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Jefferson Bruno S. Oliveira
- Departamento de Clínica e Cirurgia Veterinária, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Pâmela A. de Lima
- Departamento de Clínica e Cirurgia Veterinária, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Paulo Roberto A. Nogueira
- Departamento de Clínica e Cirurgia Veterinária, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Tatiane A. da Paixão
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Renato Lima Santos
- Departamento de Clínica e Cirurgia Veterinária, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
5
|
Sun Z, Zhao H, Ma L, Shi Y, Ji M, Sun X, Ma D, Zhou W, Huang T, Zhang D. The quest for nanoparticle-powered vaccines in cancer immunotherapy. J Nanobiotechnology 2024; 22:61. [PMID: 38355548 PMCID: PMC10865557 DOI: 10.1186/s12951-024-02311-z] [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: 10/18/2023] [Accepted: 01/26/2024] [Indexed: 02/16/2024] Open
Abstract
Despite recent advancements in cancer treatment, this disease still poses a serious threat to public health. Vaccines play an important role in preventing illness by preparing the body's adaptive and innate immune responses to combat diseases. As our understanding of malignancies and their connection to the immune system improves, there has been a growing interest in priming the immune system to fight malignancies more effectively and comprehensively. One promising approach involves utilizing nanoparticle systems for antigen delivery, which has been shown to potentiate immune responses as vaccines and/or adjuvants. In this review, we comprehensively summarized the immunological mechanisms of cancer vaccines while focusing specifically on the recent applications of various types of nanoparticles in the field of cancer immunotherapy. By exploring these recent breakthroughs, we hope to identify significant challenges and obstacles in making nanoparticle-based vaccines and adjuvants feasible for clinical application. This review serves to assess recent breakthroughs in nanoparticle-based cancer vaccinations and shed light on their prospects and potential barriers. By doing so, we aim to inspire future immunotherapies for cancer that harness the potential of nanotechnology to deliver more effective and targeted treatments.
Collapse
Affiliation(s)
- Zhe Sun
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Hui Zhao
- Department of Endodontics, East Branch of Jinan Stomatological Hospital, Jinan, 250000, Shandong, China
| | - Li Ma
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Yanli Shi
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Mei Ji
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Xiaodong Sun
- Department of Endodontics, Gaoxin Branch of Jinan Stomatological Hospital, Jinan, 250000, Shandong, China
| | - Dan Ma
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Wei Zhou
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Tao Huang
- Department of Biomedical Engineering, Graeme Clark Institute, The University of Melbourne, Parkville, VIC, 3010, Australia.
| | - Dongsheng Zhang
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| |
Collapse
|
6
|
Gaglio SC, Perduca M, Zipeto D, Bardi G. Efficiency of Chitosan Nanocarriers in Vaccinology for Mucosal Immunization. Vaccines (Basel) 2023; 11:1333. [PMID: 37631901 PMCID: PMC10459455 DOI: 10.3390/vaccines11081333] [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: 06/30/2023] [Revised: 08/02/2023] [Accepted: 08/04/2023] [Indexed: 08/29/2023] Open
Abstract
The mucosal barrier constitutes a huge surface area, close to 40 m2 in humans, located mostly in the respiratory, gastrointestinal and urogenital tracts and ocular cavities. It plays a crucial role in tissue interactions with the microbiome, dietary antigens and other environmental materials. Effective vaccinations to achieve highly protective mucosal immunity are evolving strategies to counteract several serious diseases including tuberculosis, diphtheria, influenzae B, severe acute respiratory syndrome, Human Papilloma Virus infection and Acquired Immune Deficiency Syndrome. Interestingly, one of the reasons behind the rapid spread of severe acute respiratory syndrome coronavirus 2 variants has been the weakness of local immunization at the level of the respiratory mucosa. Mucosal vaccines can outperform parenteral vaccination as they specifically elicit protective mucosal immune responses blocking infection and transmission. In this scenario, chitosan-based nanovaccines are promising adjuvants-carrier systems that rely on the ability of chitosan to cross tight junctions and enhance particle uptake due to chitosan-specific mucoadhesive properties. Indeed, chitosan not only improves the adhesion of antigens to the mucosa promoting their absorption but also shows intrinsic immunostimulant abilities. Furthermore, by finely tuning the colloidal properties of chitosan, it can provide sustained antigen release to strongly activate the humoral defense. In the present review, we agnostically discuss the potential reasons why chitosan-based vaccine carriers, that efficiently elicit strong immune responses in experimental setups and in some pre-clinical/clinical studies, are still poorly considered for therapeutic formulations.
Collapse
Affiliation(s)
- Salvatore Calogero Gaglio
- Department of Biotechnology, University of Verona, Cà Vignal 1, Strada Le Grazie 15, 37134 Verona, Italy;
| | - Massimiliano Perduca
- Department of Biotechnology, University of Verona, Cà Vignal 1, Strada Le Grazie 15, 37134 Verona, Italy;
| | - Donato Zipeto
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy
| | - Giuseppe Bardi
- Nanobiointeractions & Nanodiagnostics, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| |
Collapse
|
7
|
Süer Ö, Gül A, Hameş EE. Adjuvant action of needle-shaped BC microfibrils. CELLULOSE (LONDON, ENGLAND) 2023; 30:4263-4276. [PMID: 37113141 PMCID: PMC10061392 DOI: 10.1007/s10570-023-05138-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 03/05/2023] [Indexed: 06/19/2023]
Abstract
Bacterial cellulose (BC) is an unbranched biopolymer produced by microorganisms and composed of glucopyranose units linked by β-1,4 bonds. This study investigates the adjuvant action of needle-shaped BC microfibrils (BCmFs) in vitro using bovine serum albumin (BSA) as a model antigen. BC produced by the static culture of Komagataibacter xylinus was then microparticled (1-5 μm) by acid hydrolysis and characterized using Dynamic Light Scattering and Scanning Electron Microscopy. Subsequently, Attenuated Total Reflectance-Fourier-Transform Infrared Spectroscopy, cytotoxicity, TNF-α (tumour necrosis factor-alpha) and IL-6 (interleukin-6) cytokine secretion, and cellular uptake of the BCmFs-BSA conjugate on the human monocyte cell line (U937) differentiated into macrophages were performed. The microfibrils were determined to be 1-5 μm in size, needle-shaped, with a zeta potential of - 32 mV. Their conjugation with the model antigen, BSA, was demonstrated by FTIR analysis. In the cytotoxicity assay, BCmFs-BSA in macrophage cells showed high viability (over 70%). Although the highest TNF-α cytokine level (113 pg/ml) was obtained with BCmFs-BSA (Bovine serum albumin) conjugate (500 µg/ml) and was statistically significant (p = 0.0001) compared to the positive control group (BSA-aluminium hydroxide), IL-6 cytokine levels were not statistically different from those in the control group as desired. It has been shown in macrophage-differentiated U937 cells that microbially synthesized BC in the form of needle-shaped microfibrils (BCmFs) has a high cellular uptake capacity and increases the immunogenicity of the antigen. These results demonstrate for the first time that BCmFs have the potential to serve as a vaccine adjuvant.
Collapse
Affiliation(s)
- Özge Süer
- Department of Bioengineering, Graduate School of Natural and Applied Sciences, Ege University, Izmir, Türkiye
- Department of Food Engineering, Faculty of Engineering, Izmir University of Economics, Izmir, Türkiye
| | - Aytül Gül
- Department of Bioengineering, Graduate School of Natural and Applied Sciences, Ege University, Izmir, Türkiye
| | - Elif Esin Hameş
- Department of Bioengineering, Graduate School of Natural and Applied Sciences, Ege University, Izmir, Türkiye
- Department of Bioengineering, Faculty of Engineering, Ege University, 35100 Bornova, Izmir, Türkiye
| |
Collapse
|
8
|
Reay SL, Jackson EL, Salthouse D, Ferreira AM, Hilkens CMU, Novakovic K. Effective Endotoxin Removal from Chitosan That Preserves Chemical Structure and Improves Compatibility with Immune Cells. Polymers (Basel) 2023; 15:polym15071592. [PMID: 37050208 PMCID: PMC10096541 DOI: 10.3390/polym15071592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/13/2023] [Accepted: 03/17/2023] [Indexed: 04/14/2023] Open
Abstract
Chitosan is one of the most researched biopolymers for healthcare applications, however, being a naturally derived polymer, it is susceptible to endotoxin contamination, which elicits pro-inflammatory responses, skewing chitosan's performance and leading to inaccurate conclusions. It is therefore critical that endotoxins are quantified and removed for in vivo use. Here, heat and mild NaOH treatment are investigated as facile endotoxin removal methods from chitosan. Both treatments effectively removed endotoxin to below the FDA limit for medical devices (<0.5 EU/mL). However, in co-culture with peripheral blood mononuclear cells (PBMCs), only NaOH-treated chitosan prevented TNF-α production. While endotoxin removal is the principal task, the preservation of chitosan's structure is vital for the synthesis and lysozyme degradation of chitosan-based hydrogels. The chemical properties of NaOH-treated chitosan (by FTIR-ATR) were significantly similar to its native composition, whereas the heat-treated chitosan evidenced macroscopic chemical and physical changes associated with the Maillard reaction, deeming this treatment unsuitable for further applications. Degradation studies conducted with lysozyme demonstrated that the degradation rates of native and NaOH-treated chitosan-genipin hydrogels were similar. In vitro co-culture studies showed that NaOH hydrogels did not negatively affect the cell viability of monocyte-derived dendritic cells (moDCs), nor induce phenotypical maturation or pro-inflammatory cytokine release.
Collapse
Affiliation(s)
- Sophie L Reay
- School of Engineering, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| | - Emma L Jackson
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Daniel Salthouse
- School of Engineering, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| | - Ana Marina Ferreira
- School of Engineering, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| | - Catharien M U Hilkens
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Katarina Novakovic
- School of Engineering, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| |
Collapse
|
9
|
Enhancing neutralizing antibodies against receptor binding domain of SARS-CoV-2 by a safe natural adjuvant system. Virus Res 2023; 326:199047. [PMID: 36693449 PMCID: PMC9867563 DOI: 10.1016/j.virusres.2023.199047] [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: 07/15/2022] [Revised: 01/09/2023] [Accepted: 01/15/2023] [Indexed: 01/22/2023]
Abstract
The receptor binding domain (RBD) plays a pivotal role in the viral entry as it enables the engagement of severe acute respiratory syndrome 2 (SARS-CoV-2) with the human angiotensin-converting enzyme 2 (ACE2) receptor for host cell entry. RBD is the major target for developing viral inhibitors and vaccines. Expression of recombinant RBD in E.coli is highly scalable with a low-cost procedure despite its high expression level compared to expression in mammalian and yeast cells. Using an alternative natural adjuvant system instead of alum adjuvant, increased immunogenicity of RBD antigen in serological assay including direct ELISA and surrogate Virus Neutralization Test (sVNT) was demonstrated with high levels of IgGs and neutralizing antibodies in mice sera immunized with RBD:AlSa (Alum and Sodium alginate) formulation. The sVNT is a simple and fast test that can be used instead of the conventional virus neutralization test requiring live virus and BSL3 laboratory to detect total neutralizing antibodies against RBD. Additionally, results showed a safety profile for sodium alginate which supported using it as an alternative natural adjuvant.
Collapse
|
10
|
Yang N, Jin X, Zhu C, Gao F, Weng Z, Du X, Feng G. Subunit vaccines for Acinetobacter baumannii. Front Immunol 2023; 13:1088130. [PMID: 36713441 PMCID: PMC9878323 DOI: 10.3389/fimmu.2022.1088130] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 12/22/2022] [Indexed: 01/15/2023] Open
Abstract
Acinetobacter baumannii is a gram-negative bacterium and a crucial opportunistic pathogen in hospitals. A. baumannii infection has become a challenging problem in clinical practice due to the increasing number of multidrug-resistant strains and their prevalence worldwide. Vaccines are effective tools to prevent and control A. baumannii infection. Many researchers are studying subunit vaccines against A. baumannii. Subunit vaccines have the advantages of high purity, safety, and stability, ease of production, and highly targeted induced immune responses. To date, no A. baumannii subunit vaccine candidate has entered clinical trials. This may be related to the easy degradation of subunit vaccines in vivo and weak immunogenicity. Using adjuvants or delivery vehicles to prepare subunit vaccines can slow down degradation and improve immunogenicity. The common immunization routes include intramuscular injection, subcutaneous injection, intraperitoneal injection and mucosal vaccination. The appropriate immunization method can also enhance the immune effect of subunit vaccines. Therefore, selecting an appropriate adjuvant and immunization method is essential for subunit vaccine research. This review summarizes the past exploration of A. baumannii subunit vaccines, hoping to guide current and future research on these vaccines.
Collapse
Affiliation(s)
- Ning Yang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiao Jin
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chenghua Zhu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Fenglin Gao
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zheqi Weng
- The Second Clinical Medical School of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xingran Du
- Department of Infectious Disease, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China,*Correspondence: Xingran Du, ; Ganzhu Feng,
| | - Ganzhu Feng
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China,*Correspondence: Xingran Du, ; Ganzhu Feng,
| |
Collapse
|
11
|
Nguyen KG, Mantooth SM, Vrabel MR, Zaharoff DA. Intranasal Delivery of Thermostable Subunit Vaccine for Cross-Reactive Mucosal and Systemic Antibody Responses Against SARS-CoV-2. Front Immunol 2022; 13:858904. [PMID: 35592324 PMCID: PMC9110812 DOI: 10.3389/fimmu.2022.858904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 04/01/2022] [Indexed: 11/16/2022] Open
Abstract
Despite the remarkable efficacy of currently approved COVID-19 vaccines, there are several opportunities for continued vaccine development against SARS-CoV-2 and future lethal respiratory viruses. In particular, restricted vaccine access and hesitancy have limited immunization rates. In addition, current vaccines are unable to prevent breakthrough infections, leading to prolonged virus circulation. To improve access, a subunit vaccine with enhanced thermostability was designed to eliminate the need for an ultra-cold chain. The exclusion of infectious and genetic materials from this vaccine may also help reduce vaccine hesitancy. In an effort to prevent breakthrough infections, intranasal immunization to induce mucosal immunity was explored. A prototype vaccine comprised of receptor-binding domain (RBD) polypeptides formulated with additional immunoadjuvants in a chitosan (CS) solution induced high levels of RBD-specific antibodies in laboratory mice after 1 or 2 immunizations. Antibody responses were durable with high titers persisting for at least five months following subcutaneous vaccination. Serum anti-RBD antibodies contained both IgG1 and IgG2a isotypes suggesting that the vaccine induced a mixed Th1/Th2 response. RBD vaccination without CS formulation resulted in minimal anti-RBD responses. The addition of CpG oligonucleotides to the CS plus RBD vaccine formulation increased antibody titers more effectively than interleukin-12 (IL-12). Importantly, generated antibodies were cross-reactive against RBD mutants associated with SARS-CoV-2 variants of concern, including alpha, beta and delta variants, and inhibited binding of RBD to its cognate receptor angiotensin converting enzyme 2 (ACE2). With respect to stability, vaccines did not lose activity when stored at either room temperature (21-22°C) or 4°C for at least one month. When delivered intranasally, vaccines induced RBD-specific mucosal IgA antibodies, which may protect against breakthrough infections in the upper respiratory tract. Altogether, data indicate that the designed vaccine platform is versatile, adaptable and capable of overcoming key constraints of current COVID-19 vaccines.
Collapse
Affiliation(s)
- Khue G. Nguyen
- Joint Department of Biomedical Engineering, University of North Carolina-Chapel Hill and North Carolina State University, Raleigh, NC, United States
| | - Siena M. Mantooth
- Joint Department of Biomedical Engineering, University of North Carolina-Chapel Hill and North Carolina State University, Raleigh, NC, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
| | - Maura R. Vrabel
- Joint Department of Biomedical Engineering, University of North Carolina-Chapel Hill and North Carolina State University, Raleigh, NC, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
| | - David A. Zaharoff
- Joint Department of Biomedical Engineering, University of North Carolina-Chapel Hill and North Carolina State University, Raleigh, NC, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
| |
Collapse
|
12
|
Safarzadeh M, Mohammadi-Yeganeh S, Ghorbani-Bidkorbeh F, Haji Molla Hoseini M. Chitosan based nanoformulation expressing miR-155 as a promising adjuvant to enhance Th1-biased immune responses. Life Sci 2022; 297:120459. [PMID: 35248524 DOI: 10.1016/j.lfs.2022.120459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 02/18/2022] [Accepted: 03/01/2022] [Indexed: 11/15/2022]
Abstract
BACKGROUND AND AIM MiR-155 could act as a key modulator of different aspects of immune system including Th1 responses. In this study, we designed chitosan nanoparticles containing miR-155-expressing plasmid and explored their effects as an adjuvant to enhance Th1 responses for potential future application against intracellular pathogens. METHODS Nanoparticles were formulated by complex coacervation method and characterized for physicochemical and functional characteristics. Transfection efficiency in Raw 264.7 cells, effects on miR-155 target genes and NO production were evaluated. The prepared nanoparticles were co-administered as an adjuvant with ovalbumin to immunize mice and finally production of IFN-γ and IL-4 were measured by ELISA in splenocyte recall responses. RESULTS The prepared nanoparticles had the mean size of 244 nm and zeta potential of +17 mV, respectively. Electrophoresis analysis indicated the high capability of nanoparticles to protect the plasmid from DNaseI degradation. Furthermore, nanoparticles showed an appropriate transfection efficiency in Raw 264.7 cells and could downregulate the expression of miR-155 target genes and also upregulate NO production. In vivo immunization examinations revealed successful shift of T cell responses toward Th1. CONCLUSION Our data suggests the high potential of chitosan nanoparticles containing miR-155-expressing plasmid as an adjuvant for significantly enhanced Th1-biased immune responses upon immunization with a given antigen.
Collapse
Affiliation(s)
- Mehrnoush Safarzadeh
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samira Mohammadi-Yeganeh
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ghorbani-Bidkorbeh
- Department of Pharmaceutics, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mostafa Haji Molla Hoseini
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
13
|
Van Herck S, Feng B, Tang L. Delivery of STING agonists for adjuvanting subunit vaccines. Adv Drug Deliv Rev 2021; 179:114020. [PMID: 34756942 DOI: 10.1016/j.addr.2021.114020] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/16/2021] [Accepted: 10/19/2021] [Indexed: 02/06/2023]
Abstract
Adjuvant is an essential component in subunit vaccines. Many agonists of pathogen recognition receptors have been developed as potent adjuvants to optimize the immunogenicity and efficacy of vaccines. Recently discovered cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway has attracted much attention as it is a key mediator for modulating immune responses. Vaccines adjuvanted with STING agonists are found to mediate a robust immune defense against infections and cancer. In this review, we first discuss the mechanisms of STING agonists in the context of vaccination. Next, we present recent progress in novel STING agonist discovery and the delivery strategies. We next highlight recent work in optimizing the efficacy while minimizing toxicity of STING agonist-assisted subunit vaccines for protection against infectious diseases or treatment of cancer. Finally, we share our perspectives of current issues and future directions in further developing STING agonists for adjuvanting subunit vaccines.
Collapse
Affiliation(s)
- Simon Van Herck
- Institute of Bioengineering, École polytechnique fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland; Department of Pharmaceutics, Ghent University, 9000 Ghent, Belgium
| | - Bing Feng
- Institute of Bioengineering, École polytechnique fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland; Institute of Materials Science & Engineering, EPFL, 1015 Lausanne, Switzerland
| | - Li Tang
- Institute of Bioengineering, École polytechnique fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland; Institute of Materials Science & Engineering, EPFL, 1015 Lausanne, Switzerland.
| |
Collapse
|
14
|
Huang L, Chu Y, Zhang L, Liu X, Hao W, Chen Y, Dai J. High strength pure chitosan hydrogels via double crosslinking strategy. Biomed Mater 2021; 16. [PMID: 34038891 DOI: 10.1088/1748-605x/ac058c] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/26/2021] [Indexed: 11/12/2022]
Abstract
Chitosan (CS) hydrogels have been widely used throughout basic tissue engineering and regenerative medicine research and it is very desirable to develop advanced CS materials with superior mechanical and topographical properties for more extensive applications. Herein, we present the design of a double crosslinking pure CS hydrogel material via the synergic effect of the chemical covalent network, hydrophobic interactions, enhanced intermolecular hydrogen bonding and the formation of the CS crystallite. The resultant pure CS hydrogel possesses increases in strength and toughness by two orders of magnitude (fracture energy ∼7.733 J m-2; maximal compression stress ∼10.81 MPa, elastic modulus ∼1.33 MPa). We utilize1H NMR and FT-IR to prove the success of chemical modification. The results of Raman spectra and WXRD have proved the existence of physical interaction between CS hydrogels and microcrystals, thus explaining the enhancement mechanism of mechanical strength of CS hydrogel. The live and death results also show that MSCs can grow well on CS hydrogels, and the results of CCK-8 indicate low cytotoxicity of CS hydrogels. This CS hydrogel shows great potential applications in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Lei Huang
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, People's Republic of China.,Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| | - Yun Chu
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, People's Republic of China.,Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| | - Lulu Zhang
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, People's Republic of China.,Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| | - Xiru Liu
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| | - Wangping Hao
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| | - Yanyan Chen
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| | - Jianwu Dai
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China.,State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China
| |
Collapse
|
15
|
Abbasi S, Uchida S. Multifunctional Immunoadjuvants for Use in Minimalist Nucleic Acid Vaccines. Pharmaceutics 2021; 13:644. [PMID: 34062771 PMCID: PMC8147386 DOI: 10.3390/pharmaceutics13050644] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/25/2021] [Accepted: 04/27/2021] [Indexed: 12/13/2022] Open
Abstract
Subunit vaccines based on antigen-encoding nucleic acids have shown great promise for antigen-specific immunization against cancer and infectious diseases. Vaccines require immunostimulatory adjuvants to activate the innate immune system and trigger specific adaptive immune responses. However, the incorporation of immunoadjuvants into nonviral nucleic acid delivery systems often results in fairly complex structures that are difficult to mass-produce and characterize. In recent years, minimalist approaches have emerged to reduce the number of components used in vaccines. In these approaches, delivery materials, such as lipids and polymers, and/or pDNA/mRNA are designed to simultaneously possess several functionalities of immunostimulatory adjuvants. Such multifunctional immunoadjuvants encode antigens, encapsulate nucleic acids, and control their pharmacokinetic or cellular fate. Herein, we review a diverse class of multifunctional immunoadjuvants in nucleic acid subunit vaccines and provide a detailed description of their mechanisms of adjuvanticity and induction of specific immune responses.
Collapse
Affiliation(s)
- Saed Abbasi
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Satoshi Uchida
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
- Medical Chemistry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 1-5 Shimogamohangi-cho, Sakyo-ku, Kyoto 606-0823, Japan
| |
Collapse
|
16
|
Pautu V, Lepeltier E, Mellinger A, Riou J, Debuigne A, Jérôme C, Clere N, Passirani C. pH-Responsive Lipid Nanocapsules: A Promising Strategy for Improved Resistant Melanoma Cell Internalization. Cancers (Basel) 2021; 13:2028. [PMID: 33922267 PMCID: PMC8122844 DOI: 10.3390/cancers13092028] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/12/2021] [Accepted: 04/16/2021] [Indexed: 12/12/2022] Open
Abstract
Despite significant advances in melanoma therapy, low response rates and multidrug resistance (MDR) have been described, reducing the anticancer efficacy of the administered molecules. Among the causes to explain these resistances, the decreased intratumoral pH is known to potentiate MDR and to reduce the sensitivity to anticancer molecules. Nanomedicines have been widely exploited as the carriers of MDR reversing molecules. Lipid nanocapsules (LNC) are nanoparticles that have already demonstrated their ability to improve cancer treatment. Here, LNC were modified with novel copolymers that combine N-vinylpyrrolidone (NVP) to impart stealth properties and vinyl imidazole (Vim), providing pH-responsive ability to address classical chemoresistance by improving tumor cell entry. These copolymers could be post-inserted at the LNC surface, leading to the property of going from neutral charge under physiological pH to positive charge under acidic conditions. LNC modified with polymer P5 (C18H37-P(NVP21-co-Vim15)) showed in vitro pH-responsive properties characterized by an enhanced cellular uptake under acidic conditions. Moreover, P5 surface modification led to an increased biological effect by protecting the nanocarrier from opsonization by complement activation. These data suggest that pH-sensitive LNC responds to what is expected from a promising nanocarrier to target metastatic melanoma.
Collapse
Affiliation(s)
- Vincent Pautu
- Micro & Nanomedecines Translationnelles (MINT), University of Angers, Inserm, The National Center for Scientific Research (CNRS), SFR ICAT, F-49000 Angers, France; (V.P.); (E.L.); (A.M.); (J.R.); (N.C.)
- Center for Education and Research on Macromolecules (CERM), Complex and Entangled Systems from Atoms to Materials Research Unit (CESAM-RU), University of Liège, 4000 Liège, Belgium; (A.D.); (C.J.)
| | - Elise Lepeltier
- Micro & Nanomedecines Translationnelles (MINT), University of Angers, Inserm, The National Center for Scientific Research (CNRS), SFR ICAT, F-49000 Angers, France; (V.P.); (E.L.); (A.M.); (J.R.); (N.C.)
| | - Adélie Mellinger
- Micro & Nanomedecines Translationnelles (MINT), University of Angers, Inserm, The National Center for Scientific Research (CNRS), SFR ICAT, F-49000 Angers, France; (V.P.); (E.L.); (A.M.); (J.R.); (N.C.)
| | - Jérémie Riou
- Micro & Nanomedecines Translationnelles (MINT), University of Angers, Inserm, The National Center for Scientific Research (CNRS), SFR ICAT, F-49000 Angers, France; (V.P.); (E.L.); (A.M.); (J.R.); (N.C.)
| | - Antoine Debuigne
- Center for Education and Research on Macromolecules (CERM), Complex and Entangled Systems from Atoms to Materials Research Unit (CESAM-RU), University of Liège, 4000 Liège, Belgium; (A.D.); (C.J.)
| | - Christine Jérôme
- Center for Education and Research on Macromolecules (CERM), Complex and Entangled Systems from Atoms to Materials Research Unit (CESAM-RU), University of Liège, 4000 Liège, Belgium; (A.D.); (C.J.)
| | - Nicolas Clere
- Micro & Nanomedecines Translationnelles (MINT), University of Angers, Inserm, The National Center for Scientific Research (CNRS), SFR ICAT, F-49000 Angers, France; (V.P.); (E.L.); (A.M.); (J.R.); (N.C.)
| | - Catherine Passirani
- Micro & Nanomedecines Translationnelles (MINT), University of Angers, Inserm, The National Center for Scientific Research (CNRS), SFR ICAT, F-49000 Angers, France; (V.P.); (E.L.); (A.M.); (J.R.); (N.C.)
| |
Collapse
|
17
|
Natural and synthetic carbohydrate-based vaccine adjuvants and their mechanisms of action. Nat Rev Chem 2021; 5:197-216. [PMID: 37117529 PMCID: PMC7829660 DOI: 10.1038/s41570-020-00244-3] [Citation(s) in RCA: 143] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/03/2020] [Indexed: 01/31/2023]
Abstract
Modern subunit vaccines based on homogeneous antigens offer more precise targeting and improved safety compared with traditional whole-pathogen vaccines. However, they are also less immunogenic and require an adjuvant to increase the immunogenicity of the antigen and potentiate the immune response. Unfortunately, few adjuvants have sufficient potency and low enough toxicity for clinical use, highlighting the urgent need for new, potent and safe adjuvants. Notably, a number of natural and synthetic carbohydrate structures have been used as adjuvants in clinical trials, and two have recently been approved in human vaccines. However, naturally derived carbohydrate adjuvants are heterogeneous, difficult to obtain and, in some cases, unstable. In addition, their molecular mechanisms of action are generally not fully understood, partly owing to the lack of tools to elucidate their immune-potentiating effects, thus hampering the rational development of optimized adjuvants. To address these challenges, modification of the natural product structure using synthetic chemistry emerges as an attractive approach to develop well-defined, improved carbohydrate-containing adjuvants and chemical probes for mechanistic investigation. This Review describes selected examples of natural and synthetic carbohydrate-based adjuvants and their application in synthetic self-adjuvanting vaccines, while also discussing current understanding of their molecular mechanisms of action.
Collapse
|
18
|
Abstract
Vaccination is one of the main methods for the specific prevention of infectious diseases. The disadvantage of vaccination is the use of pathogens (live or attenuated viruses and bacteria) that can lead to the development of a disease. Recombinant technologies are capable of producing specific DNA or protein molecules that possess antigenic properties and do not cause disease. However, individual antigen molecules are low-immunogenic, and therefore, require conjugation with a compound possessing stronger immunogenic properties. In this study, we examined the immunogenic properties of the new anionic copolymer consisting of glycidyl methacrylate, butyl acrylate, triethylene glycol dimethacrylate, and acrylic acid, in mice. The experimental polymer induced a stronger immunogenic response than aluminum hydroxide. The histological studies have established that immunization both with aluminum hydroxide and the polymer studied does not cause damage to the liver, kidneys, or the spleen. No negative side effects were observed. It has been concluded that the new synthetic anionic polyelectrolyte hydrogel (PHG) has a potential as an adjuvant for vaccine development.
Collapse
|
19
|
Castro F, Pinto ML, Pereira CL, Serre K, Barbosa MA, Vermaelen K, Gärtner F, Gonçalves RM, De Wever O, Oliveira MJ. Chitosan/γ-PGA nanoparticles-based immunotherapy as adjuvant to radiotherapy in breast cancer. Biomaterials 2020; 257:120218. [DOI: 10.1016/j.biomaterials.2020.120218] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/05/2020] [Accepted: 06/22/2020] [Indexed: 02/07/2023]
|
20
|
Zheng Y, Bian L, Zhao H, Liu Y, Lu J, Liu D, Zhang K, Song Y, Luo Y, Jiang C, Chen Y, Zhang Y, Kong W. Respiratory Syncytial Virus F Subunit Vaccine With AS02 Adjuvant Elicits Balanced, Robust Humoral and Cellular Immunity in BALB/c Mice. Front Immunol 2020; 11:526965. [PMID: 33013922 PMCID: PMC7516270 DOI: 10.3389/fimmu.2020.526965] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 08/18/2020] [Indexed: 01/08/2023] Open
Abstract
Respiratory syncytial virus (RSV) is a leading cause of lower respiratory illness, particularly in infants, the elderly, and immunocompromised adults. There is no licensed commercial vaccine against RSV. Importantly, formalin-inactivated RSV vaccines mediate enhanced respiratory disease. RSV fusion (F) protein with pre-fusion conformation is a promising candidate subunit vaccine. However, some problems remain to be solved, such as low immunogenicity and humoral immunity bias. Adjuvants can effectively enhance and adjust vaccine immune responses. In this study, we formulated pre-fusion RSV-F protein with the adjuvants, Alhydrogel, MF59, AS03, AS02, and glycol chitosan (GCS). We then conducted head-to-head comparisons of vaccine-induced immune responses in BALB/c mice. All adjuvanted vaccines enhanced antigen-specific and neutralizing antibody titers and viral clearance and gave an order of adjuvant activity: AS02 > AS03, MF59 > GCS, and Alhydrogel. Among them, AS02 elicited the highest antibody expression, which persisted until week 18. Moreover, AS02 significantly enhanced Th1 type immune response in immunized mice. Mice in the AS02 group also showed faster recovery from viral attacks in challenge tests. Further transcriptome analysis revealed that AS02 regulates immune balance by activating TLR-4 and promotes Th1-type immune responses. These results suggest that AS02 may be an excellent candidate adjuvant for RSV-F subunit vaccines. This study also provides valuable information regarding the effect of other adjuvants on immune responses of RSV-F subunit vaccines.
Collapse
Affiliation(s)
- Yu Zheng
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Lijun Bian
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Huiting Zhao
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Yulan Liu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Jingcai Lu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China.,R&D Center, Changchun BCHT Biotechnology Co., Changchun, China
| | - Dawei Liu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China.,R&D Center, Changchun BCHT Biotechnology Co., Changchun, China
| | - Ke Zhang
- The Key and Characteristic Laboratory of Modern Pathogen Biology, Department of Parasitology, Basic Medical College, Guizhou Medical University, Guiyang, China
| | - Yueshuang Song
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China.,R&D Center, Changchun BCHT Biotechnology Co., Changchun, China
| | - Yusi Luo
- Intensive Care Unit, Department of Emergency, Guizhou Medical University Affiliated Hospital, Guiyang, China
| | - Chunlai Jiang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| | - Yan Chen
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| | - Yong Zhang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| | - Wei Kong
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| |
Collapse
|
21
|
Maeyama JI, Kurata-Iesato Y, Isaka M, Komiya T, Sakurai S. Induction of antibody responses in mice immunized intranasally with Type I interferon as adjuvant and synergistic effect of chitosan. Microbiol Immunol 2020; 64:610-619. [PMID: 32662896 DOI: 10.1111/1348-0421.12832] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 06/24/2020] [Accepted: 07/09/2020] [Indexed: 02/05/2023]
Abstract
Type I IFNs are a range of host-derived molecules with adjuvant potential; they have been used for many years in the treatment of cancer and viral hepatitis. Therefore, the safety of IFNs for human use has been established. In this study, we evaluated the mucosal adjuvanticity of IFN-β administered intranasally to mice with diphtheria toxoid, and suggested a method to improve its adjuvanticity. When IFN-β alone was used as a mucosal adjuvant, no clear results were obtained. However, simultaneous administration of IFN-β and chitosan resulted in an enhancement of the specific serum immunoglobulin G (IgG) and IgA antibody responses, the mucosal IgA antibody response, and antitoxin titers. Furthermore, the intranasal administration of IFN-α alone resulted in a greater increase in antibody titer than IFN-β, and a synergistic effect with chitosan was also observed. These findings suggest that intranasal administration of chitosan and Type I IFNs may display an effective synergistic mucosal adjuvant activity.
Collapse
Affiliation(s)
- Jun-Ichi Maeyama
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yuko Kurata-Iesato
- Department of Pharmaceutical Quality Assurance, Toray Industries Inc., Mishima, Japan
| | - Masanori Isaka
- Department of Microbiology, Nagoya City University Medical School, Nagoya, Japan
| | - Takako Komiya
- Department of Bacterial Pathogenesis and Infection, National Institute of Infectious Diseases, Tokyo, Japan
| | - Shingou Sakurai
- Faculty of Pharmacy, Tokyo University of Science, Tokyo, Japan
| |
Collapse
|
22
|
Simón-Vázquez R, Peleteiro M, González-Fernández Á. Polymeric nanostructure vaccines: applications and challenges. Expert Opin Drug Deliv 2020; 17:1007-1023. [PMID: 32476491 DOI: 10.1080/17425247.2020.1776259] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION The use of biocompatible polymers, from natural or synthetic sources, opened the door for a new era in vaccine research. These polymers offer the possibility to develop nanostructured antigen carriers that can be easily internalized by antigen-presenting cells, due to their nanometric size. Besides, the incorporation of an adjuvant allows increasing and modulating the immune response for both, polymers with or without self-adjuvant properties. AREAS COVERED The historical background and the state-of-the-art in the use of polymers as antigen carriers are addressed in the first part of this review. Then, an overview of the immunology of vaccination is provided. Finally, the main advances in the field, based on the prototypes that are licensed or undergoing clinical trials, but also the challenges that limit the translation of many polymer-based nanostructure vaccines with promising preclinical results, are discussed. EXPERT OPINION Polymeric nanostructured vaccines have a great potential in modern vaccinology. However, the translation into the market is hampered due to several limitations. Studies on correlates of protection to provide suitable biomarkers, new and better methods of synthesis to produce more reproducible nanovaccines, a deeper knowledge in the immune system and in the physiopathology of the infectious diseases will surely improve and boost the field in the next years.
Collapse
Affiliation(s)
- Rosana Simón-Vázquez
- Immunology Group, Centro de Investigaciones Biomédicas, CINBIO, Universidade de Vigo, Campus Universitario Lagoas Marcosende , Vigo, Spain.,Instituto De Investigación Sanitaria Galicia Sur (IIS-GS), Hospital Álvaro Cunqueiro, Estrada Clara Campoamor , Vigo, Pontevedra, Spain
| | - Mercedes Peleteiro
- Instituto De Investigación Sanitaria Galicia Sur (IIS-GS), Hospital Álvaro Cunqueiro, Estrada Clara Campoamor , Vigo, Pontevedra, Spain.,Flow Cytometry Core Facility, CINBIO, Universidade de Vigo, Campus Universitario Lagoas Marcosende, Vigo, Spain
| | - África González-Fernández
- Immunology Group, Centro de Investigaciones Biomédicas, CINBIO, Universidade de Vigo, Campus Universitario Lagoas Marcosende , Vigo, Spain.,Instituto De Investigación Sanitaria Galicia Sur (IIS-GS), Hospital Álvaro Cunqueiro, Estrada Clara Campoamor , Vigo, Pontevedra, Spain
| |
Collapse
|
23
|
Marques C, Som C, Schmutz M, Borges O, Borchard G. How the Lack of Chitosan Characterization Precludes Implementation of the Safe-by-Design Concept. Front Bioeng Biotechnol 2020; 8:165. [PMID: 32211394 PMCID: PMC7077258 DOI: 10.3389/fbioe.2020.00165] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 02/18/2020] [Indexed: 01/01/2023] Open
Abstract
Efficacy and safety of nanomedicines based on polymeric (bio)materials will benefit from a rational implementation of a Safe-by-Design (SbD) approach throughout their development. In order to achieve this goal, however, a standardization of preparation and characterization methods and their accurate reporting is needed. Focusing on the example of chitosan, a biopolymer derived from chitin and frequently used in drug and vaccine delivery vector preparation, this review discusses the challenges still to be met and overcome prior to a successful implementation of the SbD approach to the preparation of chitosan-based protein drug delivery systems.
Collapse
Affiliation(s)
- Cíntia Marques
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland.,Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Claudia Som
- Empa, Swiss Federal Laboratories for Materials Science and Technology, Technology and Society Laboratory, St. Gallen, Switzerland
| | - Mélanie Schmutz
- Empa, Swiss Federal Laboratories for Materials Science and Technology, Technology and Society Laboratory, St. Gallen, Switzerland
| | - Olga Borges
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.,Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Gerrit Borchard
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| |
Collapse
|
24
|
AbdelAllah NH, Gaber Y, Rashed ME, Azmy AF, Abou-Taleb HA, AbdelGhani S. Alginate-coated chitosan nanoparticles act as effective adjuvant for hepatitis A vaccine in mice. Int J Biol Macromol 2020; 152:904-912. [PMID: 32114177 DOI: 10.1016/j.ijbiomac.2020.02.287] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/24/2020] [Accepted: 02/25/2020] [Indexed: 01/28/2023]
Abstract
The numerous recent hepatitis A outbreaks emphasize the need for vaccination; despite the effectiveness of the current ones, developments are needed to overcome its high cost plus some immune response limitations. Our study aims to evaluate the use of chitosan and alginate-coated chitosan nanoparticles as an adjuvant/carrier for the hepatitis A vaccine (HAV) against the traditional adjuvant alum. Immune responses towards (HAV-Al) with alum, (HAV-Ch) with chitosan, and (HAV-aCNP) with alginate-coated chitosan nanoparticles, were assessed in mice. HAV-aCNP significantly improved the immunogenicity by increasing the seroconversion rate (100%), the hepatitis A antibodies level, and the splenocytes proliferation. Thus, the HAV-aCNP adjuvant was superior to other classes in IFN-γ and IL-10 development. Meanwhile, the solution formula of HAV with chitosan showed comparable humoral and cellular immune responses to alum-adjuvanted suspension with a balanced Th1/Th2 immune pathway. The current study showed the potential of alginate-coated chitosan nanoparticles as an effective carrier for HAV. Consequently, this would impact the cost of HAV production positively.
Collapse
Affiliation(s)
- Nourhan H AbdelAllah
- Department of Microbiology and Immunology, Faculty of Pharmacy, Beni-Suef University, 62511 Beni-Suef, Egypt; Viral Control Unit, National Organization for Research and Control of Biologicals (NORCB), Cairo 12654, Egypt
| | - Yasser Gaber
- Department of Microbiology and Immunology, Faculty of Pharmacy, Beni-Suef University, 62511 Beni-Suef, Egypt; Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Mutah University, Al-karak 61710, Jordan
| | - Mohamed E Rashed
- Microbiology Department, National Organization for Research and Control of Biologicals (NORCB), Cairo 12654, Egypt
| | - Ahmed F Azmy
- Department of Microbiology and Immunology, Faculty of Pharmacy, Beni-Suef University, 62511 Beni-Suef, Egypt
| | - Heba A Abou-Taleb
- Pharmaceutics and Industrial Pharmacy Department, Faculty of Pharmacy, Nahda University (NUB), Beni-Suef, Egypt
| | - Sameh AbdelGhani
- Department of Microbiology and Immunology, Faculty of Pharmacy, Beni-Suef University, 62511 Beni-Suef, Egypt; Department of Pathology and Medical Laboratory, University of Louisville, KY 40202, USA.
| |
Collapse
|
25
|
|
26
|
Meng F, Kwon S, Wang J, Yeo Y. Immunoactive drug carriers in cancer therapy. BIOMATERIALS FOR CANCER THERAPEUTICS 2020:53-94. [DOI: 10.1016/b978-0-08-102983-1.00003-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
27
|
Chen YH, Lai KY, Chiu YH, Wu YW, Shiau AL, Chen MC. Implantable microneedles with an immune-boosting function for effective intradermal influenza vaccination. Acta Biomater 2019; 97:230-238. [PMID: 31362139 DOI: 10.1016/j.actbio.2019.07.048] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 07/21/2019] [Accepted: 07/25/2019] [Indexed: 12/11/2022]
Abstract
This study details effective influenza vaccination via sustained intradermal (ID) release of vaccines using implantable and patch-free chitosan microneedles (MNs). The microneedle (MN) patch is composed of vaccine-loaded chitosan MNs with a dissolvable supporting array that gives extra length for complete insertion of MNs and is dissolved within the skin during insertion. Chitosan MNs can be quickly and entirely implanted into the dermis to function as a depot and an immune-boosting agent for the extended release of vaccines and simultaneous activation of the immune system. We found the influenza virus-specific antibody levels induced by chitosan MN vaccination were significantly higher than those elicited by intramuscular (IM) immunization with influenza vaccine alone. The MN induced immune-enhancing effect was obvious 4 week after the vaccination and lasted for at least 16 weeks. Most importantly, MN-immunized mice were completely protected from H1N1 viral challenge without major weight loss, whereas mice receiving IM injection at the same dose had a mortality rate of 60% and experienced notable weight loss after challenge. Our results suggest that the chitosan MNs cannot only be a viable tool for precise ID vaccine delivery but also exert strong adjuvanticity to enhance vaccine potency and induce protective immunity against influenza virus infections. STATEMENT OF SIGNIFICANCE: There is an urgent need for generating a new vaccination strategy to address the threat of global pandemic influenza. This study presents implantable chitosan microneedles (MNs) with immune-boosting function for effective influenza vaccination. We demonstrate that the chitosan MN can not only be an efficient tool for sustained intradermal delivery but also serve as an immunological adjuvant to boost vaccine efficacy. Continuous antigen exposure and immune stimulation provided by the implanted MNs may enhance the immunogenicity of influenza vaccines and evoke long-lasting immune responses to completely protect mice from lethal influenza challenge. The proposed MN system has great potential to be used as a new adjuvanted vaccine formulation and make influenza vaccination more effective and more accessible.
Collapse
|
28
|
Yousefi S, Abbassi-Daloii T, Tahmoorespur M, Sekhavati MH. Nanoparticle or conventional adjuvants: which one improves immune response against Brucellosis? IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2019; 22:360-366. [PMID: 31168339 PMCID: PMC6535204 DOI: 10.22038/ijbms.2019.31748.7642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Objective(s): Brucellosis is a common infectious disease among animals and humans. While subunit vaccines could be used as an efficient strategy against pathogens, they usually seem to be less immunogenic than live or killed vaccines. However, the use of a suitable adjuvant accompanied by subunit vaccines can be a good alternative to enhance the immune response. Materials and Methods: To find a proper adjuvant against Brucellosis, the immune response of induced mice by Aluminum Hydroxide (AH), Incomplete Freund (IFA), and Chitosan Nanoparticle (CS) adjuvants in individuals and in combination with CS were assessed. Results: Immunization with CS stimulated higher interferon gamma (IFN-γ) immunity, while there were no significant differences between rOMP25 (IFA), rOMP25 (AH), rOMP25 (AH-CS) and rOMP25 (IFA-CS) recombinant proteins. Tumor necrosis factor alpha (TNF-α) analysis revealed there were no significant differencesbetween immunized groups and the positive control group, except for the treatment formulated in single IFA. Furthermore, unlike IFN-γ, there was a reverse interleukin-4 (IL-4) immune response trend for treatments, as rOMP25 (CS) displayed the lowest response. rOMP25 (CS) induced higher titer of total antibody than the other ones. Although the recombinant proteins emulsified in different adjuvants induced similar titer of IgG1 antibody, the ones that were formulated in CS, IFA and IFA-CS showed a higher titer of IgG2a. The cell proliferation assay demonstrating the antigen-specific cell proliferative response could be promoted after immunization with CS. Conclusion: CS whether single or in combination with IF adjuvants has potential to improve Th1-Th2 responses.
Collapse
Affiliation(s)
- Soheil Yousefi
- Department of Animal Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | | | | | | |
Collapse
|
29
|
Cohen E, Merzendorfer H. Chitin/Chitosan: Versatile Ecological, Industrial, and Biomedical Applications. EXTRACELLULAR SUGAR-BASED BIOPOLYMERS MATRICES 2019; 12. [PMCID: PMC7115017 DOI: 10.1007/978-3-030-12919-4_14] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Chitin is a linear polysaccharide of N-acetylglucosamine, which is highly abundant in nature and mainly produced by marine crustaceans. Chitosan is obtained by hydrolytic deacetylation. Both polysaccharides are renewable resources, simply and cost-effectively extracted from waste material of fish industry, mainly crab and shrimp shells. Research over the past five decades has revealed that chitosan, in particular, possesses unique and useful characteristics such as chemical versatility, polyelectrolyte properties, gel- and film-forming ability, high adsorption capacity, antimicrobial and antioxidative properties, low toxicity, and biocompatibility and biodegradability features. A plethora of chemical chitosan derivatives have been synthesized yielding improved materials with suggested or effective applications in water treatment, biosensor engineering, agriculture, food processing and storage, textile additives, cosmetics fabrication, and in veterinary and human medicine. The number of studies in this research field has exploded particularly during the last two decades. Here, we review recent advances in utilizing chitosan and chitosan derivatives in different technical, agricultural, and biomedical fields.
Collapse
Affiliation(s)
- Ephraim Cohen
- Department of Entomology, The Robert H. Smith Faculty of Agriculture Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Hans Merzendorfer
- School of Science and Technology, Institute of Biology – Molecular Biology, University of Siegen, Siegen, Germany
| |
Collapse
|
30
|
Markushin SG, Akopova II, Blagodatskikh IV, Kulikov SN, Bezrodnykh EA, Muranov AV, Yamskov IA, Tikhonov VE. Effect of Molecular Weight and Degree of Acetylation on Adjuvantive Properties of Chitosan Derivatives. APPL BIOCHEM MICRO+ 2018. [DOI: 10.1134/s0003683818050149] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
31
|
Tandberg J, Lagos L, Ropstad E, Smistad G, Hiorth M, Winther-Larsen HC. The Use of Chitosan-Coated Membrane Vesicles for Immunization Against Salmonid Rickettsial Septicemia in an Adult Zebrafish Model. Zebrafish 2018; 15:372-381. [PMID: 29957152 DOI: 10.1089/zeb.2017.1556] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The introduction of fish vaccination has had a tremendous impact on the aquaculture industry by providing an important measurement in regard to disease control. Infectious diseases caused by intracellular pathogens do, however, remain an unsolved problem for the industry. This is in many cases directly connected to the inability of vaccines to evoke a cellular immunity needed for long-term protection. Thus, there is a need for new and improved vaccines and adjuvants able to induce a strong humoral and cellular immune response. We have previously shown that membrane vesicles (MVs) from the intracellular fish pathogen Piscirickettsia salmonis are able to induce a protective response in adult zebrafish, but the incorporation of an adjuvant has not been evaluated. In this study, we report the use of chitosan as an adjuvant in combination with the P. salmonis-derived MVs for improved immunization against P. salmonis. Both free chitosan and chitosan-coated MVs (cMVs) were injected into adult zebrafish and their efficacy evaluated. The cMVs provided a significant protection (p < 0.05), while a small but nonsignificant reduction in mortalities was registered for fish injected with free chitosan. Both free chitosan and the cMVs were shown to induce an increased immune gene expression of CD 4, CD 8, MHC I, Mpeg1.1, TNFα, IL-1β, IL-10, and IL-6, but to a higher degree in the cMV group. Taken together, the results indicate a potential use of chitosan-coated MVs for vaccination, and that zebrafish is a promising model for aquaculture-relevant studies.
Collapse
Affiliation(s)
- Julia Tandberg
- 1 Department of Pharmaceutical Biosciences, Faculty of Mathematics and Natural Science, School of Pharmacy, University of Oslo , Oslo, Norway
| | - Leidy Lagos
- 2 Department of Animal and Aquacultural Sciences, Norwegian University of Life Sciences , Ås, Norway
| | - Erik Ropstad
- 3 Department of Production Animal Clinical Sciences, Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences , Oslo, Norway
| | - Gro Smistad
- 4 Department of Pharmacy, School of Pharmacy, University of Oslo , Oslo, Norway
| | - Marianne Hiorth
- 4 Department of Pharmacy, School of Pharmacy, University of Oslo , Oslo, Norway
| | - Hanne C Winther-Larsen
- 1 Department of Pharmaceutical Biosciences, Faculty of Mathematics and Natural Science, School of Pharmacy, University of Oslo , Oslo, Norway
| |
Collapse
|
32
|
Chen MC, Lai KY, Ling MH, Lin CW. Enhancing immunogenicity of antigens through sustained intradermal delivery using chitosan microneedles with a patch-dissolvable design. Acta Biomater 2018; 65:66-75. [PMID: 29109028 DOI: 10.1016/j.actbio.2017.11.004] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 11/01/2017] [Accepted: 11/02/2017] [Indexed: 12/31/2022]
Abstract
Reducing the dosage required for vaccination is highly desirable, particularly in cases of epidemic emergencies. This study evaluated the potential of a chitosan microneedle (MN) system with a patch-dissolvable design for low-dose immunization. This system comprises antigen-loaded chitosan MNs and a hydrophilic polyvinyl alcohol/polyvinyl pyrrolidone supporting array patch, which provides extra strength to achieve complete MN insertion and then quickly dissolves in the skin to reduce patch-induced skin irritation. After insertion, MNs could be directly implanted in the dermal layer as an intradermal (ID) depot to allow a sustained release of the model antigen ovalbumin (OVA) for up to 28 days. We found that rats immunized with MNs containing low-dose OVA (approximately 200 μg) had persistently high antibody levels for 18 weeks, which were significantly higher than those observed after an intramuscular injection of full-dose OVA (approximately 500 μg), demonstrating at least 2.5-fold dose sparing. Moreover, OVA-encapsulated chitosan MNs had superior immunogenicity to OVA plus chitosan solution, indicating that MN-based delivery and prolonged skin exposure can further enhance chitosan's adjuvanticity. Therefore, this patch-dissolvable MN system offers a needle-free, accurate, and reliable ID delivery of antigens and has potential as a sustained ID delivery device to improve vaccine efficacy and facilitate dose sparing with existing vaccines. STATEMENT OF SIGNIFICANCE This study developed implantable chitosan microneedles (MNs) with a patch-dissolvable design for the sustained intradermal (ID) delivery of antigens and demonstrated their antigen dose-sparing potential. We found that rats immunized with chitosan MNs containing low-dose OVA had persistently high antibody levels for 18 weeks, which were significantly higher than those observed after an intramuscular injection of full-dose OVA, demonstrating at least 2.5-fold dose sparing. Our results indicate that chitosan MNs can not only serve as an efficient vaccine delivery system but also exert their promising adjuvant activity by forming an ID depot for prolonged antigen exposure and activating dendritic cells for promoting immune responses.
Collapse
|
33
|
Jesus S, Soares E, Borchard G, Borges O. Poly-ϵ-caprolactone/chitosan nanoparticles provide strong adjuvant effect for hepatitis B antigen. Nanomedicine (Lond) 2017; 12:2335-2348. [DOI: 10.2217/nnm-2017-0138] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Aim: This work aims to investigate the adjuvant effect of poly-ϵ-caprolactone/chitosan nanoparticles (NPs) for hepatitis B surface antigen (HBsAg) and the plasmid DNA encoding HBsAg (pRC/CMV-HBs). Methods: Both antigens were adsorbed onto preformed NPs. Vaccination studies were performed in C57BL/6 mice. Transfection efficiency was investigated in A549 cell line. Results: HBsAg-adsorbed NPs generated strong anti-HBsAg IgG titers, mainly of IgG1 isotype, and induced antigen-specific IFN-γ and IL-17 secretion by spleen cells. The addition of pRC/CMV-HBs to the HBsAg-adsorbed NPs inhibited IL-17 secretion but had minor effect on IFN-γ levels. Lastly, pRC/CMV-HBs-loaded NPs generated a weak serum antibody response. Conclusion: Poly-ϵ-caprolactone/chitosan NPs provide a strong humoral adjuvant effect for HBsAg and induce a Th1/Th17-mediated cellular immune responses worth explore for hepatitis B virus vaccination.
Collapse
Affiliation(s)
- Sandra Jesus
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Neuroscience & Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Edna Soares
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Neuroscience & Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Gerrit Borchard
- Department of Pharmaceutical Sciences, Faculty of Science, University of Geneva, CH-1211 Geneva, Switzerland
- The Geneva-Lausanne School of Pharmacy, University of Geneva, University of Lausanne, CH-1211 Geneva, Switzerland
| | - Olga Borges
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Neuroscience & Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| |
Collapse
|
34
|
Bernocchi B, Carpentier R, Betbeder D. Nasal nanovaccines. Int J Pharm 2017; 530:128-138. [PMID: 28698066 DOI: 10.1016/j.ijpharm.2017.07.012] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 07/03/2017] [Accepted: 07/04/2017] [Indexed: 01/08/2023]
Abstract
Nasal administration of vaccines is convenient for the potential stimulation of mucosal and systemic immune protection. Moreover the easy accessibility of the intranasal route renders it optimal for pandemic vaccination. Nanoparticles have been identified as ideal delivery systems and adjuvants for vaccine application. Heterogeneous protocols have been used for animal studies. This complicates the understanding of the formulation influence on the immune response and the comparison of the different nanoparticles approaches developed. Moreover anatomical and immunological differences between rodents and humans provide an additional hurdle in the rational development of nasal nanovaccines. This review will give a comprehensive expertise of the state of the art in nasal nanovaccines in animals and humans focusing on the nanomaterial used.
Collapse
Affiliation(s)
- B Bernocchi
- Inserm, LIRIC-UMR 995, F-59000 Lille, France; Université de Lille, LIRIC-UMR 995, F-59000 Lille, France; CHRU de Lille, LIRIC-UMR 995, F-59000 Lille, France
| | - R Carpentier
- Inserm, LIRIC-UMR 995, F-59000 Lille, France; Université de Lille, LIRIC-UMR 995, F-59000 Lille, France; CHRU de Lille, LIRIC-UMR 995, F-59000 Lille, France.
| | - D Betbeder
- Inserm, LIRIC-UMR 995, F-59000 Lille, France; Université de Lille, LIRIC-UMR 995, F-59000 Lille, France; CHRU de Lille, LIRIC-UMR 995, F-59000 Lille, France; University of Artois, 62000 Arras, France
| |
Collapse
|
35
|
Yang Z, Xu M, Jia Z, Zhang Y, Wang L, Zhang H, Wang J, Song M, Zhao Y, Wu Z, Zhao L, Yin Z, Hong Z. A novel antigen delivery system induces strong humoral and CTL immune responses. Biomaterials 2017; 134:51-63. [DOI: 10.1016/j.biomaterials.2017.04.035] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 04/17/2017] [Accepted: 04/19/2017] [Indexed: 12/17/2022]
|
36
|
Babu A, Ramesh R. Multifaceted Applications of Chitosan in Cancer Drug Delivery and Therapy. Mar Drugs 2017; 15:E96. [PMID: 28346381 PMCID: PMC5408242 DOI: 10.3390/md15040096] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 03/17/2017] [Accepted: 03/20/2017] [Indexed: 12/15/2022] Open
Abstract
Chitosan is a versatile polysaccharide of biological origin. Due to the biocompatible and biodegradable nature of chitosan, it is intensively utilized in biomedical applications in scaffold engineering as an absorption enhancer, and for bioactive and controlled drug release. In cancer therapy, chitosan has multifaceted applications, such as assisting in gene delivery and chemotherapeutic delivery, and as an immunoadjuvant for vaccines. The present review highlights the recent applications of chitosan and chitosan derivatives in cancer therapy.
Collapse
Affiliation(s)
- Anish Babu
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| | - Rajagopal Ramesh
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
- Graduate Program in Biomedical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| |
Collapse
|
37
|
Tao W, Fu T, He Z, Hu R, Jia L, Hong Y. Evaluation of Immunostimulatory Effects of N-(2-Hydroxy) Propyl-3-Trimethylammonium Chitosan Chloride for Improving Live Attenuated Hepatitis A Virus Vaccine Efficacy. Viral Immunol 2016; 30:120-126. [PMID: 27918250 DOI: 10.1089/vim.2016.0099] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
This study was to evaluate the immunostimulatory effects of N-(2-hydroxy) propyl-3-trimethylammonium chitosan chloride (HTCC) as an adjuvant for improving a commercial live attenuated hepatitis A virus (HAV) vaccine efficacy in mice. Mice in the experimental group were intraperitoneally immunized with a solution of HTCC and live attenuated HAV vaccine. And for those injected with sterile water, HTCC or live attenuated HAV vaccine were treated as mock group, negative group, and positive group in turn. The serum HAV-specific IgG titers and the ratios of the serum HAV-specific IgG2a/IgG1 in the experimental group were significantly increased (p = 0.00042 and p = 0.040, respectively). Splenocyte proliferation stimulation index in experimental group was higher than positive group (p = 0.021), and significantly higher than mock group and negative group (p = 0.0078 and p = 0.0050, respectively). The percentages of CD4+ T lymphocytes in the peripheral blood in experimental group were significantly higher than positive group, negative group, and mock group (p = 0.012, p = 0.012, and p = 0.045, respectively). Compared to the other three groups, experimental group showed a slightly higher ratio of CD4+/CD8+, but there were no significant differences (p > 0.05). In the percentages of CD8+ T lymphocytes, there were no significant differences among the four groups (p > 0.05). HTCC can enhance live attenuated HAV vaccine to generate stronger humoral responses and induce a Th1-biased immune response, as well as IgG2a class switching, compared with the live attenuated HAV vaccine alone. This study validated an important concept for further development of a safe and potent vaccine adjuvant.
Collapse
Affiliation(s)
- Wei Tao
- Institute of Bioengineering , Zhejiang Academy of Medical Sciences, Hangzhou, China
| | - Ting Fu
- Institute of Bioengineering , Zhejiang Academy of Medical Sciences, Hangzhou, China
| | - Zhuojing He
- Institute of Bioengineering , Zhejiang Academy of Medical Sciences, Hangzhou, China
| | - Ruxi Hu
- Institute of Bioengineering , Zhejiang Academy of Medical Sciences, Hangzhou, China
| | - Lan Jia
- Institute of Bioengineering , Zhejiang Academy of Medical Sciences, Hangzhou, China
| | - Yan Hong
- Institute of Bioengineering , Zhejiang Academy of Medical Sciences, Hangzhou, China
| |
Collapse
|
38
|
Philibert T, Lee BH, Fabien N. Current Status and New Perspectives on Chitin and Chitosan as Functional Biopolymers. Appl Biochem Biotechnol 2016; 181:1314-1337. [PMID: 27787767 DOI: 10.1007/s12010-016-2286-2] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 10/10/2016] [Indexed: 11/24/2022]
Abstract
The natural biopolymer chitin and its deacetylated product chitosan are found abundantly in nature as structural building blocks and are used in all sectors of human activities like materials science, nutrition, health care, and energy. Far from being fully recognized, these polymers are able to open opportunities for completely novel applications due to their exceptional properties which an economic value is intrinsically entrapped. On a commercial scale, chitosan is mainly obtained from crustacean shells rather than from the fungal and insect sources. Significant efforts have been devoted to commercialize chitosan extracted from fungal and insect sources to completely replace crustacean-derived chitosan. However, the traditional chitin extraction processes are laden with many disadvantages. The present review discusses the potential bioextraction of chitosan from fungal, insect, and crustacean as well as its superior physico-chemical properties. The different aspects of fungal, insects, and crustacean chitosan extraction methods and various parameters having an effect on the yield of chitin and chitosan are discussed in detail. In addition, this review also deals with essential attributes of chitosan for high value-added applications in different fields and highlighted new perspectives on the production of chitin and deacetylated chitosan from different sources with the concomitant reduction of the environmental impact.
Collapse
Affiliation(s)
- Tuyishime Philibert
- School of Biotechnology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, China
| | - Byong H Lee
- School of Biotechnology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, China. .,Department of Food Science and Biotechnology, Kangwon National University, Chuncheon, 24341, South Korea. .,Department of Microbiology/Immunology, McGill University, Montreal, QC, H9X3V9, Canada.
| | - Nsanzabera Fabien
- School of Biotechnology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, China
| |
Collapse
|
39
|
Mathews PD, Mertins O. Dispersion of chitosan in liquid crystalline lamellar phase: Production of biofriendly hydrogel of nano cubic topology. Carbohydr Polym 2016; 157:850-857. [PMID: 27987999 DOI: 10.1016/j.carbpol.2016.10.071] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 09/28/2016] [Accepted: 10/23/2016] [Indexed: 02/04/2023]
Abstract
Bicontinuous cubic phases were produced with introduction of chitosan in phospholipid/water hydrogel, providing composites of defined molecular organization. The ratio of lipid/water was constant and swelling of lipids bilayer is delimited by incorporation of polymer molecules into the structure. By means of synchrotron small angle X-ray scattering we identified topologies of coexisting cubic phases. The expected liquid crystalline Lα lamellar phase was suppressed by 0.2wt% chitosan leading to formation of diamond Pn3m and gyroid Ia3d cubic topology, with close lattice distances. An increment to 0.4wt% chitosan caused large increase in Pn3m lattice distance. However a higher 0.6wt% evolved this phase to a newly acquired primitive Im3m topology. The structuring process of the three-dimensional complex network is principally governed by demands of chitosan physical requirements over lipids bilayers interfacial curvature. The composite hydrogel of specific topologies presents reduced time release of gallic acid and may find application as new material for time-sustained delivery of bioactive compounds.
Collapse
Affiliation(s)
- Patrick D Mathews
- Department of Animal Biology, Institute of Biology, State University of Campinas, 13083-970 Campinas, Brazil.
| | - Omar Mertins
- Department of Biophysics, Paulista School of Medicine, Federal University of Sao Paulo, 04023-062 Sao Paulo, Brazil.
| |
Collapse
|
40
|
Veazey RS, Siddiqui A, Klein K, Buffa V, Fischetti L, Doyle-Meyers L, King DF, Tregoning JS, Shattock RJ. Evaluation of mucosal adjuvants and immunization routes for the induction of systemic and mucosal humoral immune responses in macaques. Hum Vaccin Immunother 2016; 11:2913-22. [PMID: 26697975 DOI: 10.1080/21645515.2015.1070998] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Delivering vaccine antigens to mucosal surfaces is potentially very attractive, especially as protection from mucosal infections may be mediated by local immune responses. However, to date mucosal immunization has had limited successes, with issues of both safety and poor immunogenicity. One approach to improve immunogenicity is to develop adjuvants that are effective and safe at mucosal surfaces. Differences in immune responses between mice and men have overstated the value of some experimental adjuvants which have subsequently performed poorly in the clinic. Due to their closer similarity, non-human primates can provide a more accurate picture of adjuvant performance. In this study we immunised rhesus macaques (Macaca mulatta) using a unique matrix experimental design that maximised the number of adjuvants screened while reducing the animal usage. Macaques were immunised by the intranasal, sublingual and intrarectal routes with the model protein antigens keyhole limpet haemocyanin (KLH), β-galactosidase (β-Gal) and ovalbumin (OVA) in combination with the experimental adjuvants Poly(I:C), Pam3CSK4, chitosan, Thymic Stromal Lymphopoietin (TSLP), MPLA and R848 (Resiquimod). Of the routes used, only intranasal immunization with KLH and R848 induced a detectable antibody response. When compared to intramuscular immunization, intranasal administration gave slightly lower levels of antigen specific antibody in the plasma, but enhanced local responses. Following intranasal delivery of R848, we observed a mildly inflammatory response, but no difference to the control. From this we conclude that R848 is able to boost antibody responses to mucosally delivered antigen, without causing excess local inflammation.
Collapse
Affiliation(s)
- Ronald S Veazey
- a Tulane National Primate Research Center; Tulane University School of Medicine ; Covington , LA USA
| | - Asna Siddiqui
- b Mucosal Infection & Immunity Group; Section of Virology; Imperial College London; St. Mary's Campus ; London , UK
| | - Katja Klein
- b Mucosal Infection & Immunity Group; Section of Virology; Imperial College London; St. Mary's Campus ; London , UK.,c Present affiliation: University of Western Ontario ; Ontario , Canada
| | - Viviana Buffa
- b Mucosal Infection & Immunity Group; Section of Virology; Imperial College London; St. Mary's Campus ; London , UK
| | - Lucia Fischetti
- b Mucosal Infection & Immunity Group; Section of Virology; Imperial College London; St. Mary's Campus ; London , UK
| | - Lara Doyle-Meyers
- a Tulane National Primate Research Center; Tulane University School of Medicine ; Covington , LA USA
| | - Deborah F King
- b Mucosal Infection & Immunity Group; Section of Virology; Imperial College London; St. Mary's Campus ; London , UK.,d Present affiliation: IAVI Human Immunology Lab; Chelsea and Westminster; Imperial College London ; London , UK
| | - John S Tregoning
- b Mucosal Infection & Immunity Group; Section of Virology; Imperial College London; St. Mary's Campus ; London , UK
| | - Robin J Shattock
- b Mucosal Infection & Immunity Group; Section of Virology; Imperial College London; St. Mary's Campus ; London , UK
| |
Collapse
|
41
|
Chitosan and Sodium Alginate Combinations Are Alternative, Efficient, and Safe Natural Adjuvant Systems for Hepatitis B Vaccine in Mouse Model. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:7659684. [PMID: 27493674 PMCID: PMC4963576 DOI: 10.1155/2016/7659684] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 06/09/2016] [Indexed: 02/06/2023]
Abstract
Hepatitis B viral (HBV) infections represent major public health problem and are an occupational hazard for healthcare workers. Current alum-adjuvanted HBV vaccine is the most effective measure to prevent HBV infection. However, the vaccine has some limitations including poor response in some vaccinee and being a frost-sensitive suspension. The goal of our study was to use an alternative natural adjuvant system strongly immunogenic allowing for a reduction in dose and cost. We tested HBV surface antigen (HBsAg) adjuvanted with chitosan (Ch) and sodium alginate (S), both natural adjuvants, either alone or combined with alum in mouse model. Mice groups were immunized subcutaneously with HBsAg adjuvanted with Ch or S, or triple adjuvant formula with alum (Al), Ch, and S, or double formulations with AlCh or AlS. These were compared to control groups immunized with current vaccine formula or unadjuvanted HBsAg. We evaluated the rate of seroconversion, serum HBsAg antibody, IL-4, and IFN-γ levels. The results showed that the solution formula with Ch or S exhibited comparable immunogenic responses to Al-adjuvanted suspension. The AlChS gave significantly higher immunogenic response compared to controls. Collectively, our results indicated that Ch and S are effective HBV adjuvants offering natural alternatives, potentially reducing dose.
Collapse
|
42
|
Ravindranathan S, Koppolu BP, Smith SG, Zaharoff DA. Effect of Chitosan Properties on Immunoreactivity. Mar Drugs 2016; 14:md14050091. [PMID: 27187416 PMCID: PMC4882565 DOI: 10.3390/md14050091] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 04/15/2016] [Accepted: 05/02/2016] [Indexed: 11/16/2022] Open
Abstract
Chitosan is a widely investigated biopolymer in drug and gene delivery, tissue engineering and vaccine development. However, the immune response to chitosan is not clearly understood due to contradicting results in literature regarding its immunoreactivity. Thus, in this study, we analyzed effects of various biochemical properties, namely degree of deacetylation (DDA), viscosity/polymer length and endotoxin levels, on immune responses by antigen presenting cells (APCs). Chitosan solutions from various sources were treated with mouse and human APCs (macrophages and/or dendritic cells) and the amount of tumor necrosis factor-α (TNF-α) released by the cells was used as an indicator of immunoreactivity. Our results indicate that only endotoxin content and not DDA or viscosity influenced chitosan-induced immune responses. Our data also indicate that low endotoxin chitosan (<0.01 EU/mg) ranging from 20 to 600 cP and 80% to 97% DDA is essentially inert. This study emphasizes the need for more complete characterization and purification of chitosan in preclinical studies in order for this valuable biomaterial to achieve widespread clinical application.
Collapse
Affiliation(s)
- Sruthi Ravindranathan
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR 72701, USA.
| | - Bhanu Prasanth Koppolu
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR 72701, USA.
| | - Sean G Smith
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR 72701, USA.
| | - David A Zaharoff
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR 72701, USA.
| |
Collapse
|
43
|
The Vaccine Adjuvant Chitosan Promotes Cellular Immunity via DNA Sensor cGAS-STING-Dependent Induction of Type I Interferons. Immunity 2016; 44:597-608. [PMID: 26944200 DOI: 10.1016/j.immuni.2016.02.004] [Citation(s) in RCA: 434] [Impact Index Per Article: 48.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 11/03/2015] [Accepted: 12/03/2015] [Indexed: 01/17/2023]
Abstract
The cationic polysaccharide chitosan is an attractive candidate adjuvant capable of driving potent cell-mediated immunity, but the mechanism by which it acts is not clear. We show that chitosan promotes dendritic cell maturation by inducing type I interferons (IFNs) and enhances antigen-specific T helper 1 (Th1) responses in a type I IFN receptor-dependent manner. The induction of type I IFNs, IFN-stimulated genes and dendritic cell maturation by chitosan required the cytoplasmic DNA sensor cGAS and STING, implicating this pathway in dendritic cell activation. Additionally, this process was dependent on mitochondrial reactive oxygen species and the presence of cytoplasmic DNA. Chitosan-mediated enhancement of antigen specific Th1 and immunoglobulin G2c responses following vaccination was dependent on both cGAS and STING. These findings demonstrate that a cationic polymer can engage the STING-cGAS pathway to trigger innate and adaptive immune responses.
Collapse
|
44
|
Immune cell impact of three differently coated lipid nanocapsules: pluronic, chitosan and polyethylene glycol. Sci Rep 2016; 6:18423. [PMID: 26728491 PMCID: PMC4700454 DOI: 10.1038/srep18423] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 11/09/2015] [Indexed: 12/23/2022] Open
Abstract
Lipid nanocapsules (NCs) represent promising tools in clinical practice for diagnosis and therapy applications. However, the NC appropriate functionalization is essential to guarantee high biocompatibility and molecule loading ability. In any medical application, the immune system-impact of differently functionalized NCs still remains to be fully understood. A comprehensive study on the action exerted on human peripheral blood mononuclear cells (PBMCs) and major immune subpopulations by three different NC coatings: pluronic, chitosan and polyethylene glycol-polylactic acid (PEG) is reported. After a deep particle characterization, the uptake was assessed by flow-cytometry and confocal microscopy, focusing then on apoptosis, necrosis and proliferation impact in T cells and monocytes. Cell functionality by cell diameter variations, different activation marker analysis and cytokine assays were performed. We demonstrated that the NCs impact on the immune cell response is strongly correlated to their coating. Pluronic-NCs were able to induce immunomodulation of innate immunity inducing monocyte activations. Immunomodulation was observed in monocytes and T lymphocytes treated with Chitosan-NCs. Conversely, PEG-NCs were completely inert. These findings are of particular value towards a pre-selection of specific NC coatings depending on biomedical purposes for pre-clinical investigations; i.e. the immune-specific action of particular NC coating can be excellent for immunotherapy applications.
Collapse
|
45
|
Jesus S, Soares E, Borges O. Poly-ε-caprolactone/Chitosan and Chitosan Particles: Two Recombinant Antigen Delivery Systems for Intranasal Vaccination. Methods Mol Biol 2016; 1404:697-713. [PMID: 27076331 DOI: 10.1007/978-1-4939-3389-1_45] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Several evidences converge on the idea that among the mucosal administration routes, the nasal mucosa is the most attractive site for the delivery of vaccines. Mucoadhesive particulate adjuvants should be able to increase the residence time of antigens in nasal cavity in order to increase their probability of being taken up by nasopharynx-associated lymphoid tissue (NALT) cells and subsequently to initiate the innate and adaptive immune response. Focusing on chitosan, a mucoadhesive biopolymer, we describe in this chapter a method to prepare antigen loaded chitosan nanoparticles and a second method to prepare antigen loaded poly-ε-caprolactone/chitosan nanoparticles. Additionally the methodology for the assessment of mucoadhesivity of the delivery system is also described. The two critical procedures in mice intranasal immunization experiments include challenges in the intranasal administration itself due to the small mouse nose, and the other is related with the collection of mucosal secretions to assess the sIgA. The techniques are difficult to perform without advanced training. Therefore, protocols followed in our laboratory, as well as some tips, are described in this chapter.
Collapse
Affiliation(s)
- Sandra Jesus
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Edna Soares
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Olga Borges
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal.
- Faculty of Pharmacy, University of Coimbra, 3000-548, Coimbra, Portugal.
| |
Collapse
|
46
|
González-Aramundiz JV, Peleteiro Olmedo M, González-Fernández Á, Alonso Fernández MJ, Csaba NS. Protamine-based nanoparticles as new antigen delivery systems. Eur J Pharm Biopharm 2015; 97:51-9. [PMID: 26455338 DOI: 10.1016/j.ejpb.2015.09.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 09/29/2015] [Accepted: 09/30/2015] [Indexed: 12/17/2022]
Abstract
The use of biodegradable nanoparticles as antigen delivery vehicles is an attractive approach to overcome the problems associated with the use of Alum-based classical adjuvants. Herein we report, the design and development of protamine-based nanoparticles as novel antigen delivery systems, using recombinant hepatitis B surface antigen as a model viral antigen. The nanoparticles, composed of protamine and a polysaccharide (hyaluronic acid or alginate), were obtained using a mild ionic cross-linking technique. The size and surface charge of the nanoparticles could be modulated by adjusting the ratio of the components. Prototypes with optimal physicochemical characteristics and satisfactory colloidal stability were selected for the assessment of their antigen loading capacity, antigen stability during storage and in vitro and in vivo proof-of-concept studies. In vitro studies showed that antigen-loaded nanoparticles induced the secretion of cytokines by macrophages more efficiently than the antigen in solution, thus indicating a potential adjuvant effect of the nanoparticles. Finally, in vivo studies showed the capacity of these systems to trigger efficient immune responses against the hepatitis B antigen following intramuscular administration, suggesting the potential interest of protamine-polysaccharide nanoparticles as antigen delivery systems.
Collapse
Affiliation(s)
- José Vicente González-Aramundiz
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Health Research Institute of Santiago de Compostela (IDIS), Dept. of Pharmacy and Pharmaceutical Technology, School of Pharmacy, Univ. of Santiago de Compostela, Santiago de Compostela, Spain; Departamento de Farmacia, Facultad de Química, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Mercedes Peleteiro Olmedo
- Immunology, Biomedical Research Center (CINBIO) and Institute of Biomedical Research of Vigo (IBIV), Universidad de Vigo, Campus Lagoas Marcosende, Vigo, Pontevedra, Spain.
| | - África González-Fernández
- Immunology, Biomedical Research Center (CINBIO) and Institute of Biomedical Research of Vigo (IBIV), Universidad de Vigo, Campus Lagoas Marcosende, Vigo, Pontevedra, Spain.
| | - María José Alonso Fernández
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Health Research Institute of Santiago de Compostela (IDIS), Dept. of Pharmacy and Pharmaceutical Technology, School of Pharmacy, Univ. of Santiago de Compostela, Santiago de Compostela, Spain.
| | - Noemi Stefánia Csaba
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Health Research Institute of Santiago de Compostela (IDIS), Dept. of Pharmacy and Pharmaceutical Technology, School of Pharmacy, Univ. of Santiago de Compostela, Santiago de Compostela, Spain.
| |
Collapse
|
47
|
Powell BS, Andrianov AK, Fusco PC. Polyionic vaccine adjuvants: another look at aluminum salts and polyelectrolytes. Clin Exp Vaccine Res 2015; 4:23-45. [PMID: 25648619 PMCID: PMC4313107 DOI: 10.7774/cevr.2015.4.1.23] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 11/24/2014] [Accepted: 11/29/2014] [Indexed: 12/22/2022] Open
Abstract
Adjuvants improve the adaptive immune response to a vaccine antigen by modulating innate immunity or facilitating transport and presentation. The selection of an appropriate adjuvant has become vital as new vaccines trend toward narrower composition, expanded application, and improved safety. Functionally, adjuvants act directly or indirectly on antigen presenting cells (APCs) including dendritic cells (DCs) and are perceived as having molecular patterns associated either with pathogen invasion or endogenous cell damage (known as pathogen associated molecular patterns [PAMPs] and damage associated molecular patterns [DAMPs]), thereby initiating sensing and response pathways. PAMP-type adjuvants are ligands for toll-like receptors (TLRs) and can directly affect DCs to alter the strength, potency, speed, duration, bias, breadth, and scope of adaptive immunity. DAMP-type adjuvants signal via proinflammatory pathways and promote immune cell infiltration, antigen presentation, and effector cell maturation. This class of adjuvants includes mineral salts, oil emulsions, nanoparticles, and polyelectrolytes and comprises colloids and molecular assemblies exhibiting complex, heterogeneous structures. Today innovation in adjuvant technology is driven by rapidly expanding knowledge in immunology, cross-fertilization from other areas including systems biology and materials sciences, and regulatory requirements for quality, safety, efficacy and understanding as part of the vaccine product. Standardizations will aid efforts to better define and compare the structure, function and safety of adjuvants. This article briefly surveys the genesis of adjuvant technology and then re-examines polyionic macromolecules and polyelectrolyte materials, adjuvants currently not known to employ TLR. Specific updates are provided for aluminum-based formulations and polyelectrolytes as examples of improvements to the oldest and emerging classes of vaccine adjuvants in use.
Collapse
Affiliation(s)
| | - Alexander K Andrianov
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD, USA
| | | |
Collapse
|