1
|
Bai Z, Wan D, Lan T, Hong W, Dong H, Wei Y, Wei X. Nanoplatform Based Intranasal Vaccines: Current Progress and Clinical Challenges. ACS NANO 2024; 18:24650-24681. [PMID: 39185745 PMCID: PMC11394369 DOI: 10.1021/acsnano.3c10797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Multiple vaccine platforms have been employed to develop the nasal SARS-CoV-2 vaccines in preclinical studies, and the dominating pipelines are viral vectored as protein-based vaccines. Among them, several viral vectored-based vaccines have entered clinical development. Nevertheless, some unsatisfactory results were reported in these clinical studies. In the face of such urgent situations, it is imperative to rapidly develop the next-generation intranasal COVID-19 vaccine utilizing other technologies. Nanobased intranasal vaccines have emerged as an approach against respiratory infectious diseases. Harnessing the power of nanotechnology, these vaccines offer a noninvasive yet potent defense against pathogens, including the threat of COVID-19. The improvements made in vaccine mucosal delivery technologies based on nanoparticles, such as lipid nanoparticles, polymeric nanoparticles, inorganic nanoparticles etc., not only provide stability and controlled release but also enhance mucosal adhesion, effectively overcoming the limitations of conventional vaccines. Hence, in this review, we overview the evaluation of intranasal vaccine and highlight the current barriers. Next, the modern delivery systems based on nanoplatforms are summarized. The challenges in clinical application of nanoplatform based intranasal vaccine are finally discussed.
Collapse
Affiliation(s)
- Ziyi Bai
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Dandan Wan
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Tianxia Lan
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Weiqi Hong
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Haohao Dong
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| |
Collapse
|
2
|
Guo F, Wei J, Song Y, Song J, Wang Y, Li K, Li B, Qian Z, Wang X, Wang H, Xu T. Immune responses induced by Mycobacterium tuberculosis heat-resistant antigen (Mtb-HAg) upon co-administration with Bacillus Calmette-Guérin in mice. Cytokine 2024; 179:156610. [PMID: 38640558 DOI: 10.1016/j.cyto.2024.156610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/01/2024] [Accepted: 04/06/2024] [Indexed: 04/21/2024]
Abstract
OBJECTIVES To preliminarily assess the immunogenicity of Mtb-HAg in mice and the synergistic effect provided by HAg when co-immunised with BCG. METHODS Mice were randomly grouped for different immunisations and then spleens were aseptically removed and lymphocytes were extracted for immediate detection of cytokines transcript levels and stimulation index(SI), cytokine secretion and multifunctional antigen-specific T cells were detected after incubation for different times. RESULTS HAg extracted from active Mtb is a group of mixed polypeptides with molecular weights of (10-14) kDa. It can significantly stimulate lymphocytes proliferation and increase SI. Injection of HAg alone and in combination with BCG induced significantly higher numbers of multifunctional antigen-specific T cells including CD4+ IFN-γ+, CD4+ IL-2+, CD8+ IFN-γ+, and CD8+ IL-2+ cells than that in BCG-treated mice. Co-immunisation induced the secretion of higher levels of IFN-γ, TNF-α, IL-2 and IL-4 and increased their mRNA expression levels. Significant increases in the transcription levels of IL-10, IL-12 and IL-17 were observed in the co-immunised group with the assistance of HAg. CONCLUSION We demonstrated that HAg has favourable immunogenicity, triggers a stronger Th1-type immune response and proposed the hypothesis that HAg can be used as a BCG booster to further enhance the benefits of BCG.
Collapse
Affiliation(s)
- Fangzheng Guo
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Research Center of Laboratory, School of Laboratory, Bengbu Medical University, Bengbu 233000, China; Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, First Affiliated Hospital of Bengbu Medical University, China.
| | - Jing Wei
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Research Center of Laboratory, School of Laboratory, Bengbu Medical University, Bengbu 233000, China; Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, First Affiliated Hospital of Bengbu Medical University, China.
| | - Yamin Song
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Research Center of Laboratory, School of Laboratory, Bengbu Medical University, Bengbu 233000, China; Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, First Affiliated Hospital of Bengbu Medical University, China.
| | - Jianhan Song
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, First Affiliated Hospital of Bengbu Medical University, China.
| | - Ying Wang
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, First Affiliated Hospital of Bengbu Medical University, China.
| | - Kangsheng Li
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, First Affiliated Hospital of Bengbu Medical University, China.
| | - Baiqing Li
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Research Center of Laboratory, School of Laboratory, Bengbu Medical University, Bengbu 233000, China; Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, First Affiliated Hospital of Bengbu Medical University, China; Department of Clinical Laboratory, School of Laboratory, Bengbu Medical University, Bengbu 233000, China.
| | - Zhongqing Qian
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Research Center of Laboratory, School of Laboratory, Bengbu Medical University, Bengbu 233000, China; Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, First Affiliated Hospital of Bengbu Medical University, China; Department of Clinical Laboratory, School of Laboratory, Bengbu Medical University, Bengbu 233000, China.
| | - Xiaojing Wang
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Research Center of Laboratory, School of Laboratory, Bengbu Medical University, Bengbu 233000, China; Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, First Affiliated Hospital of Bengbu Medical University, China.
| | - Hongtao Wang
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Research Center of Laboratory, School of Laboratory, Bengbu Medical University, Bengbu 233000, China; Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, First Affiliated Hospital of Bengbu Medical University, China; Department of Immunology, School of Laboratory, Bengbu Medical University, Bengbu 233000, China; Department of Laboratory Medicine and Rehabilitation, College of Xinjiang Uyghur Medicine, Hetian 848000, China.
| | - Tao Xu
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Research Center of Laboratory, School of Laboratory, Bengbu Medical University, Bengbu 233000, China; Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, First Affiliated Hospital of Bengbu Medical University, China; Department of Clinical Laboratory, School of Laboratory, Bengbu Medical University, Bengbu 233000, China.
| |
Collapse
|
3
|
Silva S, Bicker J, Falcão A, Fortuna A. Air-liquid interface (ALI) impact on different respiratory cell cultures. Eur J Pharm Biopharm 2023; 184:62-82. [PMID: 36696943 DOI: 10.1016/j.ejpb.2023.01.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 09/24/2022] [Accepted: 01/19/2023] [Indexed: 01/23/2023]
Abstract
The intranasal route has been receiving greater attention from the scientific community not only for systemic drug delivery but also for the treatment of pulmonary and neurological diseases. Along with it, drug transport and permeability studies across the nasal mucosa have exponentially increased. Nevertheless, the translation of data from in vitro cell lines to in vivo studies is not always reliable, due to the difficulty in generating an in vitro model that resembles respiratory human physiology. Among all currently available methodologies, the air-liquid interface (ALI) method is advantageous to promote cell differentiation and optimize the morphological and histological characteristics of airway epithelium cells. Cells grown under ALI conditions, in alternative to submerged conditions, appear to provide relevant input for inhalation and pulmonary toxicology and complement in vivo experiments. Different methodologies and a variety of materials have been used to induce ALI conditions in primary cells and numerous cell lines. Until this day, with only exploratory results, no consensus has been reached regarding the validation of the ALI method, hampering data comparison. The present review describes the most adequate cell models of airway epithelium and how these models are differently affected by ALI conditions. It includes the evaluation of cellular features before and after ALI, and the application of the method in primary cell cultures, commercial 3D primary cells, cell lines and stem-cell derived models. A variety of these models have been recently applied for pharmacological studies against severe acute respiratory syndrome-coronavirus(-2) SARS-CoV(-2), namely primary cultures with alveolar type II epithelium cells and organotypic 3D models. The herein compiled data suggest that ALI conditions must be optimized bearing in mind the type of cells (nasal, bronchial, alveolar), their origin and the objective of the study.
Collapse
Affiliation(s)
- Soraia Silva
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Joana Bicker
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Amílcar Falcão
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Ana Fortuna
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
4
|
Respiratory Vaccination with Hemagglutinin Nanoliposomes Protects Mice from Homologous and Heterologous Strains of Influenza Virus. J Virol 2022; 96:e0100622. [PMID: 36106872 PMCID: PMC9555155 DOI: 10.1128/jvi.01006-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Intranasal vaccination offers the potential advantage of needle-free prevention of respiratory pathogens such as influenza viruses with induction of mucosal immune responses. Optimal design of adjuvants and antigen delivery vehicles for intranasal delivery has not yet been well established. Here, we report that an adjuvant-containing nanoliposome antigen display system that converts soluble influenza hemagglutinin antigens into nanoparticles is effective for intranasal immunization. Intranasal delivery of nanoliposomes in mice delivers the particles to resident immune cells in the respiratory tract, inducing a mucosal response in the respiratory system as evidenced by nasal and lung localized IgA antibody production, while also producing systemic IgG antibodies. Intranasal vaccination with nanoliposome particles decorated with nanogram doses of hemagglutinin protected mice from homologous and heterologous H3N2 and H1N1 influenza virus challenge. IMPORTANCE A self-assembling influenza virus vaccine platform that seamlessly converts soluble antigens into nanoparticles is demonstrated with various H1N1 and H3N2 influenza antigens to protect mice against influenza virus challenge following intranasal vaccination. Mucosal immune responses following liposome delivery to lung antigen-presenting cells are demonstrated.
Collapse
|
5
|
Nian X, Zhang J, Huang S, Duan K, Li X, Yang X. Development of Nasal Vaccines and the Associated Challenges. Pharmaceutics 2022; 14:1983. [PMID: 36297419 PMCID: PMC9609876 DOI: 10.3390/pharmaceutics14101983] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 09/16/2022] [Accepted: 09/16/2022] [Indexed: 02/02/2024] Open
Abstract
Viruses, bacteria, fungi, and several other pathogenic microorganisms usually infect the host via the surface cells of respiratory mucosa. Nasal vaccination could provide a strong mucosal and systemic immunity to combat these infections. The intranasal route of vaccination offers the advantage of easy accessibility over the injection administration. Therefore, nasal immunization is considered a promising strategy for disease prevention, particularly in the case of infectious diseases of the respiratory system. The development of a nasal vaccine, particularly the strategies of adjuvant and antigens design and optimization, enabling rapid induction of protective mucosal and systemic responses against the disease. In recent times, the development of efficacious nasal vaccines with an adequate safety profile has progressed rapidly, with effective handling and overcoming of the challenges encountered during the process. In this context, the present report summarizes the most recent findings regarding the strategies used for developing nasal vaccines as an efficient alternative to conventional vaccines.
Collapse
Affiliation(s)
- Xuanxuan Nian
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
| | - Jiayou Zhang
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
| | - Shihe Huang
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
| | - Kai Duan
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
| | - Xinguo Li
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
| | - Xiaoming Yang
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- China National Biotech Group Company Limited, Beijing 100029, China
| |
Collapse
|
6
|
Fan J, Jin S, Gilmartin L, Toth I, Hussein WM, Stephenson RJ. Advances in Infectious Disease Vaccine Adjuvants. Vaccines (Basel) 2022; 10:1120. [PMID: 35891284 PMCID: PMC9316175 DOI: 10.3390/vaccines10071120] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 02/01/2023] Open
Abstract
Vaccines are one of the most significant medical interventions in the fight against infectious diseases. Since their discovery by Edward Jenner in 1796, vaccines have reduced the worldwide transmission to eradication levels of infectious diseases, including smallpox, diphtheria, hepatitis, malaria, and influenza. However, the complexity of developing safe and effective vaccines remains a barrier for combating many more infectious diseases. Immune stimulants (or adjuvants) are an indispensable factor in vaccine development, especially for inactivated and subunit-based vaccines due to their decreased immunogenicity compared to whole pathogen vaccines. Adjuvants are widely diverse in structure; however, their overall function in vaccine constructs is the same: to enhance and/or prolong an immunological response. The potential for adverse effects as a result of adjuvant use, though, must be acknowledged and carefully managed. Understanding the specific mechanisms of adjuvant efficacy and safety is a key prerequisite for adjuvant use in vaccination. Therefore, rigorous pre-clinical and clinical research into adjuvant development is essential. Overall, the incorporation of adjuvants allows for greater opportunities in advancing vaccine development and the importance of immune stimulants drives the emergence of novel and more effective adjuvants. This article highlights recent advances in vaccine adjuvant development and provides detailed data from pre-clinical and clinical studies specific to infectious diseases. Future perspectives into vaccine adjuvant development are also highlighted.
Collapse
Affiliation(s)
- Jingyi Fan
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.F.); (S.J.); (L.G.); (I.T.); (W.M.H.)
| | - Shengbin Jin
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.F.); (S.J.); (L.G.); (I.T.); (W.M.H.)
| | - Lachlan Gilmartin
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.F.); (S.J.); (L.G.); (I.T.); (W.M.H.)
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.F.); (S.J.); (L.G.); (I.T.); (W.M.H.)
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Waleed M. Hussein
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.F.); (S.J.); (L.G.); (I.T.); (W.M.H.)
| | - Rachel J. Stephenson
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.F.); (S.J.); (L.G.); (I.T.); (W.M.H.)
| |
Collapse
|
7
|
Song H, Zhang S, Yang B, Liu Y, Kang Y, Li Y, Qian A, Yuan Z, Cong B, Shan X. Effects of four different adjuvants separately combined with Aeromonas veronii inactivated vaccine on haematoimmunological state, enzymatic activity, inflammatory response and disease resistance in crucian carp. FISH & SHELLFISH IMMUNOLOGY 2022; 120:658-673. [PMID: 34500055 DOI: 10.1016/j.fsi.2021.09.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 09/01/2021] [Accepted: 09/04/2021] [Indexed: 06/13/2023]
Abstract
The purpose of the current study was to explore the immunomodulatory effects of different adjuvants combined with inactivated vaccines under Aeromonas veronii TH0426 infection in crucian carp. This study explored the best conditions for A. veronii as an inactivated vaccine, and included an animal safety test. Furthermore, we expressed the flagellin FlaA of the A. veronii TH0426 strain for use as an adjuvant supplemented in the diet. Crucian carp were fed 12 different experimental diets for 35 days, including the administration of 10 different adjuvants and inactivated vaccine combinations (50% aluminum hydroxide gel and inactivated vaccine combination, and inactivated vaccine with 20%, 30%, or 50% glucan, astragalus polysaccharide or flagellin), inactivated vaccine alone, and PBS control without adjuvant and inactivated vaccine. After the 42 day feeding trials, the fish were challenged with A. veronii TH0426, and the survival rate over 14 days was recorded. In addition, flagellin FlaA can be expressed normally in large amounts. All experimental groups produced higher levels of IgM serum titres than the control group in the different feeding cycles. Moreover, the activity of serum ACP, AKP, SOD, and LZM, and the expression of inflammatory factors were significantly increased in the experimental groups compared with the control group. The results of qRT-PCR analysis showed that the transcription levels of the IL-10, IL-1β, IFN-γ and TNF-α genes in heart, liver, spleen and kidney tissues were significantly enhanced by adjuvant treatment, indicating that the addition of adjuvants can significantly promote the body's inflammatory response. In addition, the phagocytic activity of leukocytes in each adjuvant treated group was significantly enhanced compared to that in the groups without adjuvant. After the A. veronii challenge, the survival rate of all adjuvant-treated groups was significantly higher than that of the control group, and the 50% flagellin adjuvant group had the highest rate of 78.37%. Overall, our findings strongly indicate that adjuvants not only significantly improve the body's immunity, but also exhibit a strong anti-infection ability. Importantly, this work provides a new perspective for the prevention and control of aquaculture diseases.
Collapse
Affiliation(s)
- Haichao Song
- College of Veterinary Medicine, College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, Jilin, China
| | - Siqi Zhang
- Dunhua Agricultural and Rural Bureau, Dunhua, Jilin, China
| | - Bintong Yang
- Marine College, Shandong University, Weihai, China
| | - Yanhui Liu
- Jilin Academy of Sciences, Changchun, Jilin, China
| | | | - Ying Li
- College of Veterinary Medicine, College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, Jilin, China
| | - Aidong Qian
- College of Veterinary Medicine, College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, Jilin, China
| | - Zhonghua Yuan
- College of Veterinary Medicine, College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, Jilin, China
| | - Bo Cong
- Institute of Special Animal and Plant Sciences of CAAS, Changchun, Jilin, China.
| | - Xiaofeng Shan
- College of Veterinary Medicine, College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, Jilin, China.
| |
Collapse
|
8
|
Inaku K, Ene A, Kasimu S, Bolarin D. Development of Severe Acute Respiratory Syndrome Corona Virus-2 (SARS-COV-2) Vaccines. NIGERIAN JOURNAL OF MEDICINE 2022. [DOI: 10.4103/njm.njm_172_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
9
|
Nakhaei P, Margiana R, Bokov DO, Abdelbasset WK, Jadidi Kouhbanani MA, Varma RS, Marofi F, Jarahian M, Beheshtkhoo N. Liposomes: Structure, Biomedical Applications, and Stability Parameters With Emphasis on Cholesterol. Front Bioeng Biotechnol 2021; 9:705886. [PMID: 34568298 PMCID: PMC8459376 DOI: 10.3389/fbioe.2021.705886] [Citation(s) in RCA: 217] [Impact Index Per Article: 72.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 08/09/2021] [Indexed: 12/12/2022] Open
Abstract
Liposomes are essentially a subtype of nanoparticles comprising a hydrophobic tail and a hydrophilic head constituting a phospholipid membrane. The spherical or multilayered spherical structures of liposomes are highly rich in lipid contents with numerous criteria for their classification, including structural features, structural parameters, and size, synthesis methods, preparation, and drug loading. Despite various liposomal applications, such as drug, vaccine/gene delivery, biosensors fabrication, diagnosis, and food products applications, their use encounters many limitations due to physico-chemical instability as their stability is vigorously affected by the constituting ingredients wherein cholesterol performs a vital role in the stability of the liposomal membrane. It has well established that cholesterol exerts its impact by controlling fluidity, permeability, membrane strength, elasticity and stiffness, transition temperature (Tm), drug retention, phospholipid packing, and plasma stability. Although the undetermined optimum amount of cholesterol for preparing a stable and controlled release vehicle has been the downside, but researchers are still focused on cholesterol as a promising material for the stability of liposomes necessitating explanation for the stability promotion of liposomes. Herein, the prior art pertaining to the liposomal appliances, especially for drug delivery in cancer therapy, and their stability emphasizing the roles of cholesterol.
Collapse
Affiliation(s)
- Pooria Nakhaei
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ria Margiana
- Department of Anatomy, Faculty of Medicine, Universitas Indonesia, Depok, Indonesia
- Cipto Mangunkusumo Hospital, The National Referral Hospital, Central Jakarta, Indonesia
- Master’s Programme Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Depok, Indonesia
| | - Dmitry O. Bokov
- Institute of Pharmacy, Sechenov First Moscow State Medical University, Moscow, Russia
- Laboratory of Food Chemistry, Federal Research Center of Nutrition, Biotechnology, and Food Safety, Moscow, Russia
| | - Walid Kamal Abdelbasset
- Department of Health and Rehabilitation Sciences, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Al Kharj, Saudi Arabia
- Department of Physical Therapy, Kasr Al-Aini Hospital, Cairo University, Giza, Egypt
| | - Mohammad Amin Jadidi Kouhbanani
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Czechia
| | - Rajender S. Varma
- Regional Centre of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute, Palacký University in Olomouc, Olomouc, Czechia
| | - Faroogh Marofi
- Department of Hematology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mostafa Jarahian
- Toxicology and Chemotherapy Unit (G401), German Cancer Research Center, Heidelberg, Germany
| | - Nasrin Beheshtkhoo
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Czechia
| |
Collapse
|
10
|
Razim A, Pyclik M, Pacyga K, Górska S, Xu J, Olszewski MA, Gamian A, Myc A. Silicone Oil-Based Nanoadjuvants as Candidates for a New Formulation of Intranasal Vaccines. Vaccines (Basel) 2021; 9:vaccines9030234. [PMID: 33800507 PMCID: PMC7999606 DOI: 10.3390/vaccines9030234] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/03/2021] [Accepted: 03/05/2021] [Indexed: 01/05/2023] Open
Abstract
Many conventional vaccines are administered via a needle injection, while most pathogens primarily invade the host via mucosal surfaces. Moreover, protective IgA antibodies are insufficiently induced by parenteral vaccines. Mucosal immunity induces both local and systemic response to pathogens and typically lasts for long periods of time. Therefore, vaccination via mucosal routes has been increasingly explored. However, mucosal vaccines require potent adjuvants to become efficacious. Despite many efforts to develop safe and robust adjuvants for mucosal vaccines, only a few have been approved for use in human formulations. The aim of our study was to design, develop and characterize new silicone oil-based nanoadjuvant candidates for intranasal vaccines with potential to become mucosal adjuvants. We have developed an array of nanoadjuvant candidates (NACs), based on well-defined ingredients. NAC1, 2 and 3 are based on silicone oil, but differ in the used detergents and organic solvents, which results in variations in their droplet size and zeta potential. NACs' cytotoxicity, Tumor Necrosis Factor α (TNF-α) induction and their effect on antigen engulfment by immune cells were tested in vitro. Adjuvant properties of NACs were verified by intranasal vaccination of mice together with ovalbumin (OVA). NACs show remarkable stability and do not require any special storage conditions. They exhibit bio-adhesiveness and influence the degree of model protein engulfment by epithelial cells. Moreover, they induce high specific anti-OVA IgG antibody titers after two intranasal administrations. Nanoadjuvant candidates composed of silicone oil and cationic detergents are stable, exhibit remarkable adjuvant properties and can be used as adjuvants for intranasal immunization.
Collapse
Affiliation(s)
- Agnieszka Razim
- Department of Microbiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (M.P.); (K.P.); (S.G.)
- Correspondence:
| | - Marcelina Pyclik
- Department of Microbiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (M.P.); (K.P.); (S.G.)
| | - Katarzyna Pacyga
- Department of Microbiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (M.P.); (K.P.); (S.G.)
| | - Sabina Górska
- Department of Microbiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (M.P.); (K.P.); (S.G.)
| | - Jintao Xu
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, MI 48109, USA; (J.X.); (M.A.O.)
- Research Service, Department of Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA
| | - Michal A. Olszewski
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, MI 48109, USA; (J.X.); (M.A.O.)
- Research Service, Department of Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA
| | - Andrzej Gamian
- Department of Immunology of Infectious Diseases, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (A.G.); (A.M.)
| | - Andrzej Myc
- Department of Immunology of Infectious Diseases, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (A.G.); (A.M.)
- MNIMBS, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
11
|
Flanagan KL, Best E, Crawford NW, Giles M, Koirala A, Macartney K, Russell F, Teh BW, Wen SCH. Progress and Pitfalls in the Quest for Effective SARS-CoV-2 (COVID-19) Vaccines. Front Immunol 2020; 11:579250. [PMID: 33123165 PMCID: PMC7566192 DOI: 10.3389/fimmu.2020.579250] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 08/31/2020] [Indexed: 12/15/2022] Open
Abstract
There are currently around 200 SARS-CoV-2 candidate vaccines in preclinical and clinical trials throughout the world. The various candidates employ a range of vaccine strategies including some novel approaches. Currently, the goal is to prove that they are safe and immunogenic in humans (phase 1/2 studies) with several now advancing into phase 2 and 3 trials to demonstrate efficacy and gather comprehensive data on safety. It is highly likely that many vaccines will be shown to stimulate antibody and T cell responses in healthy individuals and have an acceptable safety profile, but the key will be to confirm that they protect against COVID-19. There is much hope that SARS-CoV-2 vaccines will be rolled out to the entire world to contain the pandemic and avert its most damaging impacts. However, in all likelihood this will initially require a targeted approach toward key vulnerable groups. Collaborative efforts are underway to ensure manufacturing can occur at the unprecedented scale and speed required to immunize billions of people. Ensuring deployment also occurs equitably across the globe will be critical. Careful evaluation and ongoing surveillance for safety will be required to address theoretical concerns regarding immune enhancement seen in previous contexts. Herein, we review the current knowledge about the immune response to this novel virus as it pertains to the design of effective and safe SARS-CoV-2 vaccines and the range of novel and established approaches to vaccine development being taken. We provide details of some of the frontrunner vaccines and discuss potential issues including adverse effects, scale-up and delivery.
Collapse
Affiliation(s)
- Katie L. Flanagan
- Department of Infectious Diseases, Launceston General Hospital, Launceston, TAS, Australia
- Faculty of Health Sciences and School of Medicine, University of Tasmania, Launceston, TAS, Australia
- School of Health and Biomedical Science, Royal Melbourne Institute of Technology (RMIT) University, Melbourne, VIC, Australia
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia
| | - Emma Best
- Department of Paediatric Infectious Diseases, Starship Children's Hospital, Auckland, New Zealand
- Department of Paediatrics: Child and Youth Health, University of Auckland, Auckland, New Zealand
| | - Nigel W. Crawford
- Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
- Murdoch Children's Research Institute, Royal Children's Hospital Immunisation Service, Melbourne, VIC, Australia
| | - Michelle Giles
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
- Infectious Diseases Unit, Alfred Health, Melbourne, VIC, Australia
| | - Archana Koirala
- Department of Child and Adolescent Health, University of Sydney, Sydney, NSW, Australia
- National Centre for Immunisation Research & Surveillance (NCIRS), Sydney, NSW, Australia
- Department of Infectious Diseases, Nepean Hospital, Sydney, NSW, Australia
| | - Kristine Macartney
- Department of Child and Adolescent Health, University of Sydney, Sydney, NSW, Australia
- National Centre for Immunisation Research & Surveillance (NCIRS), Sydney, NSW, Australia
| | - Fiona Russell
- Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
- Murdoch Children's Research Institute, Royal Children's Hospital Immunisation Service, Melbourne, VIC, Australia
| | - Benjamin W. Teh
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Sophie CH Wen
- Infection Management Prevention Services, Queensland Children's Hospital, South Brisbane, QLD, Australia
- University of Queensland Centre for Clinical Research (UQCCR), University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
12
|
Calzas C, Chevalier C. Innovative Mucosal Vaccine Formulations Against Influenza A Virus Infections. Front Immunol 2019; 10:1605. [PMID: 31379823 PMCID: PMC6650573 DOI: 10.3389/fimmu.2019.01605] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 06/27/2019] [Indexed: 12/11/2022] Open
Abstract
Despite efforts made to develop efficient preventive strategies, infections with influenza A viruses (IAV) continue to cause serious clinical and economic problems. Current licensed human vaccines are mainly inactivated whole virus particles or split-virion administered via the parenteral route. These vaccines provide incomplete protection against IAV in high-risk groups and are poorly/not effective against the constant antigenic drift/shift occurring in circulating strains. Advances in mucosal vaccinology and in the understanding of the protective anti-influenza immune mechanisms suggest that intranasal immunization is a promising strategy to fight against IAV. To date, human mucosal anti-influenza vaccines consist of live attenuated strains administered intranasally, which elicit higher local humoral and cellular immune responses than conventional parenteral vaccines. However, because of inconsistent protective efficacy and safety concerns regarding the use of live viral strains, new vaccine candidates are urgently needed. To prime and induce potent and long-lived protective immune responses, mucosal vaccine formulations need to ensure the immunoavailability and the immunostimulating capacity of the vaccine antigen(s) at the mucosal surfaces, while being minimally reactogenic/toxic. The purpose of this review is to compile innovative delivery/adjuvant systems tested for intranasal administration of inactivated influenza vaccines, including micro/nanosized particulate carriers such as lipid-based particles, virus-like particles and polymers associated or not with immunopotentiatory molecules including microorganism-derived toxins, Toll-like receptor ligands and cytokines. The capacity of these vaccines to trigger specific mucosal and systemic humoral and cellular responses against IAV and their (cross)-protective potential are considered.
Collapse
Affiliation(s)
- Cynthia Calzas
- VIM, UR892, Equipe Virus Influenza, INRA, University PARIS-SACLAY, Jouy-en-Josas, France
| | - Christophe Chevalier
- VIM, UR892, Equipe Virus Influenza, INRA, University PARIS-SACLAY, Jouy-en-Josas, France
| |
Collapse
|
13
|
Blakney AK, McKay PF, Christensen D, Yus BI, Aldon Y, Follmann F, Shattock RJ. Effects of cationic adjuvant formulation particle type, fluidity and immunomodulators on delivery and immunogenicity of saRNA. J Control Release 2019; 304:65-74. [PMID: 31071377 DOI: 10.1016/j.jconrel.2019.04.043] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 04/07/2019] [Accepted: 04/29/2019] [Indexed: 01/07/2023]
Abstract
Self-amplifying RNA (saRNA) is well suited as a vaccine platform against chlamydia, as it is relatively affordable and scalable, has been shown to induce immunity against multivalent antigens, and can result in protein expression for up to 60 days. Cationic adjuvant formulations (CAFs) have been previously investigated as an adjuvant for protein subunit vaccines; here we optimize the CAFs for delivery of saRNA in vivo and observe the immunogenicity profile in the context of both cellular and humoral immunity against the major outer membrane protein (MOMP) of Chlamydia trachomatis. We tested both liposomal and emulsion based CAFs with solid and fluid phase lipids, with or without the TLR agonists R848 and 3M-052, for in vitro transfection efficiency and cytotoxicity. We then optimized the RNA/delivery system ratio for in vivo delivery using saRNA coding for firefly luciferase (fLuc) as a reporter protein in vivo. We observed that while the fluid phase liposome formulations showed the highest in vitro transfection efficiency, the fluid and solid phase liposomes had equivalent luciferase expression in vivo. Incorporation of R848 or 3M-052 into the formulation was not observed to affect the delivery efficiency of saRNA either in vitro or in vivo. MOMP-encoding saRNA complexed with CAFs resulted in both MOMP-specific cellular and humoral immunity, and while there was a slight enhancement of IFN-γ+ T-cell responses when R848 was incorporated into the formulation, the self-adjuvanting effects of RNA appeared to dominate the immune response. These studies establish that CAFs are efficient delivery vehicles for saRNA both for in vitro transfections and in vivo immunogenicity and generate cellular and humoral responses that are proportionate to protein expression.
Collapse
Affiliation(s)
- Anna K Blakney
- Department of Medicine, Imperial College London, London, UK
| | - Paul F McKay
- Department of Medicine, Imperial College London, London, UK
| | - Dennis Christensen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | | | - Yoann Aldon
- Department of Medicine, Imperial College London, London, UK
| | - Frank Follmann
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | | |
Collapse
|
14
|
Riccomi A, Piccaro G, Christensen D, Palma C, Andersen P, Vendetti S. Parenteral Vaccination With a Tuberculosis Subunit Vaccine in Presence of Retinoic Acid Provides Early but Transient Protection to M. Tuberculosis Infection. Front Immunol 2019; 10:934. [PMID: 31130946 PMCID: PMC6509564 DOI: 10.3389/fimmu.2019.00934] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 04/11/2019] [Indexed: 12/17/2022] Open
Abstract
Most microbes invading through mucosal surfaces cause disease and therefore strategies to induce mucosal immune responses are strongly needed. Vitamin A metabolites, such as retinoic acid (RA), play crucial roles in programming T and B cells to home to mucosal compartments, therefore we evaluated the capacity of RA to elicit mucosal immune responses against tuberculosis (TB) after parenteral vaccination. We found that mice immunized through subcutaneous injections with the TB subunit vaccine (CAF01+H56) in presence of RA show enhanced mucosal H56-specific IgA responses and enhanced Ag-specific CD4+ T lymphocytes homing to the lung as compared with control mice. Immunization with CAF01+H56 in presence of RA resulted in lower bacterial loads in the lungs of mice 14 days after challenge with virulent Mycobacterium tuberculosis (Mtb) as compared to mice immunized in the absence of RA or vaccinated with BCG. Higher amounts of IFNγ and IL-17 pro-inflammatory cytokines were found in lung homogenates of mice immunized with CAF01+H56 and RA 24 h after Mtb infection. However, 6 weeks after infection the protection was comparable in vaccinated mice with or without RA even though treatment with RA during immunization is able to better contain the inflammatory response by the host. Furthermore, at later stage of the infection a higher percentage of Mtb specific CD4+PD1+ T lymphocytes were found in the lungs of mice immunized with CAF01+H56 and RA. These data show that an enhanced mucosal immune response is generated during parenteral vaccination in presence of RA. Furthermore, RA treatment contained the bacterial growth at an early stage of the infection and limited the inflammatory response in the lung at later time points.
Collapse
Affiliation(s)
- Antonella Riccomi
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | | | - Dennis Christensen
- Department of Infectious Diseases Immunology, Statens Serum Institute, Copenhagen, Denmark
| | - Carla Palma
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Peter Andersen
- Department of Infectious Diseases Immunology, Statens Serum Institute, Copenhagen, Denmark
| | - Silvia Vendetti
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
15
|
Corthésy B, Bioley G. Lipid-Based Particles: Versatile Delivery Systems for Mucosal Vaccination against Infection. Front Immunol 2018; 9:431. [PMID: 29563912 PMCID: PMC5845866 DOI: 10.3389/fimmu.2018.00431] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 02/19/2018] [Indexed: 12/19/2022] Open
Abstract
Vaccination is the process of administering immunogenic formulations in order to induce or harness antigen (Ag)-specific antibody and T cell responses in order to protect against infections. Important successes have been obtained in protecting individuals against many deleterious pathological situations after parenteral vaccination. However, one of the major limitations of the current vaccination strategies is the administration route that may not be optimal for the induction of immunity at the site of pathogen entry, i.e., mucosal surfaces. It is now well documented that immune responses along the genital, respiratory, or gastrointestinal tracts have to be elicited locally to ensure efficient trafficking of effector and memory B and T cells to mucosal tissues. Moreover, needle-free mucosal delivery of vaccines is advantageous in terms of safety, compliance, and ease of administration. However, the quest for mucosal vaccines is challenging due to (1) the fact that Ag sampling has to be performed across the epithelium through a relatively limited number of portals of entry; (2) the deleterious acidic and proteolytic environment of the mucosae that affect the stability, integrity, and retention time of the applied Ags; and (3) the tolerogenic environment of mucosae, which requires the addition of adjuvants to elicit efficient effector immune responses. Until now, only few mucosally applicable vaccine formulations have been developed and successfully tested. In animal models and clinical trials, the use of lipidic structures such as liposomes, virosomes, immune stimulating complexes, gas-filled microbubbles and emulsions has proven efficient for the mucosal delivery of associated Ags and the induction of local and systemic immune reponses. Such particles are suitable for mucosal delivery because they protect the associated payload from degradation and deliver concentrated amounts of Ags via specialized sampling cells (microfold cells) within the mucosal epithelium to underlying antigen-presenting cells. The review aims at summarizing recent development in the field of mucosal vaccination using lipid-based particles. The modularity ensured by tailoring the lipidic design and content of particles, and their known safety as already established in humans, make the continuing appraisal of these vaccine candidates a promising development in the field of targeted mucosal vaccination.
Collapse
Affiliation(s)
- Blaise Corthésy
- R&D Laboratory, Division of Immunology and Allergy, Centre des Laboratoires d'Epalinges, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Gilles Bioley
- R&D Laboratory, Division of Immunology and Allergy, Centre des Laboratoires d'Epalinges, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| |
Collapse
|
16
|
Qu W, Li N, Yu R, Zuo W, Fu T, Fei W, Hou Y, Liu Y, Yang J. Cationic DDA/TDB liposome as a mucosal vaccine adjuvant for uptake by dendritic cells in vitro induces potent humoural immunity. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:852-860. [PMID: 29447484 DOI: 10.1080/21691401.2018.1438450] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The cationic dimethyldioctadecylammonium/trehalose 6,6,9-dibehenate (DDA/TDB) liposome is as a strong adjuvant system for vaccines, with remarkable immunostimulatory activity. The mucosal administration of vaccines is a potential strategy for inducing earlier and stronger mucosal immune responses to infectious diseases. In this study, we assessed whether the intranasal administration of cationic DDA/TDB liposomes combined with influenza antigen A (H3N2) can be used as a highly efficacious vaccine to induce mucosal and systemic antibody responses. Confocal laser scanning microscopy and a flow-cytometric analysis showed that the uptake of the cationic DDA/TDB liposome carrier was significantly higher than that of neutral 1,2-distearoyl-sn-glycero-3-phosphocholine/cholesterol (DSPC/Chol) or cationic 1,2-dioleoyl-3-trimethylammonium-propane/3β-(N-[N',N'-dimethylaminoethane]-carbamoyl (DOTAP/DC-Chol) liposomes. Our results indicate that the cationic DDA/TDB liposome is more effective in facilitating its uptake by dendritic cells (DCs) in vitro than the DSPC/Chol or DOTAP/DC-Chol liposome. DCs treated with DDA/TDB liposomes strongly expressed CD80, CD86, and MHC II molecules, whereas those treated with DSPC/Chol or DOTAP/DC-Chol liposomes did not. C57BL/6 mice intranasally immunized with H3N2-encapsulating cationic DDA/TDB liposomes had significantly higher H3N2-specific s-IgA levels in their nasal wash fluid than those treated with other formulations. The DDA/TDB liposomes also simultaneously enhanced the serum IgG IgG2a, IgG1, and IgG2b antibody responses. In summary, DDA/TDB liposomes effectively facilitated their uptake by DCs and DCs maturation in vitro, and induced significantly higher mucosal IgA, systemic IgG, IgG1, and IgG2b antibody titres than other formulations after their intranasal administration in vivo. These results indicate that DDA/TDB liposomes are a promising antigen delivery carrier for clinical antiviral applications.
Collapse
Affiliation(s)
- Wenjing Qu
- a Department of Pharmaceutics, School of Pharmacy , Ningxia Medical University , Yinchuan , PR China
| | - Na Li
- a Department of Pharmaceutics, School of Pharmacy , Ningxia Medical University , Yinchuan , PR China
| | - Rui Yu
- a Department of Pharmaceutics, School of Pharmacy , Ningxia Medical University , Yinchuan , PR China
| | - Wenbao Zuo
- a Department of Pharmaceutics, School of Pharmacy , Ningxia Medical University , Yinchuan , PR China
| | - Tingting Fu
- a Department of Pharmaceutics, School of Pharmacy , Ningxia Medical University , Yinchuan , PR China
| | - Wenling Fei
- a Department of Pharmaceutics, School of Pharmacy , Ningxia Medical University , Yinchuan , PR China
| | - Yanhui Hou
- a Department of Pharmaceutics, School of Pharmacy , Ningxia Medical University , Yinchuan , PR China
| | - Yanhua Liu
- a Department of Pharmaceutics, School of Pharmacy , Ningxia Medical University , Yinchuan , PR China
| | - Jianhong Yang
- a Department of Pharmaceutics, School of Pharmacy , Ningxia Medical University , Yinchuan , PR China
| |
Collapse
|
17
|
Pal S, Tifrea DF, Follmann F, Andersen P, de la Maza LM. The cationic liposomal adjuvants CAF01 and CAF09 formulated with the major outer membrane protein elicit robust protection in mice against a Chlamydia muridarum respiratory challenge. Vaccine 2017; 35:1705-1711. [PMID: 28238632 DOI: 10.1016/j.vaccine.2017.02.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 01/20/2017] [Accepted: 02/09/2017] [Indexed: 12/31/2022]
Abstract
Two cationic liposomal adjuvants CAF01 and CAF09 were formulated with the native or the recombinant Chlamydia muridarum major outer membrane protein (nMOMP and rMOMP). BALB/c mice were immunized with the four vaccine formulations using the subcutaneous followed by the intranasal (i.n.) routes. As positive controls mice were inoculated i.n. with live C. muridarum and negative controls received i.n. minimal essential medium (MEM). Four weeks after the last immunization mice were challenged i.n. with 104 inclusion forming units (IFU) of C. muridarum. Following the challenge the mice were weighed daily. At 10days post-challenge the mice were euthanized, their lungs weighed and the number of C. muridarum IFU determined. Serum collected the day before the challenge showed that all four groups of mice immunized with CAF01, or CAF09 and MOMP had significant C. muridarum-specific antibody titers. As determined by a T-cell lymphoproliferative assay, these four groups of mice also mounted robust cell mediated immune responses with high production of IFN-γ and IL17 and low levels of IL-4. Following the challenge the four groups of mice lost significantly less body weight than the MEM-immunized group. Lungs of mice vaccinated with CAF01, or CAF09, and nMOMP were significantly lighter than those from mice immunized using rMOMP. The number of IFU recovered from the lungs of mice vaccinated with CAF01, or CAF09, and nMOMP was similar to the number of IFU recovered from mice immunized with live EB. Mice that received rMOMP had significantly higher numbers of IFU than other groups. In conclusion, CAF01 and CAF09 elicited very robust protective humoral and cellular immune responses and were equally effective at adjuntavizing the C. muridarum MOMP. Mice vaccinated with nMOMP were significantly better protected than those immunized with rMOMP, indicative of the importance of the structural conformation of this antigen in protection.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Administration, Intranasal
- Animals
- Antibodies, Bacterial/blood
- Bacterial Outer Membrane Proteins/immunology
- Bacterial Vaccines/administration & dosage
- Bacterial Vaccines/immunology
- Body Weight
- Cell Proliferation
- Chlamydia Infections/pathology
- Chlamydia Infections/prevention & control
- Chlamydia muridarum/immunology
- Cytokines/metabolism
- Disease Models, Animal
- Injections, Subcutaneous
- Liposomes/administration & dosage
- Lung/pathology
- Mice, Inbred BALB C
- Pneumonia, Bacterial/pathology
- Pneumonia, Bacterial/prevention & control
- T-Lymphocytes/immunology
- Treatment Outcome
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/immunology
Collapse
Affiliation(s)
- Sukumar Pal
- Department of Pathology and Laboratory Medicine, Medical Sciences I, Room D440, University of California, Irvine, Irvine, CA 92697-4800, USA
| | - Delia F Tifrea
- Department of Pathology and Laboratory Medicine, Medical Sciences I, Room D440, University of California, Irvine, Irvine, CA 92697-4800, USA
| | - Frank Follmann
- Department of Infectious Disease Immunology, Adjuvant Research, Staten Serum Institute, Copenhagen, Denmark
| | - Peter Andersen
- Department of Infectious Disease Immunology, Adjuvant Research, Staten Serum Institute, Copenhagen, Denmark
| | - Luis M de la Maza
- Department of Pathology and Laboratory Medicine, Medical Sciences I, Room D440, University of California, Irvine, Irvine, CA 92697-4800, USA.
| |
Collapse
|
18
|
Wu H, Bao Y, Wang X, Zhou D, Wu W. Alkyl polyglycoside, a highly promising adjuvant in intranasal split influenza vaccines. Hum Vaccin Immunother 2017; 13:1-9. [PMID: 28129034 DOI: 10.1080/21645515.2016.1278098] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Influenza viral infections are significant global public health concerns due to the morbidity and mortality associated with acute respiratory disease, secondary complications, and pandemic threats; thus, continuous efforts have been made to develop potent influenza vaccines. In this study, 3 different mucosal adjuvants-alkyl polyglycoside (APG), gellan gum, and chitosan (CS)-were evaluated for their efficacy in intranasal A/H1N1 or B split influenza vaccines administered to BALB/c mice. Protective immunity was monitored by serum analysis for IgG, hemagglutination inhibition (HI), and neutralizing antibody levels, as well as mucosal IgA levels in nasal and pulmonary lavage fluids. Survival, body weight, lung viral titer, and pulmonary immunopathology were also examined following lethal influenza challenge. Notably, all adjuvants amplified the IgG and IgA immune responses (not detected in immunization of influenza B) and increased survival rate compared with controls administered adjuvant-free intranasal vaccines. Alternatively, intramuscular immunization stimulated IgG production, but had no effect on IgA levels. Our collective analysis identified that APG was the most effective intranasal adjuvant, as all mice survived influenza challenge with limited body weight loss, viral titer, and pulmonary pathology, similar to those observed with intramuscular vaccination. This evidence supports that APG can elicit both systemic and mucosal immunity, and may be an effective adjuvant in intranasal split influenza A/H1N1 and B vaccines.
Collapse
Affiliation(s)
- Hui Wu
- a National Pharmaceutical Engineering Research Center, China State Institute of Pharmaceutical Industry , Shanghai , China
| | - Yuanyuan Bao
- a National Pharmaceutical Engineering Research Center, China State Institute of Pharmaceutical Industry , Shanghai , China
| | - Xiang Wang
- b Vaccine Research Center, Institut Pasteur of Shanghai, Chinese Academy of Sciences , Shanghai , China
| | - Dongming Zhou
- b Vaccine Research Center, Institut Pasteur of Shanghai, Chinese Academy of Sciences , Shanghai , China
| | - Wenzhe Wu
- a National Pharmaceutical Engineering Research Center, China State Institute of Pharmaceutical Industry , Shanghai , China
| |
Collapse
|
19
|
Kakhi Z, Frisch B, Heurtault B, Pons F. Liposomal constructs for antitumoral vaccination by the nasal route. Biochimie 2016; 130:14-22. [DOI: 10.1016/j.biochi.2016.07.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 07/06/2016] [Indexed: 11/27/2022]
|
20
|
Heegaard PMH, Fang Y, Jungersen G. Novel Adjuvants and Immunomodulators for Veterinary Vaccines. Methods Mol Biol 2016; 1349:63-82. [PMID: 26458830 DOI: 10.1007/978-1-4939-3008-1_5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Adjuvants are crucial for efficacy of vaccines, especially subunit and recombinant vaccines. Rational vaccine design, including knowledge-based and molecularly defined adjuvants tailored for directing and potentiating specific types of host immune responses towards the antigens included in the vaccine is becoming a reality with our increased understanding of innate and adaptive immune activation. This will allow future vaccines to induce immune reactivity having adequate specificity as well as protective and recallable immune effector mechanisms in appropriate body compartments, including mucosal surfaces. Here we describe these new developments and, when possible, relate new immunological knowledge to the many years of experience with traditional, empirical adjuvants. Finally, some protocols are given for production of emulsion (oil-based) and liposome-based adjuvant/antigen formulations.
Collapse
Affiliation(s)
- Peter M H Heegaard
- Section for Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, Bülowsvej 27, 1870, Frederiksberg C, Denmark.
| | - Yongxiang Fang
- Section for Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, Bülowsvej 27, 1870, Frederiksberg C, Denmark.,State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Public Health of Ministry of Agriculture, Lanzhou Veterinary Research Institute, CAAS, Lanzhou, 730046, China
| | - Gregers Jungersen
- Section for Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, Bülowsvej 27, 1870, Frederiksberg C, Denmark
| |
Collapse
|
21
|
Lorenzen E, Follmann F, Bøje S, Erneholm K, Olsen AW, Agerholm JS, Jungersen G, Andersen P. Intramuscular Priming and Intranasal Boosting Induce Strong Genital Immunity Through Secretory IgA in Minipigs Infected with Chlamydia trachomatis. Front Immunol 2015; 6:628. [PMID: 26734002 PMCID: PMC4679855 DOI: 10.3389/fimmu.2015.00628] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 11/30/2015] [Indexed: 11/13/2022] Open
Abstract
International efforts in developing a vaccine against Chlamydia trachomatis have highlighted the need for novel immunization strategies for the induction of genital immunity. In this study, we evaluated an intramuscular (IM) prime/intranasal boost vaccination strategy in a Göttingen Minipig model with a reproductive system very similar to humans. The vaccine was composed of C. trachomatis subunit antigens formulated in the Th1/Th17 promoting CAF01 adjuvant. IM priming immunizations with CAF01 induced a significant cell-mediated interferon gamma and interleukin 17A response and a significant systemic high-titered neutralizing IgG response. Following genital challenge, intranasally boosted groups mounted an accelerated, highly significant genital IgA response that correlated with enhanced bacterial clearance on day 3 post infection. By detecting antigen-specific secretory component (SC), we showed that the genital IgA was locally produced in the genital mucosa. The highly significant inverse correlation between the vaginal IgA SC response and the chlamydial load suggests that IgA in the minipig model is involved in protection against C. trachomatis. This is important both for our understanding of protective immunity and future vaccination strategies against C. trachomatis and genital pathogens in general.
Collapse
Affiliation(s)
- Emma Lorenzen
- Section for Veterinary Reproduction and Obstetrics, Department of Large Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Infectious Disease Immunology, Chlamydia Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Frank Follmann
- Department of Infectious Disease Immunology, Chlamydia Vaccine Research, Statens Serum Institut , Copenhagen , Denmark
| | - Sarah Bøje
- Section for Veterinary Reproduction and Obstetrics, Department of Large Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Infectious Disease Immunology, Chlamydia Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Karin Erneholm
- Section for Veterinary Reproduction and Obstetrics, Department of Large Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Infectious Disease Immunology, Chlamydia Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Anja Weinreich Olsen
- Department of Infectious Disease Immunology, Chlamydia Vaccine Research, Statens Serum Institut , Copenhagen , Denmark
| | - Jørgen Steen Agerholm
- Section for Veterinary Reproduction and Obstetrics, Department of Large Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Gregers Jungersen
- Section for Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark , Copenhagen , Denmark
| | - Peter Andersen
- Department of Infectious Disease Immunology, Chlamydia Vaccine Research, Statens Serum Institut , Copenhagen , Denmark
| |
Collapse
|
22
|
Davitt CJ, Lavelle EC. Delivery strategies to enhance oral vaccination against enteric infections. Adv Drug Deliv Rev 2015; 91:52-69. [PMID: 25817337 DOI: 10.1016/j.addr.2015.03.007] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 02/25/2015] [Accepted: 03/12/2015] [Indexed: 01/22/2023]
Abstract
While the majority of human pathogens infect the body through mucosal sites, most licensed vaccines are injectable. In fact the only mucosal vaccine that has been widely used globally for infant and childhood vaccination programs is the oral polio vaccine (OPV) developed by Albert Sabin in the 1950s. While oral vaccines against Cholera, rotavirus and Salmonella typhi have also been licensed, the development of additional non-living oral vaccines against these and other enteric pathogens has been slow and challenging. Mucosal vaccines can elicit protective immunity at the gut mucosa, in part via antigen-specific secretory immunoglobulin A (SIgA). However, despite their advantages over the injectable route, oral vaccines face many hurdles. A key challenge lies in design of delivery strategies that can protect antigens from degradation in the stomach and intestine, incorporate appropriate immune-stimulatory adjuvants and control release at the appropriate gastrointestinal site. A number of systems including micro and nanoparticles, lipid-based strategies and enteric capsules have significant potential either alone or in advanced combined formulations to enhance intestinal immune responses. In this review we will outline the opportunities, challenges and potential delivery solutions to facilitate the development of improved oral vaccines for infectious enteric diseases.
Collapse
|
23
|
Newsted D, Fallahi F, Golshani A, Azizi A. Advances and challenges in mucosal adjuvant technology. Vaccine 2015; 33:2399-405. [PMID: 25865473 DOI: 10.1016/j.vaccine.2015.03.096] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 03/17/2015] [Accepted: 03/26/2015] [Indexed: 12/23/2022]
Abstract
Adjuvants play attractive roles in enhancement of immune response during vaccination; however, due to several challenges, only a limited number of adjuvants are licensed by health authorities. The lack of an effective mucosal adjuvant is even more significant as none of the licensed adjuvants revealed a strong enhancement in immune system after mucosal administration. Over the past two decades, several mucosal adjuvants have been developed to deliver antigens to the target cells in the mucosal immune system and increase specific immune responses. However, the safety and efficacy of these adjuvants for testing in human trials is still an important issue, requiring further study. In this article, we briefly review the challenges associated with most common mucosal adjuvants and discuss potential strategies for targeting the mucosal immune system.
Collapse
Affiliation(s)
- Daniel Newsted
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St W, Hamilton, ON, Canada
| | | | - Ashkan Golshani
- Department of Biology, Carleton University, 1125 Colonel by Drive, Ottawa, ON, Canada
| | - Ali Azizi
- Department of Pathology and Laboratory Medicine, University of Ottawa, 451 Smyth Rd, Ottawa, ON, Canada; Department of Biotechnology, University of Ontario Institute of Technology, Toronto, ON, Canada.
| |
Collapse
|
24
|
Srivastava A, Gowda DV, Madhunapantula SV, Shinde CG, Iyer M. Mucosal vaccines: a paradigm shift in the development of mucosal adjuvants and delivery vehicles. APMIS 2015; 123:275-88. [PMID: 25630573 DOI: 10.1111/apm.12351] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 11/05/2014] [Indexed: 12/25/2022]
Abstract
Mucosal immune responses are the first-line defensive mechanisms against a variety of infections. Therefore, immunizations of mucosal surfaces from which majority of infectious agents make their entry, helps to protect the body against infections. Hence, vaccinization of mucosal surfaces by using mucosal vaccines provides the basis for generating protective immunity both in the mucosal and systemic immune compartments. Mucosal vaccines offer several advantages over parenteral immunization. For example, (i) ease of administration; (ii) non-invasiveness; (iii) high-patient compliance; and (iv) suitability for mass vaccination. Despite these benefits, to date, only very few mucosal vaccines have been developed using whole microorganisms and approved for use in humans. This is due to various challenges associated with the development of an effective mucosal vaccine that can work against a variety of infections, and various problems concerned with the safe delivery of developed vaccine. For instance, protein antigen alone is not just sufficient enough for the optimal delivery of antigen(s) mucosally. Hence, efforts have been made to develop better prophylactic and therapeutic vaccines for improved mucosal Th1 and Th2 immune responses using an efficient and safe immunostimulatory molecule and novel delivery carriers. Therefore, in this review, we have made an attempt to cover the recent advancements in the development of adjuvants and delivery carriers for safe and effective mucosal vaccine production.
Collapse
Affiliation(s)
- Atul Srivastava
- Department of Pharmaceutics, JSS College of Pharmacy, JSS University, Mysore, India
| | | | | | | | | |
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW Tuberculosis (TB) remains a major health threat that will only be defeated by a combination of better drugs, diagnostics and vaccines. The only licensed TB vaccine, bacille Calmette-Guérin (BCG), protects against extrapulmonary TB in infants. RECENT FINDINGS Novel vaccine candidates that could protect against pulmonary TB either in TB naïve or in latent TB-infected healthy individuals have been developed and are currently being assessed in clinical trials. Subunit booster vaccines are either based on viral vectors expressing TB-specific antigens or on TB-protein antigens in adjuvants. Subunit vaccines are administered on top of BCG. Replacement vaccines for BCG are recombinant viable BCG or Mycobacterium tuberculosis. Several candidates are undergoing, or will soon start, phase IIb assessment for efficacy. The first vaccine candidate, MVA85A, to complete a phase IIb trial, unfortunately failed to show protection against TB in infants. Therapeutic vaccines composed of killed mycobacterial preparations target patients with complicated TB in adjunct to drug treatment. SUMMARY With increasing numbers of TB vaccine candidates in clinical trials, financial, regulatory and infrastructural issues arise, which would be best tackled by a global strategy. In addition, selection of the most promising vaccine candidates for further clinical development gains increasing importance.
Collapse
|
26
|
Portuondo DLF, Ferreira LS, Urbaczek AC, Batista-Duharte A, Carlos IZ. Adjuvants and delivery systems for antifungal vaccines: Current state and future developments. Med Mycol 2014; 53:69-89. [DOI: 10.1093/mmy/myu045] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
|
27
|
Ingvarsson PT, Rasmussen IS, Viaene M, Irlik PJ, Nielsen HM, Foged C. The surface charge of liposomal adjuvants is decisive for their interactions with the Calu-3 and A549 airway epithelial cell culture models. Eur J Pharm Biopharm 2014; 87:480-8. [DOI: 10.1016/j.ejpb.2014.04.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 03/29/2014] [Accepted: 04/02/2014] [Indexed: 12/31/2022]
|
28
|
Abstract
Of all infectious diseases, tuberculosis (TB) remains one of the most important causes of morbidity and mortality. Recent advances in understanding the biology of Mycobacterium tuberculosis (Mtb) infection and the immune response of the infected host have led to the development of several new vaccines, a number of which are already undergoing clinical trials. These include pre-exposure prime vaccines, which could replace bacille Calmette-Guérin (BCG), and pre-exposure booster vaccines given in addition to BCG. Infants are the target population of these two types of vaccines. In addition, several postexposure vaccines given during adolescence or adult life, in addition to BCG as a priming vaccine during infancy, are undergoing clinical testing. Therapeutic vaccines are currently being assessed for their potential to cure active TB as an adjunct to chemotherapy. BCG replacement vaccines are viable recombinant BCG or double-deletion mutants of Mtb. All booster vaccines are composed of one or several antigens, either expressed by viral vectors or formulated with adjuvants. Therapeutic vaccines are killed mycobacterial preparations. Finally, multivariate biomarkers and biosignatures are being generated from high-throughput data with the aim of providing better diagnostic tools to specifically determine TB progression. Here, we provide a technical overview of these recent developments as well of the relevant computational approaches and highlight the obstacles that still need to be overcome.
Collapse
Affiliation(s)
- J Weiner
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | | |
Collapse
|
29
|
Immune Adjuvant Effect of Molecularly-defined Toll-Like Receptor Ligands. Vaccines (Basel) 2014; 2:323-53. [PMID: 26344622 PMCID: PMC4494261 DOI: 10.3390/vaccines2020323] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 03/27/2014] [Accepted: 03/28/2014] [Indexed: 01/07/2023] Open
Abstract
Vaccine efficacy is optimized by addition of immune adjuvants. However, although adjuvants have been used for over a century, to date, only few adjuvants are approved for human use, mostly aimed at improving vaccine efficacy and antigen-specific protective antibody production. The mechanism of action of immune adjuvants is diverse, depending on their chemical and molecular nature, ranging from non-specific effects (i.e., antigen depot at the immunization site) to specific activation of immune cells leading to improved host innate and adaptive responses. Although the detailed molecular mechanism of action of many adjuvants is still elusive, the discovery of Toll-like receptors (TLRs) has provided new critical information on immunostimulatory effect of numerous bacterial components that engage TLRs. These ligands have been shown to improve both the quality and the quantity of host adaptive immune responses when used in vaccine formulations targeted to infectious diseases and cancer that require both humoral and cell-mediated immunity. The potential of such TLR adjuvants in improving the design and the outcomes of several vaccines is continuously evolving, as new agonists are discovered and tested in experimental and clinical models of vaccination. In this review, a summary of the recent progress in development of TLR adjuvants is presented.
Collapse
|
30
|
Progress in tuberculosis vaccine development and host-directed therapies--a state of the art review. THE LANCET RESPIRATORY MEDICINE 2014; 2:301-20. [PMID: 24717627 DOI: 10.1016/s2213-2600(14)70033-5] [Citation(s) in RCA: 163] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Tuberculosis continues to kill 1·4 million people annually. During the past 5 years, an alarming increase in the number of patients with multidrug-resistant tuberculosis and extensively drug-resistant tuberculosis has been noted, particularly in eastern Europe, Asia, and southern Africa. Treatment outcomes with available treatment regimens for drug-resistant tuberculosis are poor. Although substantial progress in drug development for tuberculosis has been made, scientific progress towards development of interventions for prevention and improvement of drug treatment outcomes have lagged behind. Innovative interventions are therefore needed to combat the growing pandemic of multidrug-resistant and extensively drug-resistant tuberculosis. Novel adjunct treatments are needed to accomplish improved cure rates for multidrug-resistant and extensively drug-resistant tuberculosis. A novel, safe, widely applicable, and more effective vaccine against tuberculosis is also desperately sought to achieve disease control. The quest to develop a universally protective vaccine for tuberculosis continues. So far, research and development of tuberculosis vaccines has resulted in almost 20 candidates at different stages of the clinical trial pipeline. Host-directed therapies are now being developed to refocus the anti-Mycobacterium tuberculosis-directed immune responses towards the host; a strategy that could be especially beneficial for patients with multidrug-resistant tuberculosis or extensively drug-resistant tuberculosis. As we are running short of canonical tuberculosis drugs, more attention should be given to host-directed preventive and therapeutic intervention measures.
Collapse
|
31
|
Gebril A, Alsaadi M, Acevedo R, Mullen AB, Ferro VA. Optimizing efficacy of mucosal vaccines. Expert Rev Vaccines 2014; 11:1139-55. [DOI: 10.1586/erv.12.81] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
32
|
Hussain MJ, Wilkinson A, Bramwell VW, Christensen D, Perrie Y. Th1 immune responses can be modulated by varying dimethyldioctadecylammonium and distearoyl-sn-glycero-3-phosphocholine content in liposomal adjuvants. J Pharm Pharmacol 2013; 66:358-66. [DOI: 10.1111/jphp.12173] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 10/10/2013] [Indexed: 01/12/2023]
Abstract
Abstract
Objectives
Cationic liposomes of dimethyldioctadecylammonium bromide (DDA) combined with trehalose 6,6′-dibehenate (TDB) elicit strong cell-mediated and antibody immune responses; DDA facilitates antigen adsorption and presentation while TDB potentiates the immune response. To further investigate the role of DDA, DDA was replaced with the neutral lipid of distearoyl-sn-glycero-3-phosphocholine (DSPC) over a series of concentrations and these systems investigated as adjuvants for the delivery of Ag85B–ESAT-6-Rv2660c, a multistage tuberculosis vaccine.
Methods
Liposomal were prepared at a 5 : 1 DDA–TDB weight ratio and DDA content incrementally replaced with DSPC. The physicochemical characteristics were assessed (vesicle size, zeta potential and antigen loading), and the ability of these systems to act as adjuvants was considered.
Key findings
As DDA was replaced with DSPC within the liposomal formulation, the cationic nature of the vesicles decreases as does electrostatically binding of the anionic H56 antigen (Hybrid56; Ag85B-ESAT6-Rv2660c); however, only when DDA was completed replaced with DSPC did vesicle size increase significantly. T-helper 1 (Th1)-type cell-mediated immune responses reduced. This reduction in responses was attributed to the replacement of DDA with DSPC rather than the reduction in DDA dose concentration within the formulation.
Conclusion
These results suggest Th1 responses can be controlled by tailoring the DDA/DSPC ratio within the liposomal adjuvant system.
Collapse
Affiliation(s)
| | | | | | | | - Yvonne Perrie
- School of Life and Health Sciences, Aston University, Birmingham, UK
| |
Collapse
|
33
|
Neuhaus V, Schwarz K, Klee A, Seehase S, Förster C, Pfennig O, Jonigk D, Fieguth HG, Koch W, Warnecke G, Yusibov V, Sewald K, Braun A. Functional testing of an inhalable nanoparticle based influenza vaccine using a human precision cut lung slice technique. PLoS One 2013; 8:e71728. [PMID: 23967238 PMCID: PMC3742667 DOI: 10.1371/journal.pone.0071728] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 07/02/2013] [Indexed: 12/12/2022] Open
Abstract
Annual outbreaks of influenza infections, caused by new influenza virus subtypes and high incidences of zoonosis, make seasonal influenza one of the most unpredictable and serious health threats worldwide. Currently available vaccines, though the main prevention strategy, can neither efficiently be adapted to new circulating virus subtypes nor provide high amounts to meet the global demand fast enough. New influenza vaccines quickly adapted to current virus strains are needed. In the present study we investigated the local toxicity and capacity of a new inhalable influenza vaccine to induce an antigen-specific recall response at the site of virus entry in human precision-cut lung slices (PCLS). This new vaccine combines recombinant H1N1 influenza hemagglutinin (HAC1), produced in tobacco plants, and a silica nanoparticle (NP)-based drug delivery system. We found no local cellular toxicity of the vaccine within applicable concentrations. However higher concentrations of NP (≥103 µg/ml) dose-dependently decreased viability of human PCLS. Furthermore NP, not the protein, provoked a dose-dependent induction of TNF-α and IL-1β, indicating adjuvant properties of silica. In contrast, we found an antigen-specific induction of the T cell proliferation and differentiation cytokine, IL-2, compared to baseline level (152±49 pg/mg vs. 22±5 pg/mg), which could not be seen for the NP alone. Additionally, treatment with 10 µg/ml HAC1 caused a 6-times higher secretion of IFN-γ compared to baseline (602±307 pg/mg vs. 97±51 pg/mg). This antigen-induced IFN-γ secretion was further boosted by the adjuvant effect of silica NP for the formulated vaccine to a 12-fold increase (97±51 pg/mg vs. 1226±535 pg/mg). Thus we were able to show that the plant-produced vaccine induced an adequate innate immune response and re-activated an established antigen-specific T cell response within a non-toxic range in human PCLS at the site of virus entry.
Collapse
Affiliation(s)
- Vanessa Neuhaus
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Centre for Lung Research (DZL), Hannover, Germany
| | - Katharina Schwarz
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Centre for Lung Research (DZL), Hannover, Germany
| | - Anna Klee
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Centre for Lung Research (DZL), Hannover, Germany
| | - Sophie Seehase
- Research Center Borstel, Leibniz Center for Medicine and Biosciences Airway Reserach Center North (ARCN), Borstel, Germany
- German Center for Lung Research (DZL), Hannover, Germany
| | | | - Olaf Pfennig
- KRH Klinikum Oststadt-Heidehaus, Hannover, Germany
| | - Danny Jonigk
- Institute for Pathology, Hanover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Centre for Lung Research (DZL), Hannover, Germany
| | | | - Wolfgang Koch
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Centre for Lung Research (DZL), Hannover, Germany
| | - Gregor Warnecke
- Division of Cardiac, Thoracic, Transplantation, and Vascular Surgery, Hanover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Centre for Lung Research (DZL), Hannover, Germany
| | - Vidadi Yusibov
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | - Katherina Sewald
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Centre for Lung Research (DZL), Hannover, Germany
| | - Armin Braun
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
- Institute of Immunology, Hanover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Centre for Lung Research (DZL), Hannover, Germany
- * E-mail:
| |
Collapse
|
34
|
Garlapati S. Do we know the Th1/Th2/Th17 determinants of vaccine response? Expert Rev Vaccines 2013; 11:1307-10. [PMID: 23249229 DOI: 10.1586/erv.12.111] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
EVALUATION OF: Kamath AT, Mastelic B, Christensen D et al. Synchronization of dendritic cell activation and antigen exposure is required for the induction of Th1/Th17 responses. J. Immunol. 188(10), 4828–4837 (2012).The determinants of Th1/Th2/Th17 responses elicited by vaccine formulations are largely undefined and are an intense area of research. Most of the present licensed alum-adjuvanted subunit vaccines fail to elicit Th1/Th17 immune responses, and Th2 antibody responses are weak and often require repeated immunizations. Moreover, such responses are not sufficient for eliminating intracellular pathogens. Th1 responses have been traditionally elicited by live-attenuated, vector-based or Toll-like receptor ligand-adjuvanted formulations for optimal stimulation of the innate immune system and immunomodulation. The linkage of adjuvant and antigen (Ag) physically, and/or in a formulation, is essential to overcome systemic effects of the adjuvant and elicit Th1/Th17 responses. The role of delivery systems for codelivery of adjuvant and Ag to the same dendritic cell has gained acceptance. The milieu in which dendritic cells process and present Ag to naive CD4+ T cells determines their polarization into different subsets.
Collapse
Affiliation(s)
- Srinivas Garlapati
- University of Quebec in Montreal, 606-4760 Côte-des-Neiges, Montreal, QC, H3V1G3, Canada.
| |
Collapse
|
35
|
Kaufmann SH. Tuberculosis vaccines: Time to think about the next generation. Semin Immunol 2013; 25:172-81. [DOI: 10.1016/j.smim.2013.04.006] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 03/11/2013] [Accepted: 04/17/2013] [Indexed: 10/26/2022]
|
36
|
Boegh M, Foged C, Müllertz A, Mørck Nielsen H. Mucosal drug delivery: barriers, in vitro models and formulation strategies. J Drug Deliv Sci Technol 2013. [DOI: 10.1016/s1773-2247(13)50055-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
37
|
Kaufmann SHE. Tuberculosis vaccine development: strength lies in tenacity. Trends Immunol 2012; 33:373-9. [PMID: 22560865 DOI: 10.1016/j.it.2012.03.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Revised: 03/21/2012] [Accepted: 03/26/2012] [Indexed: 02/07/2023]
Abstract
The past decade has witnessed a tremendous increase in the development of novel vaccines against tuberculosis (TB). In mice, each of these vaccine candidates stimulates an immune response that reduces the bacillary load, reflecting control but not sterilization of infection. Yet, the immune mechanisms underlying vaccine efficacy are only partially understood. In parallel to clinical assessment of current candidates, the next generation of vaccine candidates still needs to be developed. This requires basic research on how to induce the most efficacious immune response. Equally important is the dissection of immune responses in patients, latently infected healthy individuals, and participants of clinical vaccine trials. Amalgamation of this information will foster the way towards more efficacious vaccination strategies that not only prevent disease, but prevent or abolish infection.
Collapse
Affiliation(s)
- Stefan H E Kaufmann
- Max Planck Institute for Infection Biology, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
38
|
Pedersen G, Cox R. The mucosal vaccine quandary: intranasal vs. sublingual immunization against influenza. Hum Vaccin Immunother 2012; 8:689-93. [PMID: 22495121 DOI: 10.4161/hv.19568] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Intranasal vaccination can effectively induce both local and systemic immune responses and protect against influenza, but poses a risk of antigen or adjuvant delivery into the central nervous system (CNS). Sublingual vaccine delivery has recently received increased attention as a safer alternative to the intranasal route. Studies comparing the two routes have found that higher immune responses may be induced by intranasal than sublingual administration, possibly as a consequence of the differences in mucosal tissues between the two routes. Here we examine evidence of antigen transport into the CNS following intranasal immunisation and discuss possible reasons for the superiority of the intranasal as compared with the sublingual route in terms of vaccine immunogenicity. We encourage generation of more information on the safety of mucosal adjuvants and propose that the next generation of vaccines and adjuvants may be designed specifically for administration via the different mucosal routes.
Collapse
|
39
|
Fact and fiction in tuberculosis vaccine research: 10 years later. THE LANCET. INFECTIOUS DISEASES 2011; 11:633-40. [DOI: 10.1016/s1473-3099(11)70146-3] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
40
|
Rosenkrands I, Vingsbo-Lundberg C, Bundgaard TJ, Lindenstrøm T, Enouf V, van der Werf S, Andersen P, Agger EM. Enhanced humoral and cell-mediated immune responses after immunization with trivalent influenza vaccine adjuvanted with cationic liposomes. Vaccine 2011; 29:6283-91. [PMID: 21722683 DOI: 10.1016/j.vaccine.2011.06.040] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Revised: 06/09/2011] [Accepted: 06/10/2011] [Indexed: 11/18/2022]
Abstract
The recent pandemic caused by new influenza A (H1N1) has emphasized the need for improved influenza vaccines with enhanced immune responses that ideally include longlived humoral and CMI responses and mediate a broad protection. This study demonstrates that administration of trivalent influenza vaccine (TIV) with the cationic liposome adjuvant system CAF01 enhances the humoral immune response as measured by hemagglutinin inhibition titers and influenza-specific serum antibody titers, and promote a strong Th1 response with augmented levels of IL-1β, IL-2, IL-12, IFN-γ and TNF-α. Furthermore, high levels of IL-17 are detected in agreement with CAF01's ability to promote TH17 responses. Importantly, the Th1/Th17 cytokine profile is still maintained 20 weeks after the last vaccination. The CAF01 adjuvanted influenza vaccine reduces weight loss and temperature decrease and results in complete survival of mice challenged with the drifted H1N1 influenza strain A/PR/8/34. Overall, the results suggest that CAF01 is a potent adjuvant system for future, improved influenza vaccines.
Collapse
Affiliation(s)
- Ida Rosenkrands
- Department of Infectious Disease Immunology, Statens Serum Institut, 5 Orestads Boulevard, DK-2300 Copenhagen, Denmark.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Henriksen-Lacey M, Korsholm KS, Andersen P, Perrie Y, Christensen D. Liposomal vaccine delivery systems. Expert Opin Drug Deliv 2011; 8:505-19. [DOI: 10.1517/17425247.2011.558081] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
42
|
Romero EL, Morilla MJ. Topical and mucosal liposomes for vaccine delivery. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2011; 3:356-75. [PMID: 21360692 DOI: 10.1002/wnan.131] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mucosal (and in minor extent transcutanous) stimulation can induce local or distant mucosa secretory IgA. Liposomes and other vesicles as mucosal and transcutaneous adjuvants are attractive alternatives to parenteral vaccination. Liposomes can be massively produced under good manufacturing practices and stored for long periods, at high antigen/vesicle mass ratios. However, their uptake by antigen-presenting cells (APC) at the inductive sites remains as a major challenge. As neurotoxicity is a major concern in intranasal delivery, complexes between archaeosomes and calcium as well as cationic liposomes complexed with plasmids encoding for antigenic proteins could safely elicit secretory and systemic antigen-specific immune responses. Oral bilosomes generate intense immune responses that remain to be tested against challenge, but the admixing with toxins or derivatives is mandatory to reduce the amount of antigen. Most of the current experimental designs, however, underestimate the mucus blanket 100- to 1000-fold thicker than a 100-nm diameter liposome, which has first to be penetrated to access the underlying M cells. Overall, designing mucoadhesive chemoenzymatic resistant liposomes, or selectively targeted to M cells, has produced less relevant results than tailoring the liposomes to make them mucus penetrating. Opposing, the nearly 10 µm thickness stratum corneum interposed between liposomes and underlying APC can be surpassed by ultradeformable liposomes (UDL), with lipid matrices that penetrate up to the limit with the viable epidermis. UDL made of phospholipids and detergents, proved to be better transfection agents than conventional liposomes and niosomes, without the toxicity of ethosomes, in the absence of classical immunomodulators.
Collapse
Affiliation(s)
- Eder Lilia Romero
- Nanomedicine Research Program, Departamento de Ciencia y Tecnologia, Universidad Nacional de Quilmes, Bernal, Argentina.
| | | |
Collapse
|
43
|
|
44
|
|
45
|
Tiwari S, Agrawal GP, Vyas SP. Molecular basis of the mucosal immune system: from fundamental concepts to advances in liposome-based vaccines. Nanomedicine (Lond) 2010; 5:1617-40. [DOI: 10.2217/nnm.10.128] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The mucosal immune system, the primary portal for entry of most prevalent and devastating pathogens, is guarded by the special lymphoid tissues (mucosally associated lymphoid tissues) for immunity. Mucosal immune infection results in induction of IgA-manifested humoral immunity. Cell-mediated immunity may also be generated, marked by the presence of CD4+ Th1 and CD8+ cells. Furthermore, the immunity generated at the mucosal site is transported to the distal mucosal site as well as to systemic tissues. An understanding of the molecular basis of the mucosal immune system provides a unique platform for designing a mucosal vaccine. Coadministration of immunostimulatory molecules further accelerates functioning of the immune system. Mimicking receptor-mediated binding of the pathogen may be achieved by direct conjugation of antigen with an immunostimulatory molecule or encapsulation in a carrier followed by anchoring of a ligand having affinity to the cells of the mucosal immune system. Nanotechnology has played a significant role in mucosal vaccine development and among the available options liposomes are the most promising. Liposomes are phospholipid bilayered vesicles that can encapsulate protein as well as DNA-based vaccines and offer coencapsulation of adjuvant along with the antigen. At the same, time ligand-conjugated liposomes augment interaction of antigen with the cells of the mucosal immune system and thereby serve as suitable candidates for the mucosal delivery of vaccines. This article exhaustively explores strategies involved in the generation of mucosal immunity and also provides an insight to the progress that has been made in the development of liposome-based mucosal vaccine.
Collapse
Affiliation(s)
- Shailja Tiwari
- Drug Delivery Research Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, Madhya Pradesh 470003, India
| | - Govind P Agrawal
- Drug Delivery Research Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, Madhya Pradesh 470003, India
| | | |
Collapse
|
46
|
Slütter B, Bal SM, Que I, Kaijzel E, Löwik C, Bouwstra J, Jiskoot W. Antigen−Adjuvant Nanoconjugates for Nasal Vaccination: An Improvement over the Use of Nanoparticles? Mol Pharm 2010; 7:2207-15. [DOI: 10.1021/mp100210g] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Bram Slütter
- Division of Drug Delivery Technology, Leiden/Amsterdam Center for Drug Research (LACDR), Leiden University, Leiden, The Netherlands, and Department of Endocrinology and Metabolic Diseases, Leiden University Medical Centre, Leiden, The Netherlands
| | - Suzanne M. Bal
- Division of Drug Delivery Technology, Leiden/Amsterdam Center for Drug Research (LACDR), Leiden University, Leiden, The Netherlands, and Department of Endocrinology and Metabolic Diseases, Leiden University Medical Centre, Leiden, The Netherlands
| | - Ivo Que
- Division of Drug Delivery Technology, Leiden/Amsterdam Center for Drug Research (LACDR), Leiden University, Leiden, The Netherlands, and Department of Endocrinology and Metabolic Diseases, Leiden University Medical Centre, Leiden, The Netherlands
| | - Eric Kaijzel
- Division of Drug Delivery Technology, Leiden/Amsterdam Center for Drug Research (LACDR), Leiden University, Leiden, The Netherlands, and Department of Endocrinology and Metabolic Diseases, Leiden University Medical Centre, Leiden, The Netherlands
| | - Clemens Löwik
- Division of Drug Delivery Technology, Leiden/Amsterdam Center for Drug Research (LACDR), Leiden University, Leiden, The Netherlands, and Department of Endocrinology and Metabolic Diseases, Leiden University Medical Centre, Leiden, The Netherlands
| | - Joke Bouwstra
- Division of Drug Delivery Technology, Leiden/Amsterdam Center for Drug Research (LACDR), Leiden University, Leiden, The Netherlands, and Department of Endocrinology and Metabolic Diseases, Leiden University Medical Centre, Leiden, The Netherlands
| | - Wim Jiskoot
- Division of Drug Delivery Technology, Leiden/Amsterdam Center for Drug Research (LACDR), Leiden University, Leiden, The Netherlands, and Department of Endocrinology and Metabolic Diseases, Leiden University Medical Centre, Leiden, The Netherlands
| |
Collapse
|
47
|
Heurtault B, Frisch B, Pons F. Liposomes as delivery systems for nasal vaccination: strategies and outcomes. Expert Opin Drug Deliv 2010; 7:829-44. [PMID: 20459361 DOI: 10.1517/17425247.2010.488687] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
IMPORTANCE OF THE FIELD Among the particulate systems that have been envisaged in vaccine delivery, liposomes are very attractive. These phospholipid vesicles can indeed deliver a wide range of molecules. They have been shown to enhance considerably the immunogenicity of weak protein antigens or synthetic peptides. Also, they offer a wide range of pharmaceutical options for the design of vaccines. In the past decade, the nasal mucosa has emerged as an effective route for vaccine delivery, together with the opportunity to develop non-invasive approaches in vaccination. AREAS COVERED IN THIS REVIEW This review focuses on the recent strategies and outcomes that have been developed around the use of liposomes in nasal vaccination. WHAT THE READER WILL GAIN The various formulation parameters, including lipid composition, size, charge and mucoadhesiveness, that have been investigated in the design of liposomal vaccine candidates dedicated to nasal vaccination are outlined. Also, an overview of the immunological and protective responses obtained with the developed formulations is presented. TAKE HOME MESSAGE This review illustrates the high potential of liposomes as nasal vaccine delivery systems.
Collapse
Affiliation(s)
- Béatrice Heurtault
- Equipe de Biovectorologie, Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199 CNRS/Université de Strasbourg, Faculté de Pharmacie, 74, route du Rhin, 67401 Illkirch Cedex, France.
| | | | | |
Collapse
|