1
|
Domínguez-Odio A, Rodríguez-Martínez E, Paneque Zayas M, Cala-Delgado DL. Research publications and global manufacture of veterinary vaccines against avian influenza A (2019-2023). Front Vet Sci 2025; 12:1394675. [PMID: 40144520 PMCID: PMC11936895 DOI: 10.3389/fvets.2025.1394675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 02/10/2025] [Indexed: 03/28/2025] Open
Abstract
The characteristics of the avian influenza virus and its worldwide spread have led to intense and unprecedented scientific activity and industrial production for preventive veterinary vaccines. However, knowledge gaps remain regarding the best strategies to prevent epidemiological events in the future. In this context, the present study aimed to provide a global analysis on the scientific and industrial production of avian influenza type A vaccines for farm animals and pets during the period 2019 2023. The Scopus database was used as the primary source of information (12,162 keywords, 2,437 scientific articles, 659 academic journals, and 46 countries) for the academic analysis, while technical information posted on official institutional websites (136 commercial formulations, 24 vaccines manufacturers, and 17 countries) was collected to conduct the industrial analysis. 3,045, 25.0%) exhibited the highest levels of co-occurrence in the sciences; the journal Vaccine was the most productive in terms of articles (11.8%, 288/2,437), and the countries with the most publications were the USA (25.5%, 622/2,437) and China (23.1%, 564/2,437). The most internationally marketed vaccines were inactivated (86.0%, 117/136), avian (47.1%, 64/136), and combined (52.2%, 71/136) vaccines as well as those containing Newcastle antigens (38.0%, 27/71). In conclusion, the study demonstrated the fundamental role of classical production methods (based on the use of the whole pathogen) in avian influenza A research and the production of veterinary vaccines.
Collapse
|
2
|
Tang S, Zhao C, Zhu X. Engineering Escherichia coli-Derived Nanoparticles for Vaccine Development. Vaccines (Basel) 2024; 12:1287. [PMID: 39591189 PMCID: PMC11598912 DOI: 10.3390/vaccines12111287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/13/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
The development of effective vaccines necessitates a delicate balance between maximizing immunogenicity and minimizing safety concerns. Subunit vaccines, while generally considered safe, often fail to elicit robust and durable immune responses. Nanotechnology presents a promising approach to address this dilemma, enabling subunit antigens to mimic critical aspects of native pathogens, such as nanoscale dimensions, geometry, and highly repetitive antigen display. Various expression systems, including Escherichia coli (E. coli), yeast, baculovirus/insect cells, and Chinese hamster ovary (CHO) cells, have been explored for the production of nanoparticle vaccines. Among these, E. coli stands out due to its cost-effectiveness, scalability, rapid production cycle, and high yields. However, the E. coli manufacturing platform faces challenges related to its unfavorable redox environment for disulfide bond formation, lack of post-translational modifications, and difficulties in achieving proper protein folding. This review focuses on molecular and protein engineering strategies to enhance protein solubility in E. coli and facilitate the in vitro reassembly of virus-like particles (VLPs). We also discuss approaches for antigen display on nanocarrier surfaces and methods to stabilize these carriers. These bioengineering approaches, in combination with advanced nanocarrier design, hold significant potential for developing highly effective and affordable E. coli-derived nanovaccines, paving the way for improved protection against a wide range of infectious diseases.
Collapse
Affiliation(s)
- Shubing Tang
- Shanghai Reinovax Biologics Co., Ltd., Pudong New District, Shanghai 200135, China;
| | - Chen Zhao
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201058, China
| | - Xianchao Zhu
- Shanghai Reinovax Biologics Co., Ltd., Pudong New District, Shanghai 200135, China;
| |
Collapse
|
3
|
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] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 04/01/2024] [Accepted: 04/08/2024] [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)
| | | | | | - 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
|
4
|
Mixová G, Tihlaříková E, Zhu Y, Schindler L, Androvič L, Kracíková L, Hrdá E, Porsch B, Pechar M, Garliss CM, Wilson D, Welles HC, Holechek J, Ren Q, Lynn GM, Neděla V, Laga R. Synthesis and Structure Optimization of Star Copolymers as Tunable Macromolecular Carriers for Minimal Immunogen Vaccine Delivery. Bioconjug Chem 2024; 35:1218-1232. [PMID: 39081220 PMCID: PMC11342300 DOI: 10.1021/acs.bioconjchem.4c00273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/25/2024] [Accepted: 07/25/2024] [Indexed: 08/22/2024]
Abstract
Minimal immunogen vaccines are being developed to focus antibody responses against otherwise challenging targets, including human immunodeficiency virus (HIV), but multimerization of the minimal peptide immunogen on a carrier platform is required for activity. Star copolymers comprising multiple hydrophilic polymer chains ("arms") radiating from a central dendrimer unit ("core") were recently reported to be an effective platform for arraying minimal immunogens for inducing antibody responses in mice and primates. However, the impact of different parameters of the star copolymer (e.g., minimal immunogen density and hydrodynamic size) on antibody responses and the optimal synthetic route for controlling those parameters remains to be fully explored. We synthesized a library of star copolymers composed of poly[N-(2-hydroxypropyl)methacrylamide] hydrophilic arms extending from poly(amidoamine) dendrimer cores with the aim of identifying the optimal composition for use as minimal immunogen vaccines. Our results show that the length of the polymer arms has a crucial impact on the star copolymer hydrodynamic size and is precisely tunable over a range of 20-50 nm diameter, while the dendrimer generation affects the maximum number of arms (and therefore minimal immunogens) that can be attached to the surface of the dendrimer. In addition, high-resolution images of selected star copolymer taken by a custom-modified environmental scanning electron microscope enabled the acquisition of high-resolution images, providing new insights into the star copolymer structure. Finally, in vivo studies assessing a star copolymer vaccine comprising an HIV minimal immunogen showed the criticality of polymer arm length in promoting antibody responses and highlighting the importance of composition tunability to yield the desired biological effect.
Collapse
Affiliation(s)
- Gabriela Mixová
- Institute
of Macromolecular Chemistry, Czech Academy
of Sciences, Heyrovského
nám. 2, Prague 162
06, Czech Republic
| | - Eva Tihlaříková
- Institute
of Scientific Instruments, Czech Academy
of Sciences, Královopolská
147, Brno 612 64, Czech Republic
| | - Yaling Zhu
- Barinthus
Biotherapeutics North America, Inc. (formerly Avidea Technologies,
Inc.), 20400 Century
Boulevard, Germantown, Maryland 20874, United States
| | - Lucie Schindler
- Institute
of Macromolecular Chemistry, Czech Academy
of Sciences, Heyrovského
nám. 2, Prague 162
06, Czech Republic
| | - Ladislav Androvič
- Institute
of Macromolecular Chemistry, Czech Academy
of Sciences, Heyrovského
nám. 2, Prague 162
06, Czech Republic
| | - Lucie Kracíková
- Institute
of Macromolecular Chemistry, Czech Academy
of Sciences, Heyrovského
nám. 2, Prague 162
06, Czech Republic
| | - Eliška Hrdá
- Institute
of Macromolecular Chemistry, Czech Academy
of Sciences, Heyrovského
nám. 2, Prague 162
06, Czech Republic
| | - Bedřich Porsch
- Institute
of Macromolecular Chemistry, Czech Academy
of Sciences, Heyrovského
nám. 2, Prague 162
06, Czech Republic
| | - Michal Pechar
- Institute
of Macromolecular Chemistry, Czech Academy
of Sciences, Heyrovského
nám. 2, Prague 162
06, Czech Republic
| | - Christopher M. Garliss
- Barinthus
Biotherapeutics North America, Inc. (formerly Avidea Technologies,
Inc.), 20400 Century
Boulevard, Germantown, Maryland 20874, United States
| | - David Wilson
- Barinthus
Biotherapeutics North America, Inc. (formerly Avidea Technologies,
Inc.), 20400 Century
Boulevard, Germantown, Maryland 20874, United States
| | - Hugh C. Welles
- Barinthus
Biotherapeutics North America, Inc. (formerly Avidea Technologies,
Inc.), 20400 Century
Boulevard, Germantown, Maryland 20874, United States
| | - Jake Holechek
- Barinthus
Biotherapeutics North America, Inc. (formerly Avidea Technologies,
Inc.), 20400 Century
Boulevard, Germantown, Maryland 20874, United States
| | - Qiuyin Ren
- Vaccine
Research Center, National Institutes of
Health, Rockville, Maryland 20892, United States
| | - Geoffrey M. Lynn
- Barinthus
Biotherapeutics North America, Inc. (formerly Avidea Technologies,
Inc.), 20400 Century
Boulevard, Germantown, Maryland 20874, United States
| | - Vilém Neděla
- Institute
of Scientific Instruments, Czech Academy
of Sciences, Královopolská
147, Brno 612 64, Czech Republic
| | - Richard Laga
- Institute
of Macromolecular Chemistry, Czech Academy
of Sciences, Heyrovského
nám. 2, Prague 162
06, Czech Republic
| |
Collapse
|
5
|
Cui Y, Ho M, Hu Y, Shi Y. Vaccine adjuvants: current status, research and development, licensing, and future opportunities. J Mater Chem B 2024; 12:4118-4137. [PMID: 38591323 PMCID: PMC11180427 DOI: 10.1039/d3tb02861e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
Vaccines represent one of the most significant inventions in human history and have revolutionized global health. Generally, a vaccine functions by triggering the innate immune response and stimulating antigen-presenting cells, leading to a defensive adaptive immune response against a specific pathogen's antigen. As a key element, adjuvants are chemical materials often employed as additives to increase a vaccine's efficacy and immunogenicity. For over 90 years, adjuvants have been essential components in many human vaccines, improving their efficacy by enhancing, modulating, and prolonging the immune response. Here, we provide a timely and comprehensive review of the historical development and the current status of adjuvants, covering their classification, mechanisms of action, and roles in different vaccines. Additionally, we perform systematic analysis of the current licensing processes and highlights notable examples from clinical trials involving vaccine adjuvants. Looking ahead, we anticipate future trends in the field, including the development of new adjuvant formulations, the creation of innovative adjuvants, and their integration into the broader scope of systems vaccinology and vaccine delivery. The article posits that a deeper understanding of biochemistry, materials science, and vaccine immunology is crucial for advancing vaccine technology. Such advancements are expected to lead to the future development of more effective vaccines, capable of combating emerging infectious diseases and enhancing public health.
Collapse
Affiliation(s)
- Ying Cui
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, CA 90095, USA.
| | - Megan Ho
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Yongjie Hu
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, CA 90095, USA.
| | - Yuan Shi
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
6
|
Zhang Y, Tian L, Zhao X, Jiang X, Qin J, Wang Y, Yu X. Enhanced protective efficacy of an OprF/PcrV bivalent DNA vaccine against Pseudomonas aeruginosa using a hydrogel delivery system. Biomed Pharmacother 2024; 172:116264. [PMID: 38359491 DOI: 10.1016/j.biopha.2024.116264] [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: 11/29/2023] [Revised: 01/19/2024] [Accepted: 02/06/2024] [Indexed: 02/17/2024] Open
Abstract
Pseudomonas aeruginosa (PA) is one of the leading pathogens responsible for hospital-acquired infections. With the increasing antibiotic resistance of PA, clinical treatment has become increasingly challenging. DNA vaccines represent a promising approach for combating PA infection. However, the immune response induced by a single antigen is limited, and combination vaccines hold greater therapeutic potential. The highly conserved OprF and PcrV genes are attractive candidate antigens for vaccine development, but the poor delivery of such vaccines has limited their clinical application. In this study, we constructed an OprF/PcrV bivalent DNA vaccine, and a polyaspartamide/polyethylene glycol di-aldehyde (PSIH/PEG DA) hydrogel was formulated to improve DNA delivery. The OprF/PcrV DNA vaccine formulated with the PSIH/PEG DA hydrogel was carefully characterized in vitro and in vivo and showed suitable compatibility. The PSIH/PEG DA hydrogel formulation induced a mixed Th1/Th2/Th17 immune response in mice, leading to a significant increase in antibody titers, lymphocyte proliferation rates, and cytokine levels compared to those in mice treated with single or combined vaccines. The PSIH/PEG DA hydrogel delivery system significantly enhanced the immune protection of the DNA vaccine in a murine pneumonia model, as revealed by the reduced bacterial burden and inflammation in the mouse lungs and increased survival rate. In conclusion, the PSIH/PEG DA hydrogel delivery system can further enhance the immune efficacy of the combination OprF/PcrV DNA vaccine. This research provides a novel optimized strategy for the prevention and treatment of PA infection using DNA vaccines.
Collapse
Affiliation(s)
- Yating Zhang
- Phase I Clinical Trial Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Linxia Tian
- Phase I Clinical Trial Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Xuan Zhao
- Phase I Clinical Trial Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - XiaoFeng Jiang
- Phase I Clinical Trial Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Jianglei Qin
- Key Laboratory of Pathogenesis Mechanism and Control of Inflammatory-Autoimmune Diseases in Hebei Province, Baoding 071002, China
| | - Yong Wang
- Key Laboratory of Pathogenesis Mechanism and Control of Inflammatory-Autoimmune Diseases in Hebei Province, Baoding 071002, China
| | - Xian Yu
- Phase I Clinical Trial Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| |
Collapse
|
7
|
Miranda MCR, Nunes CM, Santos LF, da Silva LB, de Jesus VR, Filho NA, Pedro JAF, Lopes JLS, Oliveira CLP, Fantini MCA, Cardoso JS, Trezena AG, Ribeiro OG, Sant'Anna OA, Tino-De-Franco M, Martins TS. Ordered mesoporous silicas for potential applications in solid vaccine formulations. Vaccine 2024; 42:689-700. [PMID: 38145911 DOI: 10.1016/j.vaccine.2023.12.032] [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: 09/02/2023] [Revised: 12/06/2023] [Accepted: 12/08/2023] [Indexed: 12/27/2023]
Abstract
In an effort to develop efficient vaccine formulations, the use of ordered mesoporous silica (SBA-15) as an antigen carrier has been investigated. SBA-15 has required properties such as high surface area and pore volume, including narrow pore size distribution to protect antigens inside its matrix. This study aimed to examine the impact of solvent removal methods, specifically freeze-drying and evaporation on the intrinsic properties of an immunogenic complex. The immunogenic complexes, synthesized and incorporated with BSA, were characterized by various physicochemical techniques. Small Angle X-ray Scattering measurements revealed the characteristic reflections associated to pure SBA-15, indicating the preservation of the silica mesostructured following BSA incorporation and the formation of BSA aggregates within the macropore region. Nitrogen Adsorption Isotherm measurements demonstrated a decrease in surface area and pore volume for all samples, indicating that the BSA was incorporated into the SBA-15 matrix. Fluorescence spectroscopy evidenced that the tryptophan residues in BSA inside SBA-15 or in solution displayed similar spectra, showing the preservation of the aromatic residues' environment. The Circular Dichroism spectra of BSA in both conditions suggest the preservation of its native secondary structure after the encapsulation process. The immunogenic analysis with the detection of anti-BSA IgG did not give any significant difference between the non-dried, freeze-dried or evaporated groups. However, all groups containing BSA and SBA-15 showed results almost three times higher than the groups with pure BSA (control group). These facts indicate that none of the BSA incorporation methods interfered with the immunogenicity of the complex. In particular, the freeze-dried process is regularly used in the pharmaceutical industry, therefore its adequacy to produce immunogenic complexes was proved Furthermore, the results showed that SBA-15 increased the immunogenic activity of BSA.
Collapse
Affiliation(s)
- Matheus C R Miranda
- Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Universidade Federal de São Paulo, Diadema, SP, Brazil
| | - Carmen M Nunes
- Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Universidade Federal de São Paulo, Diadema, SP, Brazil
| | - Luana F Santos
- Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Universidade Federal de São Paulo, Diadema, SP, Brazil
| | - Leonardo B da Silva
- Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Universidade Federal de São Paulo, Diadema, SP, Brazil
| | - Vinicius R de Jesus
- Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Universidade Federal de São Paulo, Diadema, SP, Brazil
| | - Newton Andréo Filho
- Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Universidade Federal de São Paulo, Diadema, SP, Brazil
| | - Jéssica A F Pedro
- Instituto de Física, Universidade de São Paulo, São Paulo, SP, Brazil
| | - José L S Lopes
- Departamento de Física, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
| | | | | | | | | | | | | | | | - Tereza S Martins
- Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Universidade Federal de São Paulo, Diadema, SP, Brazil.
| |
Collapse
|
8
|
Song S, Wang S, Jiang X, Yang F, Gao S, Lin X, Cheng H, van der Veen S. Th1-polarized MtrE-based gonococcal vaccines display prophylactic and therapeutic efficacy. Emerg Microbes Infect 2023; 12:2249124. [PMID: 37584947 PMCID: PMC10467530 DOI: 10.1080/22221751.2023.2249124] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/10/2023] [Accepted: 08/13/2023] [Indexed: 08/17/2023]
Abstract
ABSTRACTGlobal dissemination of high-level ceftriaxone-resistant Neisseria gonorrhoeae strains associated with the FC428 clone poses a threat to the efficacy ceftriaxone-based therapies. Vaccination is the best strategy to contain multidrug-resistant infections. In this study, we investigated the efficacy of MtrE and its surface Loop2 as vaccine antigens when combined with a Th1-polarizing adjuvant, which is expected to be beneficial for gonococcal vaccine development. Using in vitro dendritic cell maturation and T cell differentiation assays, CpG1826 was identified as the optimal Th1-polarizing adjuvant for MtrE and Loop2 displayed as linear epitope (Nloop2) or structural epitope (Intraloop2) on a carrier protein. Loop2-based antigens raised strongly Th1-polarized and bactericidal antibody responses in vaccinated mice. Furthermore, the vaccine formulations provided protection against a gonococcal challenge in mouse vaginal tract infection model when provided as prophylactic vaccines. Also, the vaccine formulations accelerated gonococcal clearance when provided as a single therapeutic dose to treat an already established infection, including against a strain associated with the FC428 clone. Therefore, this study demonstrated that MtrE and Loop 2 are effective gonococcal vaccine antigens when combined with the Th1-polarizing CpG1826 adjuvant.
Collapse
Affiliation(s)
- Shuaijie Song
- Department of Microbiology, and Department of Dermatology of Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Shuyi Wang
- Department of Microbiology, and Department of Dermatology of Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Xiaoyun Jiang
- Department of Microbiology, and Department of Dermatology of Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Fan Yang
- Department of Microbiology, and Department of Dermatology of Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Shuai Gao
- Department of Microbiology, and Department of Dermatology of Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Xu’ai Lin
- Department of Microbiology, and Department of Dermatology of Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Hao Cheng
- Department of Microbiology, and Department of Dermatology of Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Stijn van der Veen
- Department of Microbiology, and Department of Dermatology of Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
- Zhejiang Provincial Key Laboratory for Microbial Biochemistry and Metabolic Engineering, Hangzhou, People’s Republic of China
| |
Collapse
|
9
|
Hou Y, Chen M, Bian Y, Zheng X, Tong R, Sun X. Advanced subunit vaccine delivery technologies: From vaccine cascade obstacles to design strategies. Acta Pharm Sin B 2023; 13:3321-3338. [PMID: 37655334 PMCID: PMC10465871 DOI: 10.1016/j.apsb.2023.01.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 11/23/2022] [Accepted: 12/03/2022] [Indexed: 01/12/2023] Open
Abstract
Designing and manufacturing safe and effective vaccines is a crucial challenge for human health worldwide. Research on adjuvant-based subunit vaccines is increasingly being explored to meet clinical needs. Nevertheless, the adaptive immune responses of subunit vaccines are still unfavorable, which may partially be attributed to the immune cascade obstacles and unsatisfactory vaccine design. An extended understanding of the crosstalk between vaccine delivery strategies and immunological mechanisms could provide scientific insight to optimize antigen delivery and improve vaccination efficacy. In this review, we summarized the advanced subunit vaccine delivery technologies from the perspective of vaccine cascade obstacles after administration. The engineered subunit vaccines with lymph node and specific cell targeting ability, antigen cross-presentation, T cell activation properties, and tailorable antigen release patterns may achieve effective immune protection with high precision, efficiency, and stability. We hope this review can provide rational design principles and inspire the exploitation of future subunit vaccines.
Collapse
Affiliation(s)
- Yingying Hou
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Min Chen
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Yuan Bian
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Xi Zheng
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Rongsheng Tong
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Xun Sun
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
10
|
Mao L, Ma P, Luo X, Cheng H, Wang Z, Ye E, Loh XJ, Wu YL, Li Z. Stimuli-Responsive Polymeric Nanovaccines Toward Next-Generation Immunotherapy. ACS NANO 2023; 17:9826-9849. [PMID: 37207347 DOI: 10.1021/acsnano.3c02273] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
The development of nanovaccines that employ polymeric delivery carriers has garnered substantial interest in therapeutic treatment of cancer and a variety of infectious diseases due to their superior biocompatibility, lower toxicity and reduced immunogenicity. Particularly, stimuli-responsive polymeric nanocarriers show great promise for delivering antigens and adjuvants to targeted immune cells, preventing antigen degradation and clearance, and increasing the uptake of specific antigen-presenting cells, thereby sustaining adaptive immune responses and improving immunotherapy for certain diseases. In this review, the most recent advances in the utilization of stimulus-responsive polymer-based nanovaccines for immunotherapeutic applications are presented. These sophisticated polymeric nanovaccines with diverse functions, aimed at therapeutic administration for disease prevention and immunotherapy, are further classified into several active domains, including pH, temperature, redox, light and ultrasound-sensitive intelligent nanodelivery systems. Finally, the potential strategies for the future design of multifunctional next-generation polymeric nanovaccines by integrating materials science with biological interface are proposed.
Collapse
Affiliation(s)
- Liuzhou Mao
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Panqin Ma
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Xi Luo
- BE/Phase I Clinical Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361000, China
| | - Hongwei Cheng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Zhanxiang Wang
- BE/Phase I Clinical Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361000, China
| | - Enyi Ye
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE2), Agency for Science, Technology and Research (A*STAR), 1 Pesek Road, Jurong Island, Singapore 627833, Republic of Singapore
| | - Xian Jun Loh
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE2), Agency for Science, Technology and Research (A*STAR), 1 Pesek Road, Jurong Island, Singapore 627833, Republic of Singapore
| | - Yun-Long Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Zibiao Li
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE2), Agency for Science, Technology and Research (A*STAR), 1 Pesek Road, Jurong Island, Singapore 627833, Republic of Singapore
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Republic of Singapore
| |
Collapse
|
11
|
Wu N, Chen Q, Zou Y, Miao C, Ma G, Wu J. Chitosan particle-emulsion complex adjuvants: The effect of particle distribution on the immune intensity and response type. Carbohydr Polym 2023; 309:120673. [PMID: 36906359 DOI: 10.1016/j.carbpol.2023.120673] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/09/2023]
Abstract
Particle-emulsion complex adjuvants as a new trend in the research of vaccine formulation, can improve the immune strength and balance the immune type. However, the location of the particle in the formulation is a key factor that has not been investigated extensively and its type of immunity. In order to investigate the effect of different combining modes of emulsion and particle on the immune response, three types of particle-emulsion complex adjuvant formulations were designed with the combination of chitosan nanoparticles (CNP) and an o/w emulsion with squalene as the oil phase. The complex adjuvants included the CNP-I group (particle inside the emulsion droplet), CNP-S group (particle on the surface of emulsion droplet) and CNP-O group (particle outside the emulsion droplet), respectively. The formulations with different particle locations behaved with different immunoprotective effects and immune-enhancing mechanisms. Compared with CNP-O, CNP-I and CNP-S significantly improve humoral and cellular immunity. CNP-O was more like two independent systems for immune enhancement. As a result, CNP-S triggered a Th1-type immune bias and CNP-I had more of a Th2-type of the immune response. These data highlight the key influence of the subtle difference of particle location in the droplets for immune response.
Collapse
Affiliation(s)
- Nan Wu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Qiuting Chen
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China; School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou 510006, PR China
| | - Yongjuan Zou
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Chunyu Miao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Guanghui Ma
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Jie Wu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China.
| |
Collapse
|
12
|
Ananya A, Holden KG, Gu Z, Nettleton D, Mallapragada SK, Wannemuehler MJ, Kohut ML, Narasimhan B. "Just right" combinations of adjuvants with nanoscale carriers activate aged dendritic cells without overt inflammation. Immun Ageing 2023; 20:10. [PMID: 36895007 PMCID: PMC9996592 DOI: 10.1186/s12979-023-00332-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 02/05/2023] [Indexed: 03/11/2023]
Abstract
BACKGROUND The loss in age-related immunological markers, known as immunosenescence, is caused by a combination of factors, one of which is inflammaging. Inflammaging is associated with the continuous basal generation of proinflammatory cytokines. Studies have demonstrated that inflammaging reduces the effectiveness of vaccines. Strategies aimed at modifying baseline inflammation are being developed to improve vaccination responses in older adults. Dendritic cells have attracted attention as an age-specific target because of their significance in immunization as antigen presenting cells that stimulate T lymphocytes. RESULTS In this study, bone marrow derived dendritic cells (BMDCs) were generated from aged mice and used to investigate the effects of combinations of adjuvants, including Toll-like receptor, NOD2, and STING agonists with polyanhydride nanoparticles and pentablock copolymer micelles under in vitro conditions. Cellular stimulation was characterized via expression of costimulatory molecules, T cell-activating cytokines, proinflammatory cytokines, and chemokines. Our results indicate that multiple TLR agonists substantially increase costimulatory molecule expression and cytokines associated with T cell activation and inflammation in culture. In contrast, NOD2 and STING agonists had only a moderate effect on BMDC activation, while nanoparticles and micelles had no effect by themselves. However, when nanoparticles and micelles were combined with a TLR9 agonist, a reduction in the production of proinflammatory cytokines was observed while maintaining increased production of T cell activating cytokines and enhancing cell surface marker expression. Additionally, combining nanoparticles and micelles with a STING agonist resulted in a synergistic impact on the upregulation of costimulatory molecules and an increase in cytokine secretion from BMDCs linked with T cell activation without excessive secretion of proinflammatory cytokines. CONCLUSIONS These studies provide new insights into rational adjuvant selection for vaccines for older adults. Combining appropriate adjuvants with nanoparticles and micelles may lead to balanced immune activation characterized by low inflammation, setting the stage for designing next generation vaccines that can induce mucosal immunity in older adults.
Collapse
Affiliation(s)
- Ananya Ananya
- Nanovaccine Institute, Iowa State University, Ames, IA, 50011, USA
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, 50011, USA
| | - Kaitlyn G Holden
- Nanovaccine Institute, Iowa State University, Ames, IA, 50011, USA
| | - Zhiling Gu
- Department of Statistics, Iowa State University, Ames, IA, 50011, USA
| | - Dan Nettleton
- Department of Statistics, Iowa State University, Ames, IA, 50011, USA
| | - Surya K Mallapragada
- Nanovaccine Institute, Iowa State University, Ames, IA, 50011, USA
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, 50011, USA
| | | | - Marian L Kohut
- Nanovaccine Institute, Iowa State University, Ames, IA, 50011, USA
- Department of Kinesiology, Iowa State University, Ames, IA, 50011, USA
| | - Balaji Narasimhan
- Nanovaccine Institute, Iowa State University, Ames, IA, 50011, USA.
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, 50011, USA.
| |
Collapse
|
13
|
Verma SK, Mahajan P, Singh NK, Gupta A, Aggarwal R, Rappuoli R, Johri AK. New-age vaccine adjuvants, their development, and future perspective. Front Immunol 2023; 14:1043109. [PMID: 36911719 PMCID: PMC9998920 DOI: 10.3389/fimmu.2023.1043109] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 01/26/2023] [Indexed: 02/26/2023] Open
Abstract
In the present scenario, immunization is of utmost importance as it keeps us safe and protects us from infectious agents. Despite the great success in the field of vaccinology, there is a need to not only develop safe and ideal vaccines to fight deadly infections but also improve the quality of existing vaccines in terms of partial or inconsistent protection. Generally, subunit vaccines are known to be safe in nature, but they are mostly found to be incapable of generating the optimum immune response. Hence, there is a great possibility of improving the potential of a vaccine in formulation with novel adjuvants, which can effectively impart superior immunity. The vaccine(s) in formulation with novel adjuvants may also be helpful in fighting pathogens of high antigenic diversity. However, due to the limitations of safety and toxicity, very few human-compatible adjuvants have been approved. In this review, we mainly focus on the need for new and improved vaccines; the definition of and the need for adjuvants; the characteristics and mechanisms of human-compatible adjuvants; the current status of vaccine adjuvants, mucosal vaccine adjuvants, and adjuvants in clinical development; and future directions.
Collapse
Affiliation(s)
| | - Pooja Mahajan
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Nikhlesh K. Singh
- Integrative Biosciences Center, Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, School of Medicine, Detroit, MI, United States
| | - Ankit Gupta
- Microbiology Division, Defence Research and Development Establishment, Gwalior, India
| | - Rupesh Aggarwal
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | | | - Atul Kumar Johri
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
14
|
Ma J, Wang S, Zhao C, Yan X, Ren Q, Dong Z, Qiu J, Liu Y, Shan Q, Xu M, Yan B, Liu S. Computer-Aided Discovery of Potent Broad-Spectrum Vaccine Adjuvants. Angew Chem Int Ed Engl 2023; 62:e202301059. [PMID: 36815280 DOI: 10.1002/anie.202301059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/21/2023] [Accepted: 02/22/2023] [Indexed: 02/24/2023]
Abstract
Adjuvants stimulate the immune system to vigorously respond to a vaccine. While current adjuvants such as aluminum salts and oil-in-water emulsions have been used for decades, they do not generate broad and long-lasting responses in many vaccines. Consequently, more potent adjuvants are needed. Here, using computer-aided molecule design and machine learning, we discovered 2 new, broad-spectrum adjuvants that can boost vaccine responses. Our library containing 46 toll-like receptor (TLR)-targeting agonist ligands were assembled on Au nanoparticles. Comprehensive in vitro, ex vivo and in vivo studies showed both leads promoted dendritic cell activation via multiple TLRs and enhanced antigen presentation to T cells. When used together with tumor-specific antigens to immunize mice against B16-OVA melanoma and 4T1-PD1 breast cancer, both adjuvants unleashed strong immune responses that suppressed tumor growth and lung metastases. Our results show computer-aided design and screening can rapidly uncover potent adjuvants for tackling waning immunity in current vaccines.
Collapse
Affiliation(s)
- Juan Ma
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, P. R. China
- School of Environmental Sciences, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Shenqing Wang
- Institute of Environmental Research at Greater Bays, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou, 510006, P. R. China
| | - Chuanfang Zhao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, P. R. China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Xiliang Yan
- Institute of Environmental Research at Greater Bays, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou, 510006, P. R. China
| | - Quanzhong Ren
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, P. R. China
- Department of toxicology and sanitary chemistry, School of public health, Capital Medical University, Beijing, 100069, P. R. China
| | - Zheng Dong
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, P. R. China
- School of Environmental Sciences, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jiahuang Qiu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, P. R. China
- School of Environmental Sciences, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yin Liu
- School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 330106, P. R. China
| | - Qing'e Shan
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, P. R. China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P. R. China
| | - Ming Xu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, P. R. China
- School of Environmental Sciences, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Bing Yan
- Institute of Environmental Research at Greater Bays, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou, 510006, P. R. China
| | - Sijin Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, P. R. China
- School of Environmental Sciences, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P. R. China
| |
Collapse
|
15
|
Wu Y, Zhang Z, Wei Y, Qian Z, Wei X. Nanovaccines for cancer immunotherapy: Current knowledge and future perspectives. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.108098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
16
|
Liu Y, Zhao D, Wang Y, Chen Z, Yang L, Li W, Gong Y, Gan C, Tang J, Zhang T, Tang D, Dong X, Yang Q, Valencia CA, Dai L, Qi S, Dong B, Chow HY, Li Y. A vaccine based on the yeast-expressed receptor-binding domain (RBD) elicits broad immune responses against SARS-CoV-2 variants. Front Immunol 2022; 13:1011484. [PMID: 36439096 PMCID: PMC9682237 DOI: 10.3389/fimmu.2022.1011484] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 10/24/2022] [Indexed: 06/23/2024] Open
Abstract
Development of safe and efficient vaccines is still necessary to deal with the COVID-19 pandemic. Herein, we reported that yeast-expressed recombinant RBD proteins either from wild-type or Delta SARS-CoV-2 were able to elicit immune responses against SARS-CoV-2 and its variants. The wild-type RBD (wtRBD) protein was overexpressed in Pichia pastoris, and the purified protein was used as the antigen to immunize mice after formulating an aluminium hydroxide (Alum) adjuvant. Three immunization programs with different intervals were compared. It was found that the immunization with an interval of 28 days exhibited the strongest immune response to SARS-CoV-2 than the one with an interval of 14 or 42 days based on binding antibody and the neutralizing antibody (NAb) analyses. The antisera from the mice immunized with wtRBD were able to neutralize the Beta variant with a similar efficiency but the Delta variant with 2~2.5-fold decreased efficiency. However, more NAbs to the Delta variant were produced when the Delta RBD protein was used to immunize mice. Interestingly, the NAbs may cross react with the Omicron variant. To increase the production of NAbs, the adjuvant combination of Alum and CpG oligonucleotides was used. Compared with the Alum adjuvant alone, the NAbs elicited by the combined adjuvants exhibited an approximate 10-fold increase for the Delta and a more than 53-fold increase for the Omicron variant. This study suggested that yeast-derived Delta RBD is a scalable and an effective vaccine candidate for SARS-CoV-2 and its variants.
Collapse
Affiliation(s)
- Yu Liu
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Danhua Zhao
- Department of Arboviral Vaccine, National Institutes for Food and Drug Control, Beijing, China
| | - Yichang Wang
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Zhian Chen
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Li Yang
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Wenjuan Li
- Department of Arboviral Vaccine, National Institutes for Food and Drug Control, Beijing, China
| | - Yanqiu Gong
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Chunmei Gan
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jieshi Tang
- College of Life Sciences, Sichuan University, Chengdu, China
| | - Tizhong Zhang
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Dan Tang
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiuju Dong
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Qingzhe Yang
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - C. Alexander Valencia
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Lunzhi Dai
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Shiqian Qi
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Biao Dong
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Sichuan Real & Best Biotech Co., Ltd., Chengdu, China
| | - Hoi Yee Chow
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yuhua Li
- Department of Arboviral Vaccine, National Institutes for Food and Drug Control, Beijing, China
| |
Collapse
|
17
|
Nanoalum adjuvanted vaccines: small details make a big difference. Semin Immunol 2021; 56:101544. [PMID: 34895823 DOI: 10.1016/j.smim.2021.101544] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/24/2021] [Accepted: 11/24/2021] [Indexed: 11/24/2022]
Abstract
Purified vaccine antigens offer important safety and reactogenicity advantages compared with live attenuated or whole killed virus and bacterial vaccines. However, they require the addition of adjuvants to induce the magnitude, duration and quality of immune response required to achieve protective immunity. Aluminium salts have been used as adjuvants in vaccines for almost a century. In the literature, they are often referred to as aluminium-based adjuvants (ABAs), or aluminium salt-containing adjuvants or more simply "alum". All these terms are used to group aluminium suspensions that are very different in terms of atomic composition, size, and shape. They differ also in stability, antigen-adsorption, and antigen-release kinetics. Critically, these parameters also have a profound effect on the character and magnitude of the immune response elicited. Recent findings suggest that, by reducing the size of aluminium from micro to nanometers, a more effective adjuvant is obtained, together with the ability to sterile filter the vaccine product. However, the behaviour of aluminium nanoparticles in vaccine formulations is different from microparticles, requiring specific formulation strategies, as well as a more detailed understanding of how formulation influences the immune response generated. Here we review the current state of art of aluminium nanoparticles as adjuvants, with a focus on their immunobiology, preparation methods, formulation optimisation and stabilisation.
Collapse
|
18
|
Intranasal vaccination with protein bodies elicit strong protection against Streptococcus pneumoniae colonization. Vaccine 2021; 39:6920-6929. [PMID: 34696934 DOI: 10.1016/j.vaccine.2021.10.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 09/22/2021] [Accepted: 10/06/2021] [Indexed: 12/11/2022]
Abstract
Protein bodies (PBs) are particles consisting of insoluble, aggregated proteins with potential as a vaccine formulation. PBs can contain high concentrations of antigen, are stable and relatively resistant to proteases, release antigen slowly and are cost-effective to manufacture. Yet, the capacity of PBs to provoke immune responses and protection in the upper respiratory tract, a major entry route of respiratory pathogens, is largely unknown. In this study, we vaccinated mice intranasally with PBs comprising antigens from Streptococcus pneumoniae and evaluated the level of protection against nasopharyngeal colonization. PBs composed of the α-helical domain of pneumococcal surface protein A (PspAα) provided superior protection against colonization with S. pneumoniae compared to soluble PspAα. Immunization with soluble protein or PBs induced differences in antibody binding to pneumococci as well as a highly distinct antigen-specific nasal cytokine profile upon in vivo stimulation with inactivated S. pneumoniae. Moreover, immunization with PBs composed of conserved putative pneumococcal antigens reduced colonization by S. pneumoniae in mice, both as a single- and as a multi-antigen formulation. In conclusion, PBs represent a vaccine formulation that elicits strong mucosal immune responses and protection. The versatility of this platform offers opportunities for development of next-generation vaccine formulations.
Collapse
|
19
|
van Beek LF, Welzen PLW, Teufel LU, Joosten I, Diavatopoulos DA, van Hest J, de Jonge MI. Bimodal Targeting of Human Leukocytes by Fc- and CpG-Decorated Polymersomes to Tune Immune Induction. Biomacromolecules 2021; 22:4422-4433. [PMID: 34554732 PMCID: PMC8512671 DOI: 10.1021/acs.biomac.1c00985] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
The use of well-defined
nanovesicles composed of amphiphilic block copolymers (polymersomes) for delivery
of adjuvants and antigens is a promising strategy for vaccine development.
However, the potency of nanoparticle vaccines depends on efficient
interaction with and activation of cells involved in antigen presentation,
which can be achieved by targeting cellular receptors. Here, we showed
that the Fc fragment display on the polymersome surface resulted in
markedly improved interactions with granulocytes, monocytes, and NK
cells, while for “naked” polymersomes, virtually no
binding to leukocytes was observed. Moreover, CpG-decorated polymersomes
were found to also interact with T and/or B cells. Interestingly,
whole blood stimulations with Fc fragment and CpG-decorated polymersomes
induced interleukin (IL)-6, IL-8, and TNF-α production, while
naked polymersomes did not induce any cytokine production. In conclusion,
specific immune induction by polymersomes can be controlled using
bimodal targeting of different immune receptors, which is an essential
feature for targeted vaccine delivery.
Collapse
Affiliation(s)
- Lucille F van Beek
- Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands.,Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Pascal L W Welzen
- Department of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands
| | - Lisa U Teufel
- Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands.,Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Irma Joosten
- Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Dimitri A Diavatopoulos
- Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands.,Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Jan van Hest
- Department of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands
| | - Marien I de Jonge
- Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands.,Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| |
Collapse
|
20
|
Durán-Lobato M, López-Estévez AM, Cordeiro AS, Dacoba TG, Crecente-Campo J, Torres D, Alonso MJ. Nanotechnologies for the delivery of biologicals: Historical perspective and current landscape. Adv Drug Deliv Rev 2021; 176:113899. [PMID: 34314784 DOI: 10.1016/j.addr.2021.113899] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 07/05/2021] [Accepted: 07/23/2021] [Indexed: 12/12/2022]
Abstract
Biological macromolecule-based therapeutics irrupted in the pharmaceutical scene generating a great hope due to their outstanding specificity and potency. However, given their susceptibility to degradation and limited capacity to overcome biological barriers new delivery technologies had to be developed for them to reach their targets. This review aims at analyzing the historical seminal advances that shaped the development of the protein/peptide delivery field, along with the emerging technologies on the lead of the current landscape. Particularly, focus is made on technologies with a potential for transmucosal systemic delivery of protein/peptide drugs, followed by approaches for the delivery of antigens as new vaccination strategies, and formulations of biological drugs in oncology, with special emphasis on mAbs. Finally, a discussion of the key challenges the field is facing, along with an overview of prospective advances are provided.
Collapse
|
21
|
Combination Adjuvants Affect the Magnitude of Effector-Like Memory CD8 T Cells and Protection against Listeriosis. Infect Immun 2021; 89:e0076820. [PMID: 33782151 DOI: 10.1128/iai.00768-20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The development of T cell-based subunit protein vaccines against diseases such as tuberculosis and malaria remains a challenge for immunologists. Here, we have identified a nanoemulsion adjuvant, Adjuplex (ADJ), which enhanced dendritic cell (DC) cross-presentation and elicited effective memory T cell-based immunity to Listeria monocytogenes. We further evaluated whether cross-presentation induced by ADJ can be combined with the immunomodulatory effects of Toll-like receptor (TLR) agonists (CpG or glucopyranosyl lipid adjuvant [GLA]) to evoke systemic CD8 T cell-based immunity to L. monocytogenes. Mechanistically, vaccination with ADJ, alone or in combination with CpG or GLA, augmented activation and antigen uptake by CD103+ migratory and CD8α+ resident DCs and upregulated CD69 expression on B and T lymphocytes in vaccine-draining lymph nodes. By engaging basic leucine zipper ATF-like transcription factor 3-dependent cross-presenting DCs, ADJ potently elicited effector CD8 T cells that differentiated into granzyme B-expressing CD27LO effector-like memory CD8 T cells, which provided effective immunity to L. monocytogenes in the spleen and liver. CpG or GLA alone did not elicit effector-like memory CD8 T cells and induced moderate protection in the spleen but not in the liver. Surprisingly, combining CpG or GLA with ADJ reduced the number of ADJ-induced memory CD8 T cells and compromised protective immunity to L. monocytogenes, especially in the liver. Taken together, the data presented in this study provide a glimpse of protective CD8 T cell memory differentiation induced by a nanoemulsion adjuvant and demonstrate the unexpected negative effects of TLR signaling on the magnitude of CD8 T cell memory and protective immunity to L. monocytogenes, a model intracellular pathogen.
Collapse
|
22
|
Pulendran B, S Arunachalam P, O'Hagan DT. Emerging concepts in the science of vaccine adjuvants. Nat Rev Drug Discov 2021; 20:454-475. [PMID: 33824489 PMCID: PMC8023785 DOI: 10.1038/s41573-021-00163-y] [Citation(s) in RCA: 800] [Impact Index Per Article: 200.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2021] [Indexed: 02/06/2023]
Abstract
Adjuvants are vaccine components that enhance the magnitude, breadth and durability of the immune response. Following its introduction in the 1920s, alum remained the only adjuvant licensed for human use for the next 70 years. Since the 1990s, a further five adjuvants have been included in licensed vaccines, but the molecular mechanisms by which these adjuvants work remain only partially understood. However, a revolution in our understanding of the activation of the innate immune system through pattern recognition receptors (PRRs) is improving the mechanistic understanding of adjuvants, and recent conceptual advances highlight the notion that tissue damage, different forms of cell death, and metabolic and nutrient sensors can all modulate the innate immune system to activate adaptive immunity. Furthermore, recent advances in the use of systems biology to probe the molecular networks driving immune response to vaccines ('systems vaccinology') are revealing mechanistic insights and providing a new paradigm for the vaccine discovery and development process. Here, we review the 'known knowns' and 'known unknowns' of adjuvants, discuss these emerging concepts and highlight how our expanding knowledge about innate immunity and systems vaccinology are revitalizing the science and development of novel adjuvants for use in vaccines against COVID-19 and future pandemics.
Collapse
Affiliation(s)
- Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
- Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
- Chemistry, Engineering & Medicine for Human Health, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
| | - Prabhu S Arunachalam
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | | |
Collapse
|
23
|
Tian Y, Hu Q, Zhang R, Zhou B, Xie D, Wang Y, Zhang X, Yang L. Rational design of innate defense regulator peptides as tumor vaccine adjuvants. NPJ Vaccines 2021; 6:75. [PMID: 34016984 PMCID: PMC8138013 DOI: 10.1038/s41541-021-00334-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 04/06/2021] [Indexed: 02/05/2023] Open
Abstract
The development of adjuvants has been an empirical process. Efforts to develop a new design and evaluation system for novel adjuvants are not only desirable but also necessary. Moreover, composite adjuvants that contain two or more types of adjuvants to synergistically enhance the immune response are important for adjuvant and vaccine design. Innate defense regulator peptides (IDRs) are promising adjuvants for clinical immunotherapy because they exhibit multifaceted immunomodulatory capabilities. However, the rational design and discovery of IDRs that have improved immunomodulatory activities have been hampered by the lack of screening techniques and the great challenges in the identification of their interaction partners. Here, we describe a screening and evaluation system for IDR design. On the basis of in vitro screening, the optimized IDR DP7 recruited neutrophils, monocytes and macrophages to the site of infection. The adjuvant, comprising the DP7 and CpG oligonucleotide (CpG), induced chemokine/cytokine expression, enhanced the antigen uptake by dendritic cells and upregulated surface marker expression in dendritic cells. Vaccination with the NY-ESO-1 or OVA antigens combined with the adjuvant alum/CpG/DP7 strongly suppressed tumor growth in mice which was due to the improvement of antigen-specific humoral and cellular immunity. Regarding the mechanism of action, GPR35 may be the potential interaction partner of DP7. Our study revealed the potential application of the screening and evaluation system as a strategy for rationally designing effective IDRs or composite adjuvants and identifying their mechanism of action.
Collapse
Affiliation(s)
- Yaomei Tian
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
- College of Bioengineering, Sichuan University of Science & Engineering, Zigong, Sichuan, PR China
| | - Qiuyue Hu
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Rui Zhang
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Bailing Zhou
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Daoyuan Xie
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Yuanda Wang
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Xueyan Zhang
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Li Yang
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China.
| |
Collapse
|
24
|
Zuo Z, Zou Y, Li Q, Guo Y, Zhang T, Wu J, He C, Eko FO. Intranasal immunization with inactivated chlamydial elementary bodies formulated in VCG-chitosan nanoparticles induces robust immunity against intranasal Chlamydia psittaci challenge. Sci Rep 2021; 11:10389. [PMID: 34001988 PMCID: PMC8129140 DOI: 10.1038/s41598-021-89940-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 05/04/2021] [Indexed: 02/03/2023] Open
Abstract
Vaccines based on live attenuated Chlamydia elementary bodies (EBs) can cause disease in vaccinated animals and the comparably safer inactivated whole EBs are only marginally protective. Recent studies show that a vaccine formulation comprising UV-inactivated EBs (EB) and appropriate mucosal delivery systems and/or adjuvants induced significant protective immunity. We tested the hypothesis that intranasal delivery of UV-inactivated C. psittaci EB formulated in Vibrio cholerae ghosts (VCG)-chitosan nanoparticles will induce protective immunity against intranasal challenge in SPF chickens. We first compared the impact of VCG and CpG adjuvants on protective immunity following IN mucosal and IM systemic delivery of EB formulated in chitosan hydrogel/microspheres. Immunologic analysis revealed that IN immunization in the presence of VCG induced higher levels of IFN-γ response than IM delivery or the CpG adjuvanted groups. Also, vaccine efficacy evaluation showed enhanced pharyngeal bacterial clearance and protection against lung lesions with the VCG adjuvanted vaccine formulation, thereby establishing the superior adjuvanticity of VCG over CpG. We next evaluated the impact of different concentrations of VCG on protective immunity following IN mucosal immunization. Interestingly, the adjuvanticity of VCG was concentration-dependent, since protective immunity induced following IN mucosal immunization showed dose-dependent immune responses and protection. These studies reveal that formulation of inactivated chlamydial antigens with adjuvants, such as VCG and chitosan increases their ability to induce protective immune responses against challenge.
Collapse
Affiliation(s)
- Zonghui Zuo
- grid.22935.3f0000 0004 0530 8290Key Lab of Animal Epidemiology and Zoonosis, College of Veterinary Medicine, China Agricultural University, Beijing, 100193 People’s Republic of China
| | - Yongjuan Zou
- grid.9227.e0000000119573309Key Laboratory of Biopharmaceutical Production and Formulation Engineering, Chinese Academy of Sciences, Beijing, 100049 People’s Republic of China
| | - Qiang Li
- grid.22935.3f0000 0004 0530 8290Key Lab of Animal Epidemiology and Zoonosis, College of Veterinary Medicine, China Agricultural University, Beijing, 100193 People’s Republic of China
| | - Yongxia Guo
- grid.22935.3f0000 0004 0530 8290Key Lab of Animal Epidemiology and Zoonosis, College of Veterinary Medicine, China Agricultural University, Beijing, 100193 People’s Republic of China
| | - Tianyuan Zhang
- grid.22935.3f0000 0004 0530 8290Key Lab of Animal Epidemiology and Zoonosis, College of Veterinary Medicine, China Agricultural University, Beijing, 100193 People’s Republic of China
| | - Jie Wu
- grid.9227.e0000000119573309Key Laboratory of Biopharmaceutical Production and Formulation Engineering, Chinese Academy of Sciences, Beijing, 100049 People’s Republic of China
| | - Cheng He
- grid.22935.3f0000 0004 0530 8290Key Lab of Animal Epidemiology and Zoonosis, College of Veterinary Medicine, China Agricultural University, Beijing, 100193 People’s Republic of China
| | - Francis O. Eko
- grid.9001.80000 0001 2228 775XDepartment of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310 USA
| |
Collapse
|
25
|
Han L, Peng K, Qiu LY, Li M, Ruan JH, He LL, Yuan ZX. Hitchhiking on Controlled-Release Drug Delivery Systems: Opportunities and Challenges for Cancer Vaccines. Front Pharmacol 2021; 12:679602. [PMID: 34040536 PMCID: PMC8141731 DOI: 10.3389/fphar.2021.679602] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 04/28/2021] [Indexed: 12/11/2022] Open
Abstract
Cancer vaccines represent among the most promising strategies in the battle against cancers. However, the clinical efficacy of current cancer vaccines is largely limited by the lack of optimized delivery systems to generate strong and persistent antitumor immune responses. Moreover, most cancer vaccines require multiple injections to boost the immune responses, leading to poor patient compliance. Controlled-release drug delivery systems are able to address these issues by presenting drugs in a controlled spatiotemporal manner, which allows co-delivery of multiple drugs, reduction of dosing frequency and avoidance of significant systemic toxicities. In this review, we outline the recent progress in cancer vaccines including subunit vaccines, genetic vaccines, dendritic cell-based vaccines, tumor cell-based vaccines and in situ vaccines. Furthermore, we highlight the efforts and challenges of controlled or sustained release drug delivery systems (e.g., microparticles, scaffolds, injectable gels, and microneedles) in ameliorating the safety, effectiveness and operability of cancer vaccines. Finally, we briefly discuss the correlations of vaccine release kinetics and the immune responses to enlighten the rational design of the next-generation platforms for cancer therapy.
Collapse
Affiliation(s)
- Lu Han
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| | - Ke Peng
- School of pharmacy, Queen's University Belfast, Belfast, United Kingdom
| | - Li-Ying Qiu
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| | - Meng Li
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| | - Jing-Hua Ruan
- The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Li-Li He
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| | - Zhi-Xiang Yuan
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| |
Collapse
|
26
|
Cuzzubbo S, Mangsbo S, Nagarajan D, Habra K, Pockley AG, McArdle SEB. Cancer Vaccines: Adjuvant Potency, Importance of Age, Lifestyle, and Treatments. Front Immunol 2021; 11:615240. [PMID: 33679703 PMCID: PMC7927599 DOI: 10.3389/fimmu.2020.615240] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 12/23/2020] [Indexed: 12/13/2022] Open
Abstract
Although the discovery and characterization of multiple tumor antigens have sparked the development of many antigen/derived cancer vaccines, many are poorly immunogenic and thus, lack clinical efficacy. Adjuvants are therefore incorporated into vaccine formulations to trigger strong and long-lasting immune responses. Adjuvants have generally been classified into two categories: those that ‘depot’ antigens (e.g. mineral salts such as aluminum hydroxide, emulsions, liposomes) and those that act as immunostimulants (Toll Like Receptor agonists, saponins, cytokines). In addition, several novel technologies using vector-based delivery of antigens have been used. Unfortunately, the immune system declines with age, a phenomenon known as immunosenescence, and this is characterized by functional changes in both innate and adaptive cellular immunity systems as well as in lymph node architecture. While many of the immune functions decline over time, others paradoxically increase. Indeed, aging is known to be associated with a low level of chronic inflammation—inflamm-aging. Given that the median age of cancer diagnosis is 66 years and that immunotherapeutic interventions such as cancer vaccines are currently given in combination with or after other forms of treatments which themselves have immune-modulating potential such as surgery, chemotherapy and radiotherapy, the choice of adjuvants requires careful consideration in order to achieve the maximum immune response in a compromised environment. In addition, more clinical trials need to be performed to carefully assess how less conventional form of immune adjuvants, such as exercise, diet and psychological care which have all be shown to influence immune responses can be incorporated to improve the efficacy of cancer vaccines. In this review, adjuvants will be discussed with respect to the above-mentioned important elements.
Collapse
Affiliation(s)
- Stefania Cuzzubbo
- Université de Paris, PARCC, INSERM U970, 75015, Paris, France.,Laboratoire de Recherches Biochirurgicales (Fondation Carpentier), Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Européen Georges Pompidou, Paris, France
| | - Sara Mangsbo
- Ultimovacs AB, Uppsala, Sweden.,Department of Pharmaceutical Biosciences, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Divya Nagarajan
- Department of Immunology, Genetics and Clinical pathology Rudbeck laboratories, Uppsala University, Uppsala, Sweden
| | - Kinana Habra
- The School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom.,The John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Alan Graham Pockley
- The John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom.,Centre for Health, Ageing and Understanding Disease (CHAUD), School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Stephanie E B McArdle
- The John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom.,Centre for Health, Ageing and Understanding Disease (CHAUD), School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| |
Collapse
|
27
|
Olsen HE, Lynn GM, Valdes PA, Cerecedo Lopez CD, Ishizuka AS, Arnaout O, Bi WL, Peruzzi PP, Chiocca EA, Friedman GK, Bernstock JD. Therapeutic cancer vaccines for pediatric malignancies: advances, challenges, and emerging technologies. Neurooncol Adv 2021; 3:vdab027. [PMID: 33860227 PMCID: PMC8034661 DOI: 10.1093/noajnl/vdab027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Though outcomes for pediatric cancer patients have significantly improved over the past several decades, too many children still experience poor outcomes and survivors suffer lifelong, debilitating late effects after conventional chemotherapy, radiation, and surgical treatment. Consequently, there has been a renewed focus on developing novel targeted therapies to improve survival outcomes. Cancer vaccines are a promising type of immunotherapy that leverage the immune system to mediate targeted, tumor-specific killing through recognition of tumor antigens, thereby minimizing off-target toxicity. As such, cancer vaccines are orthogonal to conventional cancer treatments and can therefore be used alone or in combination with other therapeutic modalities to maximize efficacy. To date, cancer vaccination has remained largely understudied in the pediatric population. In this review, we discuss the different types of tumor antigens and vaccine technologies (dendritic cells, peptides, nucleic acids, and viral vectors) evaluated in clinical trials, with a focus on those used in children. We conclude with perspectives on how advances in combination therapies, tumor antigen (eg, neoantigen) selection, and vaccine platform optimization can be translated into clinical practice to improve outcomes for children with cancer.
Collapse
Affiliation(s)
- Hannah E Olsen
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Pablo A Valdes
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Neurosurgery, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Christian D Cerecedo Lopez
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Omar Arnaout
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - W Linda Bi
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Pier Paolo Peruzzi
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - E Antonio Chiocca
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Gregory K Friedman
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Joshua D Bernstock
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Avidea Technologies, Inc., Baltimore, Maryland, USA.,Department of Neurosurgery, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
28
|
Teng Z, Sun S, Luo X, Zhang Z, Seo H, Xu X, Huang J, Dong H, Mu S, Du P, Zhang Z, Guo H. Bi-functional gold nanocages enhance specific immunological responses of foot-and-mouth disease virus-like particles vaccine as a carrier and adjuvant. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 33:102358. [PMID: 33484882 DOI: 10.1016/j.nano.2021.102358] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 12/10/2020] [Accepted: 12/26/2020] [Indexed: 10/22/2022]
Abstract
Virus-like particle (VLP) vaccines have become one of the dominant vaccine candidates for foot-and-mouth disease (FMD). To further enhance the immunogenicity of VLP vaccines, gold nanocages (AuNCs) were selected as an adjuvant for the vaccine. Our experiments demonstrated that AuNCs had little biotoxicity in vivo and in vitro and improved the uptake of VLP in BHK-21 and RAW264.7 cell lines. The VLP-AuNCs activated DCs mainly through toll-like receptor 4 (TLR4) and promoted the secretion of IL-6, IL-1β, and TNF-α. The conjugation of VLP and AuNCs triggered a strong immune response against FMD virus (FMDV) in mice and guinea pigs. The VLP-AuNCs significantly enhanced the proliferation of CD8+ T cells (P < 0.05) and the secretion of cellular immune-related cytokines (IFN-γ, P < 0.05; IL-12p70, P < 0.01) compared with VLP. The present study demonstrated that AuNCs, as a great potential adjuvant for FMDV VLP vaccines, significantly enhance the immune response.
Collapse
Affiliation(s)
- Zhidong Teng
- State Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Shiqi Sun
- State Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Xin Luo
- State Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Zhihui Zhang
- State Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Hoseong Seo
- Biotechnology Division, Korea Atomic Energy Research Institute, Jeongeup, Republic of Korea
| | - Xiaoyu Xu
- CAS Key Laboratory for Nano-Bio Interface Research, Division of Nano biomedicine, CAS Center for Excellence in Nano Science, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou,China
| | - Jie Huang
- CAS Key Laboratory for Nano-Bio Interface Research, Division of Nano biomedicine, CAS Center for Excellence in Nano Science, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou,China
| | - Hu Dong
- State Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Suyu Mu
- State Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Ping Du
- State Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Zhijun Zhang
- CAS Key Laboratory for Nano-Bio Interface Research, Division of Nano biomedicine, CAS Center for Excellence in Nano Science, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou,China.
| | - Huichen Guo
- State Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China; School of Animal Science, Yangtze University, Jingzhou, PR China..
| |
Collapse
|
29
|
Lee W, Kingstad-Bakke B, Paulson B, Larsen A, Overmyer K, Marinaik CB, Dulli K, Toy R, Vogel G, Mueller KP, Tweed K, Walsh AJ, Russell J, Saha K, Reyes L, Skala MC, Sauer JD, Shayakhmetov DM, Coon J, Roy K, Suresh M. Carbomer-based adjuvant elicits CD8 T-cell immunity by inducing a distinct metabolic state in cross-presenting dendritic cells. PLoS Pathog 2021; 17:e1009168. [PMID: 33444400 PMCID: PMC7840022 DOI: 10.1371/journal.ppat.1009168] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 01/27/2021] [Accepted: 11/16/2020] [Indexed: 01/25/2023] Open
Abstract
There is a critical need for adjuvants that can safely elicit potent and durable T cell-based immunity to intracellular pathogens. Here, we report that parenteral vaccination with a carbomer-based adjuvant, Adjuplex (ADJ), stimulated robust CD8 T-cell responses to subunit antigens and afforded effective immunity against respiratory challenge with a virus and a systemic intracellular bacterial infection. Studies to understand the metabolic and molecular basis for ADJ's effect on antigen cross-presentation by dendritic cells (DCs) revealed several unique and distinctive mechanisms. ADJ-stimulated DCs produced IL-1β and IL-18, suggestive of inflammasome activation, but in vivo activation of CD8 T cells was unaffected in caspase 1-deficient mice. Cross-presentation induced by TLR agonists requires a critical switch to anabolic metabolism, but ADJ enhanced cross presentation without this metabolic switch in DCs. Instead, ADJ induced in DCs, an unique metabolic state, typified by dampened oxidative phosphorylation and basal levels of glycolysis. In the absence of increased glycolytic flux, ADJ modulated multiple steps in the cytosolic pathway of cross-presentation by enabling accumulation of degraded antigen, reducing endosomal acidity and promoting antigen localization to early endosomes. Further, by increasing ROS production and lipid peroxidation, ADJ promoted antigen escape from endosomes to the cytosol for degradation by proteasomes into peptides for MHC I loading by TAP-dependent pathways. Furthermore, we found that induction of lipid bodies (LBs) and alterations in LB composition mediated by ADJ were also critical for DC cross-presentation. Collectively, our model challenges the prevailing metabolic paradigm by suggesting that DCs can perform effective DC cross-presentation, independent of glycolysis to induce robust T cell-dependent protective immunity to intracellular pathogens. These findings have strong implications in the rational development of safe and effective immune adjuvants to potentiate robust T-cell based immunity.
Collapse
Affiliation(s)
- Woojong Lee
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Brock Kingstad-Bakke
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Brett Paulson
- Morgridge Institute for Research, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Autumn Larsen
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Katherine Overmyer
- Morgridge Institute for Research, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Chandranaik B. Marinaik
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Kelly Dulli
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Randall Toy
- The Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University and The Parker H. Petit Institute for Bioengineering and Biosciences, Center for ImmunoEngineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Gabriela Vogel
- The Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University and The Parker H. Petit Institute for Bioengineering and Biosciences, Center for ImmunoEngineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Katherine P. Mueller
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Kelsey Tweed
- Morgridge Institute for Research, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Alex J. Walsh
- Morgridge Institute for Research, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Jason Russell
- Morgridge Institute for Research, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Krishanu Saha
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Leticia Reyes
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Melissa C. Skala
- Morgridge Institute for Research, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - John-Demian Sauer
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Dmitry M. Shayakhmetov
- Lowance Center for Human Immunology, Emory Vaccine Center, Departments of Pediatrics and Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Joshua Coon
- Morgridge Institute for Research, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Krishnendu Roy
- The Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University and The Parker H. Petit Institute for Bioengineering and Biosciences, Center for ImmunoEngineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - M. Suresh
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
30
|
Pereira B, Xu XN, Akbar AN. Targeting Inflammation and Immunosenescence to Improve Vaccine Responses in the Elderly. Front Immunol 2020; 11:583019. [PMID: 33178213 PMCID: PMC7592394 DOI: 10.3389/fimmu.2020.583019] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 09/23/2020] [Indexed: 12/19/2022] Open
Abstract
One of the most appreciated consequences of immunosenescence is an impaired response to vaccines with advanced age. While most studies report impaired antibody responses in older adults as a correlate of vaccine efficacy, it is now widely appreciated that this may fail to identify important changes occurring in the immune system with age that may affect vaccine efficacy. The impact of immunosenescence on vaccination goes beyond the defects on antibody responses as T cell-mediated responses are reshaped during aging and certainly affect vaccination. Likewise, age-related changes in the innate immune system may have important consequences on antigen presentation and priming of adaptive immune responses. Importantly, a low-level chronic inflammatory status known as inflammaging has been shown to inhibit immune responses to vaccination and pharmacological strategies aiming at blocking baseline inflammation can be potentially used to boost vaccine responses. Yet current strategies aiming at improving immunogenicity in the elderly have mainly focused on the use of adjuvants to promote local inflammation. More research is needed to understand the role of inflammation in vaccine responses and to reconcile these seemingly paradoxical observations. Alternative approaches to improve vaccine responses in the elderly include the use of higher vaccine doses or alternative routes of vaccination showing only limited benefits. This review will explore novel targets and potential new strategies for enhancing vaccine responses in older adults, including the use of anti-inflammatory drugs and immunomodulators.
Collapse
Affiliation(s)
- Branca Pereira
- HIV/GUM Directorate, Chelsea and Westminster Hospital NHS Foundation Trust, London, United Kingdom.,Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Xiao-Ning Xu
- Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Arne N Akbar
- Division of Medicine, University College London, London, United Kingdom
| |
Collapse
|
31
|
Lemoine C, Thakur A, Krajišnik D, Guyon R, Longet S, Razim A, Górska S, Pantelić I, Ilić T, Nikolić I, Lavelle EC, Gamian A, Savić S, Milicic A. Technological Approaches for Improving Vaccination Compliance and Coverage. Vaccines (Basel) 2020; 8:E304. [PMID: 32560088 PMCID: PMC7350210 DOI: 10.3390/vaccines8020304] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/13/2020] [Accepted: 06/14/2020] [Indexed: 12/18/2022] Open
Abstract
Vaccination has been well recognised as a critically important tool in preventing infectious disease, yet incomplete immunisation coverage remains a major obstacle to achieving disease control and eradication. As medical products for global access, vaccines need to be safe, effective and inexpensive. In line with these goals, continuous improvements of vaccine delivery strategies are necessary to achieve the full potential of immunisation. Novel technologies related to vaccine delivery and route of administration, use of advanced adjuvants and controlled antigen release (single-dose immunisation) approaches are expected to contribute to improved coverage and patient compliance. This review discusses the application of micro- and nano-technologies in the alternative routes of vaccine administration (mucosal and cutaneous vaccination), oral vaccine delivery as well as vaccine encapsulation with the aim of controlled antigen release for single-dose vaccination.
Collapse
Affiliation(s)
- Céline Lemoine
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Rue Michel-Servet 1, 1221 Geneva, Switzerland;
- Vaccine Formulation Institute, Chemin des Aulx 14, 1228 Plan-les-Ouates, Switzerland
| | - Aneesh Thakur
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark;
| | - Danina Krajišnik
- Department of Pharmaceutical Technology and Cosmetology, University of Belgrade-Faculty of Pharmacy, Vojvode Stepe 450, 11221 Belgrade, Serbia; (D.K.); (I.P.); (T.I.); (I.N.); (S.S.)
| | - Romain Guyon
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK;
| | - Stephanie Longet
- Virology & Pathogenesis Group, Public Health England, Manor Farm Road, Porton Down, Salisbury SP4 0JG, UK;
| | - Agnieszka Razim
- Department of Microbiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, ul. Rudolfa Weigla 12, 53-114 Wroclaw, Poland; (A.R.); (S.G.)
| | - Sabina Górska
- Department of Microbiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, ul. Rudolfa Weigla 12, 53-114 Wroclaw, Poland; (A.R.); (S.G.)
| | - Ivana Pantelić
- Department of Pharmaceutical Technology and Cosmetology, University of Belgrade-Faculty of Pharmacy, Vojvode Stepe 450, 11221 Belgrade, Serbia; (D.K.); (I.P.); (T.I.); (I.N.); (S.S.)
| | - Tanja Ilić
- Department of Pharmaceutical Technology and Cosmetology, University of Belgrade-Faculty of Pharmacy, Vojvode Stepe 450, 11221 Belgrade, Serbia; (D.K.); (I.P.); (T.I.); (I.N.); (S.S.)
| | - Ines Nikolić
- Department of Pharmaceutical Technology and Cosmetology, University of Belgrade-Faculty of Pharmacy, Vojvode Stepe 450, 11221 Belgrade, Serbia; (D.K.); (I.P.); (T.I.); (I.N.); (S.S.)
| | - Ed C. Lavelle
- The Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, DO2R590 Dublin, Ireland;
| | - Andrzej Gamian
- Department of Immunology of Infectious Diseases, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, ul. Rudolfa Weigla 12, 53-114 Wroclaw, Poland;
| | - Snežana Savić
- Department of Pharmaceutical Technology and Cosmetology, University of Belgrade-Faculty of Pharmacy, Vojvode Stepe 450, 11221 Belgrade, Serbia; (D.K.); (I.P.); (T.I.); (I.N.); (S.S.)
| | - Anita Milicic
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK;
| |
Collapse
|
32
|
Gao Z, Zhang C, Jing L, Feng M, Li R, Yang Y. The structural characterization and immune modulation activitives comparison of Codonopsis pilosula polysaccharide (CPPS) and selenizing CPPS (sCPPS) on mouse in vitro and vivo. Int J Biol Macromol 2020; 160:814-822. [PMID: 32446900 DOI: 10.1016/j.ijbiomac.2020.05.149] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 05/12/2020] [Accepted: 05/18/2020] [Indexed: 02/01/2023]
Abstract
Codonopsis pilosula polysaccharide (CPPS) and selenizing CPPS (sCPPS) were prepared and identified by a combination of chemical and instrumental analysis. Their immune modulation activities were compared by lymphocyte proliferation and flowcytometry tests in vitro or serum antibody responses and cytokines with immunization against OVA mice in vivo. The results showed that the sCPPS was successfully modified in selenylation. In vitro, the sCPPS were more effective compared with CPPS in promoting lymphocyte proliferation synergistically with PHA or LPS and increasing the ratio of CD4+ to CD8 + T cells. In vivo, sCPPS could significantly raised IgG, IgM, IFN-γ, IL-2 and IL-4 contents in the serum of mouse against OVA in comparison with CPPS. These results indicate that selenylation modification can enhance the immune modulation activitives of CPPS. sCPPS would be as a component drug of new-type immunoenhancer.
Collapse
Affiliation(s)
- Zhenzhen Gao
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, PR China.
| | - Chao Zhang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, PR China
| | - Lirong Jing
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, PR China
| | - Min Feng
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, PR China
| | - Ran Li
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, PR China
| | - Ying Yang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, PR China
| |
Collapse
|
33
|
Alshammari AM, Smith DD, Parriott J, Stewart JP, Curran SM, McCulloh RJ, Barry PA, Iyer SS, Palermo N, Phillips JA, Dong Y, Ronning DR, Vennerstrom JL, Sanderson SD, Vetro JA. Targeted Amino Acid Substitution Overcomes Scale-Up Challenges with the Human C5a-Derived Decapeptide Immunostimulant EP67. ACS Infect Dis 2020; 6:1169-1181. [PMID: 32233506 PMCID: PMC7279522 DOI: 10.1021/acsinfecdis.0c00005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
EP67 is a second-generation, human C5a-derived decapeptide agonist of C5a receptor 1 (C5aR1/CD88) that selectively activates mononuclear phagocytes over neutrophils to potentiate protective innate and adaptive immune responses while potentially minimizing neutrophil-mediated toxicity. Pro7 and N-methyl-Leu8 (Me-Leu8) amino acid residues within EP67 likely induce backbone structural changes that increase potency and selective activation of mononuclear phagocytes over neutrophils versus first-generation EP54. The low coupling efficiency between Pro7 and Me-Leu8 and challenging purification by HPLC, however, greatly increase scale-up costs of EP67 for clinical use. Thus, the goal of this study was to determine whether replacing Pro7 and/or Me-Leu8 with large-scale amenable amino acid residues predicted to induce similar structural changes (cyclohexylalanine7 and/or leucine8) sufficiently preserves EP67 activity in primary human mononuclear phagocytes and neutrophils. We found that EP67 analogues had similar potency, efficacy, and selective activation of mononuclear phagocytes over neutrophils. Thus, replacing Pro7 and/or Me-Leu8 with large-scale amenable amino acid residues predicted to induce similar structural changes is a suitable strategy to overcome scale-up challenges with EP67.
Collapse
Affiliation(s)
- Abdulraman M. Alshammari
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198-6025, USA
| | - D. David Smith
- Department of Biomedical Sciences, Creighton University, 2500 California Plaza, Omaha, NE 68178, USA
| | - Jake Parriott
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198-6025, USA
| | - Jason P. Stewart
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198-6025, USA
| | - Stephen M. Curran
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198-6025, USA
| | - Russell J. McCulloh
- Department of Pediatrics, Children’s Hospital and Medical Center, Omaha, Nebraska, 68114, USA
| | - Peter A. Barry
- Center for Immunology and Infectious Diseases, Pathology and Laboratory Medicine, UC Davis School of Medicine, Davis, CA 95817, USA
| | - Smita S. Iyer
- Center for Immunology and Infectious Diseases, Pathology, Microbiology & Immunology, UC Davis, School of Veterinary Medicine, California National Primate Research Center, Davis, CA 95817, USA
| | - Nicholas Palermo
- Holland Computing Center, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Joy A. Phillips
- Donald P. Shiley BioScience Center, San Diego State University, San Diego, CA 92115, USA
| | - Yuxiang Dong
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198-6025, USA
| | - Donald R. Ronning
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198-6025, USA
| | - Jonathan L. Vennerstrom
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198-6025, USA
| | - Sam D. Sanderson
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198-6025, USA
| | - Joseph A. Vetro
- Center for Drug Delivery and Nanomedicine, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198-6025, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198-6025, USA
| |
Collapse
|
34
|
Qi J, Yin Y, Yu W, Shen L, Xu J, Hu T. Conjugation of β-Glucan with the Hydrazone and Disulfide Linkers Markedly Improves the Immunogenicity of Zika Virus E Protein. Mol Pharm 2020; 17:1933-1944. [DOI: 10.1021/acs.molpharmaceut.0c00010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Jinming Qi
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100190, China
| | - Ying Yin
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing 100071, China
| | - Weili Yu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Lijuan Shen
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Junjie Xu
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing 100071, China
| | - Tao Hu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
35
|
Mannosylated liposomes formulated with whole parasite P. falciparum blood-stage antigens are highly immunogenic in mice. Vaccine 2020; 38:1494-1504. [DOI: 10.1016/j.vaccine.2019.11.063] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 11/03/2019] [Accepted: 11/26/2019] [Indexed: 12/20/2022]
|
36
|
Allahyari M, Mohabati R, Vatanara A, Golkar M. In-vitro and in-vivo comparison of rSAG1-loaded PLGA prepared by encapsulation and adsorption methods as an efficient vaccine against Toxoplasma gondii”. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2019.101327] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
37
|
Abstract
Mucosal surfaces represent important routes of entry into the human body for the majority of pathogens, and they constitute unique sites for targeted vaccine delivery. Nanoparticle-based drug delivery systems are emerging technologies for delivering and improving the efficacy of mucosal vaccines. Recent studies have provided new insights into formulation and delivery aspects of importance for the design of safe and efficacious mucosal subunit vaccines based on nanoparticles. These include novel nanomaterials, their physicochemical properties and formulation approaches, nanoparticle interaction with immune cells in the mucosa, and mucosal immunization and delivery strategies. Here, we present recent progress in the application of nanoparticle-based approaches for mucosal vaccine delivery and discuss future research challenges and opportunities in the field.
Collapse
|
38
|
Short KK, Miller SM, Walsh L, Cybulski V, Bazin H, Evans JT, Burkhart D. Co-encapsulation of synthetic lipidated TLR4 and TLR7/8 agonists in the liposomal bilayer results in a rapid, synergistic enhancement of vaccine-mediated humoral immunity. J Control Release 2019; 315:186-196. [PMID: 31654684 PMCID: PMC6980726 DOI: 10.1016/j.jconrel.2019.10.025] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 10/09/2019] [Accepted: 10/14/2019] [Indexed: 12/11/2022]
Abstract
To increase vaccine immunogenicity, modern vaccines incorporate adjuvants, which serve to enhance immune cross-protection, improve humoral and cell-mediated immunity, and promote antigen dose sparing. Pattern recognition receptors (PRRs), including the Toll-like receptor (TLR) family are promising targets for development of agonist formulations for use as vaccine adjuvants. Combinations of co-delivered TLR4 and TLR7/8 ligands have been demonstrated to have synergistic effects on innate and adaptive immune response. Here, we create liposomes that stably co-encapsulate CRX-601, a synthetic TLR4 agonist, and UM-3004, a lipidated TLR7/8 agonist, within the liposomal bilayer in order to achieve co-delivery, allow tunable physical properties, and induce in vitro and in vivo immune synergy. Co-encapsulation demonstrates a synergistic increase in IL-12p70 cytokine output in vitro from treated human peripheral blood mononuclear cells (hPBMCs). Further, co-encapsulated formulations give significant improvement of early IgG2a antibody titers in BALB/c mice following primary vaccination when compared to single agonist or dual agonists delivered in separate liposomes. This work demonstrates that co-encapsulation of TLR4 and lipidated TLR7/8 agonists within the liposomal bilayer leads to innate and adaptive immune synergy which biases a Th1 immune response. Thus, liposomal co-encapsulation may be a useful and flexible tool for vaccine adjuvant formulation containing multiple TLR agonists.
Collapse
Affiliation(s)
- Kristopher K Short
- Center for Translational Medicine, University of Montana, Missoula, MT 59812, USA; Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA
| | - Shannon M Miller
- Center for Translational Medicine, University of Montana, Missoula, MT 59812, USA; Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
| | - Lois Walsh
- Center for Translational Medicine, University of Montana, Missoula, MT 59812, USA; Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA
| | - Van Cybulski
- Center for Translational Medicine, University of Montana, Missoula, MT 59812, USA; Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
| | - Hélène Bazin
- Center for Translational Medicine, University of Montana, Missoula, MT 59812, USA; Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA
| | - Jay T Evans
- Center for Translational Medicine, University of Montana, Missoula, MT 59812, USA; Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
| | - David Burkhart
- Center for Translational Medicine, University of Montana, Missoula, MT 59812, USA; Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA.
| |
Collapse
|
39
|
Wang P, Ding X, Kim H, Škalamera Đ, Michalek SM, Zhang P. Vaccine Adjuvants Derivatized from Momordica Saponins I and II. J Med Chem 2019; 62:9976-9982. [PMID: 31657920 DOI: 10.1021/acs.jmedchem.9b01511] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
We have derivatized Momordica saponins (MS) I and II through their coupling at C3 glucuronic acid site with dodecylamine. The derivatives show significantly different immunostimulant activity profiles from their respective natural parent saponins. In particular, adjuvant VSA-1 (5), the derivative of MS I, potentiates a significantly higher IgG2a responose than the corresponding natural product. Its IgG1 and IgG2a production is similar to that of GPI-0100, indicating a potential mixed and antigen-specific Th1/Th2 immune response, which is different from the Th2 immunity induced by the natural saponin MS I. In addition, toxicity evaluations show that adjuvant VSA-1 (5) is much less toxic than the widely used natural saponin mixture Quil A. These results prove that derivatizing Momordica saponins can be a viable way for easy access to structurally defined saponin immunostimulants with favorable adjvuant activity and low toxicity.
Collapse
|
40
|
Yang G, Chen S, Zhang J. Bioinspired and Biomimetic Nanotherapies for the Treatment of Infectious Diseases. Front Pharmacol 2019; 10:751. [PMID: 31333467 PMCID: PMC6624236 DOI: 10.3389/fphar.2019.00751] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 06/11/2019] [Indexed: 12/21/2022] Open
Abstract
There are still great challenges for the effective treatment of infectious diseases, although considerable achievement has been made by using antiviral and antimicrobial agents varying from small-molecule drugs, peptides/proteins, to nucleic acids. The nanomedicine approach is emerging as a new strategy capable of overcoming disadvantages of molecular therapeutics and amplifying their anti-infective activities, by localized delivery to infection sites, reducing off-target effects, and/or attenuating resistance development. Nanotechnology, in combination with bioinspired and biomimetic approaches, affords additional functions to nanoparticles derived from synthetic materials. Herein, we aim to provide a state-of-the-art review on recent progress in biomimetic and bioengineered nanotherapies for the treatment of infectious disease. Different biomimetic nanoparticles, derived from viruses, bacteria, and mammalian cells, are first described, with respect to their construction and biophysicochemical properties. Then, the applications of diverse biomimetic nanoparticles in anti-infective therapy are introduced, either by their intrinsic activity or by loading and site-specifically delivering various molecular drugs. Bioinspired and biomimetic nanovaccines for prevention and/or therapy of infectious diseases are also highlighted. At the end, major translation issues and future directions of this field are discussed.
Collapse
Affiliation(s)
- Guoyu Yang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing, China
- The First Clinical College, Chongqing Medical University, Chongqing, China
| | - Sheng Chen
- Department of Pediatrics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jianxiang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing, China
| |
Collapse
|
41
|
Trimaille T, Lacroix C, Verrier B. Self-assembled amphiphilic copolymers as dual delivery system for immunotherapy. Eur J Pharm Biopharm 2019; 142:232-239. [PMID: 31229673 DOI: 10.1016/j.ejpb.2019.06.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 05/03/2019] [Accepted: 06/19/2019] [Indexed: 01/07/2023]
Abstract
Subunit vaccines using recombinant antigens appear as the privileged vaccination technology for safety reasons but still require the development of carriers/adjuvants ensuring optimal immunogenicity and efficacy. Micelles from self-assembled amphiphilic copolymers have recently emerged as highly relevant and promising candidates owing to their ease of preparation, low size (entering in lymphatic capillaries for reaching lymph nodes), size/surface tunability and chemical versatility enabling introduction of stimuli (e.g. pH) responsive features and biofunctionalization with dedicated molecules. In particular, research efforts have increasingly focused on dendritic cells (DCs) targeting and activation by co-delivering (with antigen) ligands of pattern recognition receptors (PRRs, e.g. toll-like receptors). Such strategy has appeared as one of the most effective for eliciting CD 8+ T-cell response, which is crucial in the eradication of tumors and numerous infectious diseases. In this short review, we highlight the recent advances in such micelle-based carriers in subunit vaccination and how their precise engineering can be a strong asset for guiding and controlling immune responses.
Collapse
Affiliation(s)
- Thomas Trimaille
- Aix Marseille Univ, CNRS, Institut de Chimie Radicalaire, Marseille, France.
| | - Céline Lacroix
- Université Lyon 1, CNRS, UMR 5305, Biologie Tissulaire et Ingénierie Thérapeutique, IBCP, 69367 Lyon, France
| | - Bernard Verrier
- Université Lyon 1, CNRS, UMR 5305, Biologie Tissulaire et Ingénierie Thérapeutique, IBCP, 69367 Lyon, France
| |
Collapse
|
42
|
Wang P, Škalamera Đ, Sui X, Zhang P, Michalek SM. Synthesis and Evaluation of QS-7-Based Vaccine Adjuvants. ACS Infect Dis 2019; 5:974-981. [PMID: 30920199 DOI: 10.1021/acsinfecdis.9b00039] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
We have designed and synthesized two analogs (5 and 6) of QS-7, a natural saponin compound isolated from Quillaja saponaria (QS) Molina tree bark. The only structural difference between compound 5 and 6 is that 5 is acetylated at the 3- and 4-O positions of the quillaic acid C28 fucosyl unit while 6 is not. However, the two analogs show significantly different immunostimulant profiles. Compound 5 may potentiate a mixed Th1/Th2 (Th, T helper cells) immune response against the specific antigens while compound 6 may only induce a Th2-biased immunity. These results suggest that the 3- and/or 4-O acetyl groups of the fucosyl unit may play an important role in tuning the adjuvanticity of the QS-7 analogs, and compound 5 can serve as a structurally defined synthetic adjuvant when a mixed Th1/Th2 immune responses is desired.
Collapse
|
43
|
Imitation of nature: Bionic design in the study of particle adjuvants. J Control Release 2019; 303:101-108. [DOI: 10.1016/j.jconrel.2019.04.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 03/23/2019] [Accepted: 04/03/2019] [Indexed: 12/27/2022]
|
44
|
Efficient induction of cell-mediated immunity to varicella-zoster virus glycoprotein E co-lyophilized with a cationic liposome-based adjuvant in mice. Vaccine 2019; 37:2131-2141. [DOI: 10.1016/j.vaccine.2019.02.048] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 01/31/2019] [Accepted: 02/16/2019] [Indexed: 01/01/2023]
|
45
|
Senapati S, Darling RJ, Loh D, Schneider IC, Wannemuehler MJ, Narasimhan B, Mallapragada SK. Pentablock Copolymer Micelle Nanoadjuvants Enhance Cytosolic Delivery of Antigen and Improve Vaccine Efficacy while Inducing Low Inflammation. ACS Biomater Sci Eng 2019; 5:1332-1342. [PMID: 33405651 PMCID: PMC8627116 DOI: 10.1021/acsbiomaterials.8b01591] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
As the focus has shifted from traditional killed or live, attenuated vaccines toward subunit vaccines, improvements in vaccine safety have been confronted with low immunogenicity of protein antigens. This issue has been addressed by synthesizing and designing a wide variety of antigen carriers and adjuvants, such as Toll-like receptor agonists (e.g., MPLA, CpG). Studies have focused on optimizing adjuvants for improved cellular trafficking, cytosolic availability, and improved antigen presentation. In this work, we describe the design of novel amphiphilic pentablock copolymer (PBC) adjuvants that exhibit high biocompatibility and reversible pH- and temperature-sensitive micelle formation. We demonstrate improved humoral immunity in mice in response to single-dose immunization with PBC micelle adjuvants compared with soluble antigen alone. With the motive of exploring the mechanism of action of these PBC micelles, we studied intracellular trafficking of these PBC micelles with a model antigen and demonstrated that the PBC micelles associate with the antigen and enhance its cytosolic delivery to antigen-presenting cells. We posit that these PBC micelles operate via immune-enhancing mechanisms that are different from that of traditional Toll-like receptor activating adjuvants. The metabolic profile of antigen-presenting cells stimulated with traditional adjuvants and the PBC micelles also suggests distinct mechanisms of action. A key finding from this study is the low production of nitric oxide and reactive oxygen species by antigen-presenting cells when stimulated by PBC micelle adjuvants in sharp contrast to TLR adjuvants. Together, these studies provide a basis for rationally developing novel vaccine adjuvants that are safe, that induce low inflammation, and that can efficiently deliver antigen to the cytosol.
Collapse
Affiliation(s)
- Sujata Senapati
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa 50011, United States
- Nanovaccine Institute, Iowa State University, Ames, Iowa 50011, United States
| | - Ross J. Darling
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, Iowa 50011, United States
- Nanovaccine Institute, Iowa State University, Ames, Iowa 50011, United States
| | - Darren Loh
- Department of Chemical and Biological Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Ian C. Schneider
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa 50011, United States
- Nanovaccine Institute, Iowa State University, Ames, Iowa 50011, United States
| | - Michael J. Wannemuehler
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, Iowa 50011, United States
- Nanovaccine Institute, Iowa State University, Ames, Iowa 50011, United States
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa 50011, United States
- Nanovaccine Institute, Iowa State University, Ames, Iowa 50011, United States
| | - Surya K. Mallapragada
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa 50011, United States
- Nanovaccine Institute, Iowa State University, Ames, Iowa 50011, United States
| |
Collapse
|
46
|
A comparison between adjuvant and delivering functions of calcium phosphate, aluminum hydroxide and chitosan nanoparticles, using a model protein of Brucella melitensis Omp31. Immunol Lett 2019; 207:28-35. [DOI: 10.1016/j.imlet.2019.01.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 01/10/2019] [Accepted: 01/28/2019] [Indexed: 12/16/2022]
|
47
|
Lu F, Mosley YYC, Carmichael B, Brown DD, HogenEsch H. Formulation of aluminum hydroxide adjuvant with TLR agonists poly(I:C) and CpG enhances the magnitude and avidity of the humoral immune response. Vaccine 2019; 37:1945-1953. [PMID: 30803844 DOI: 10.1016/j.vaccine.2019.02.033] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 02/08/2019] [Accepted: 02/15/2019] [Indexed: 02/02/2023]
Abstract
Subunit vaccines generally require adjuvants to achieve optimal immune responses. Toll-like receptor (TLR) agonists are promising immune potentiators, but rapid diffusion from the injection site reduces their local effective concentration and may cause systemic reactions. In this study, we investigated the potential of aluminum hydroxide adjuvant (AH) to adsorb the TLR3 agonist poly(I:C) and TLR9 agonist CpG and compared the effect of the combination adjuvant on the immune response with either the TLR agonists or AH alone in mice. Poly(I:C) and CpG readily adsorbed onto AH and this combination adjuvant induced a stronger IgG1 and IgG2a immune response with a significant increase of antibody avidity. The combination adjuvant enhanced antigen uptake and activation of dendritic cells in vitro. It induced an inflammatory response at the injection site similar to AH but without eosinophils which are typically observed with AH. A distinctive antigen-containing monocyte/macrophage population with an intermediate level of CD11c expression was identified in the draining lymph nodes after immunization with TLR agonists and the combination adjuvant. Injection of the combination adjuvant did not induce an increase of TNFα and CXCL10 in serum in contrast to the injection of soluble TLR agonists. These results indicate that this combination adjuvant is a promising formulation to solve some of the unmet needs of current vaccines.
Collapse
Affiliation(s)
- Fangjia Lu
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, United States
| | - Yung-Yi C Mosley
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, United States
| | - Brooke Carmichael
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, United States
| | - Devonte D Brown
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, United States
| | - Harm HogenEsch
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, United States; Purdue Institute of Inflammation, Immunology and Infectious Diseases, Purdue University, West Lafayette, IN 47907, United States.
| |
Collapse
|
48
|
Lynn GM, Chytil P, Francica JR, Lagová A, Kueberuwa G, Ishizuka AS, Zaidi N, Ramirez-Valdez RA, Blobel NJ, Baharom F, Leal J, Wang AQ, Gerner MY, Etrych T, Ulbrich K, Seymour LW, Seder RA, Laga R. Impact of Polymer-TLR-7/8 Agonist (Adjuvant) Morphology on the Potency and Mechanism of CD8 T Cell Induction. Biomacromolecules 2019; 20:854-870. [PMID: 30608149 DOI: 10.1021/acs.biomac.8b01473] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Small molecule Toll-like receptor-7 and -8 agonists (TLR-7/8a) can be used as vaccine adjuvants to induce CD8 T cell immunity but require formulations that prevent systemic toxicity and focus adjuvant activity in lymphoid tissues. Here, we covalently attached TLR-7/8a to polymers of varying composition, chain architecture and hydrodynamic behavior (∼300 nm submicrometer particles, ∼10 nm micelles and ∼4 nm flexible random coils) and evaluated how these parameters of polymer-TLR-7/8a conjugates impact adjuvant activity in vivo. Attachment of TLR-7/8a to any of the polymer compositions resulted in a nearly 10-fold reduction in systemic cytokines (toxicity). Moreover, both lymph node cytokine production and the magnitude of CD8 T cells induced against protein antigen increased with increasing polymer-TLR-7/8a hydrodynamic radius, with the submicrometer particle inducing the highest magnitude responses. Notably, CD8 T cell responses induced by polymer-TLR-7/8a were dependent on CCR2+ monocytes and IL-12, whereas responses by a small molecule TLR-7/8a that unexpectedly persisted in vaccine-site draining lymph nodes (T1/2 = 15 h) had less dependence on monocytes and IL-12 but required Type I IFNs. This study shows how modular properties of synthetic adjuvants can be chemically programmed to alter immunity in vivo through distinct immunological mechanisms.
Collapse
Affiliation(s)
- Geoffrey M Lynn
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases , National Institutes of Health, 40 Convent Drive , Bethesda , Maryland 20892 , United States
| | - Petr Chytil
- Institute of Macromolecular Chemistry, Czech Academy of Sciences , Heyrovského nám. 2 , 162 06 Prague 6 , Czech Republic
| | - Joseph R Francica
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases , National Institutes of Health, 40 Convent Drive , Bethesda , Maryland 20892 , United States
| | - Anna Lagová
- Department of Oncology , University of Oxford , Old Road Campus Research Building , Oxford OX3 7DQ , United Kingdom
| | - Gray Kueberuwa
- Department of Oncology , University of Oxford , Old Road Campus Research Building , Oxford OX3 7DQ , United Kingdom
| | - Andrew S Ishizuka
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases , National Institutes of Health, 40 Convent Drive , Bethesda , Maryland 20892 , United States
| | - Neeha Zaidi
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases , National Institutes of Health, 40 Convent Drive , Bethesda , Maryland 20892 , United States
| | - Ramiro A Ramirez-Valdez
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases , National Institutes of Health, 40 Convent Drive , Bethesda , Maryland 20892 , United States
| | - Nicolas J Blobel
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases , National Institutes of Health, 40 Convent Drive , Bethesda , Maryland 20892 , United States
| | - Faezzah Baharom
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases , National Institutes of Health, 40 Convent Drive , Bethesda , Maryland 20892 , United States
| | - Joseph Leal
- Department of Immunology , University of Washington , South Lake Union E-411, 750 Republican Street , Seattle , Washington 98109 , United States
| | - Amy Q Wang
- Therapeutics for Rare and Neglected Diseases, National Center for Advancing Translational Sciences , 9800 Medical Center Drive , Rockville , Maryland 20850 , United States
| | - Michael Y Gerner
- Department of Immunology , University of Washington , South Lake Union E-411, 750 Republican Street , Seattle , Washington 98109 , United States
| | - Tomáš Etrych
- Institute of Macromolecular Chemistry, Czech Academy of Sciences , Heyrovského nám. 2 , 162 06 Prague 6 , Czech Republic
| | - Karel Ulbrich
- Institute of Macromolecular Chemistry, Czech Academy of Sciences , Heyrovského nám. 2 , 162 06 Prague 6 , Czech Republic
| | - Leonard W Seymour
- Department of Oncology , University of Oxford , Old Road Campus Research Building , Oxford OX3 7DQ , United Kingdom
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases , National Institutes of Health, 40 Convent Drive , Bethesda , Maryland 20892 , United States
| | - Richard Laga
- Institute of Macromolecular Chemistry, Czech Academy of Sciences , Heyrovského nám. 2 , 162 06 Prague 6 , Czech Republic
- Department of Oncology , University of Oxford , Old Road Campus Research Building , Oxford OX3 7DQ , United Kingdom
| |
Collapse
|
49
|
Ssemaganda A, Giddam AK, Zaman M, Skwarczynski M, Toth I, Stanisic DI, Good MF. Induction of Plasmodium-Specific Immune Responses Using Liposome-Based Vaccines. Front Immunol 2019; 10:135. [PMID: 30774635 PMCID: PMC6367261 DOI: 10.3389/fimmu.2019.00135] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 01/16/2019] [Indexed: 12/30/2022] Open
Abstract
In the development of vaccines, the ability to initiate both innate and subsequent adaptive immune responses need to be considered. Live attenuated vaccines achieve this naturally, while inactivated and sub-unit vaccines generally require additional help provided through delivery systems and/or adjuvants. Liposomes present an attractive adjuvant/delivery system for antigens. Here, we review the key aspects of immunity against Plasmodium parasites, liposome design considerations and their current application in the development of a malaria vaccine.
Collapse
Affiliation(s)
| | | | - Mehfuz Zaman
- Institute for Glycomics, Griffith University, Southport, QLD, Australia
| | - Mariusz Skwarczynski
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
- School of Pharmacy, The University of Queensland, Brisbane, QLD, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | | | - Michael F. Good
- Institute for Glycomics, Griffith University, Southport, QLD, Australia
| |
Collapse
|
50
|
Wang P, Škalamera Đ, Sui X, Zhang P, Michalek SM. Synthesis and Evaluation of a QS-17/18-Based Vaccine Adjuvant. J Med Chem 2019; 62:1669-1676. [PMID: 30656932 DOI: 10.1021/acs.jmedchem.8b01997] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
We have synthesized a QS-17/18 analogue (7) and evaluated its adjuvant activity in the formulation with rHagB antigen. Compound 7 and QS-21 analogues 5 and 6 are presumably the major components of GPI-0100, a widely used complex mixture of semisynthetic derivatives of Quillaja saponaria (QS) Molina saponins. The QS-17/18 analogue 7 shows an adjuvant activity profile similar to that of GPI-0100, potentiating mixed Th-1/Th-2 immune responses, which is different from those of QS-21 analogues 5 and 6 that probably only induce a Th2-like immunity. The combination of QS-17/18 and QS-21 analogues does not show a synergistic effect. These results suggest that QS-17/18 analogue 7 might be the active component of GPI-0100 responsible for its immunostimulant property. Therefore, compound 7 can not only be a structurally defined alternative to GPI-0100 but also provide a valuable clue for rational design of new QS-based vaccine adjuvants with better adjuvant properties.
Collapse
|