1
|
Khalifa AZ, Perrie Y, Shahiwala A. Subunit antigen delivery: emulsion and liposomal adjuvants for next-generation vaccines. Expert Opin Drug Deliv 2025; 22:583-597. [PMID: 40021342 DOI: 10.1080/17425247.2025.2474088] [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: 03/18/2024] [Accepted: 02/26/2025] [Indexed: 03/03/2025]
Abstract
INTRODUCTION Developing new vaccines to combat emerging infectious diseases has gained more significance after the COVID-19 pandemic. Vaccination is the most cost-effective method for preventing infectious diseases, and subunit antigens are a safer alternative to traditional live, attenuated, and inactivated vaccines. AREAS COVERED Challenges in delivering subunit antigens and the status of different vaccine adjuvants. Recent research developments involving emulsion and liposomal adjuvants and their compositions and properties affecting their adjuvancy. EXPERT OPINION Lipid-based adjuvants, e.g. emulsions and liposomes, represent a paradigm shift in vaccine technology by enabling robust humoral and cellular immune responses with lower antigen doses, a property that is particularly critical during pandemics or in resource-limited settings. These adjuvants can optimize vaccine administration strategies by potentially reducing the frequency of booster doses, thereby improving patient compliance and lowering healthcare costs. While emulsions excel in dose-sparing and broadening immune responses, liposomes offer customization and precision in antigen delivery. However, the broader clinical application of these technologies is not without challenges. Stability issues, e.g. the susceptibility of emulsion-based adjuvants to freezing and their reliance on cold-chain logistics, pose significant barriers to their use in remote/underserved regions. Future developments will likely focus on improving manufacturing scalability and cost-effectiveness.
Collapse
Affiliation(s)
- Al Zahraa Khalifa
- Department of Pharmaceutical Sciences, Dubai Pharmacy College for Girls, Dubai Medical University, Dubai, United Arab Emirates
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Yvonne Perrie
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Aliasgar Shahiwala
- Department of Pharmaceutical Sciences, Dubai Pharmacy College for Girls, Dubai Medical University, Dubai, United Arab Emirates
| |
Collapse
|
2
|
Vega Rojas LJ, Ruíz-Manzano RA, Velasco-Elizondo MA, Carbajo-Mata MA, Hernández-Silva DJ, Rocha-Solache M, Hernández J, Pérez-Serrano RM, Zaldívar-Lelo de Larrea G, García-Gasca T, Mosqueda J. An Evaluation of the Cellular and Humoral Response of a Multi-Epitope Vaccine Candidate Against COVID-19 with Different Alum Adjuvants. Pathogens 2024; 13:1081. [PMID: 39770342 PMCID: PMC11728595 DOI: 10.3390/pathogens13121081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 01/16/2025] Open
Abstract
SARS-CoV-2 (Betacoronavirus pandemicum) is responsible for the disease identified by the World Health Organization (WHO) as COVID-19. We designed "CHIVAX 2.1", a multi-epitope vaccine, containing ten immunogenic peptides with conserved B-cell and T-cell epitopes in the receceptor binding domain (RBD) sequences of different SARS-CoV-2 variants of concern (VoCs). We evaluated the immune response of mice immunized with 20 or 60 µg of the chimeric protein with two different alum adjuvants (Alhydrogel® and Adju-Phos®), plus PHAD®, in a two-immunization regimen (0 and 21 days). Serum samples were collected on days 0, 21, 31, and 72 post first immunization, with antibody titers determined by indirect ELISA, while lymphoproliferation assays and cytokine production were evaluated by flow cytometry. The presence of neutralizing antibodies was assessed by surrogate neutralization assays. Higher titers of total IgG, IgG1, and IgG2a antibodies, as well as increased proliferation rates of specific CD4+ and CD8+ T cells, were observed in mice immunized with 60 μg of protein plus Adju-Phos®/PHAD®. This formulation also generated the highest levels of TNF-α and IFN-γ, in addition to the presence of neutralizing antibodies against Delta and Omicron VoC. These findings indicate the potential of this chimeric multi-epitope vaccine with combined adjuvants as a promising platform against viral infections, eliciting a TH1 or TH1:TH2 balanced cell response.
Collapse
MESH Headings
- Animals
- Mice
- COVID-19 Vaccines/immunology
- COVID-19/prevention & control
- COVID-19/immunology
- SARS-CoV-2/immunology
- Alum Compounds/pharmacology
- Alum Compounds/administration & dosage
- Adjuvants, Immunologic/pharmacology
- Adjuvants, Immunologic/administration & dosage
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Immunity, Humoral/drug effects
- Immunity, Humoral/immunology
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Female
- Epitopes, T-Lymphocyte/immunology
- Immunity, Cellular/drug effects
- Immunity, Cellular/immunology
- Mice, Inbred BALB C
- Adjuvants, Vaccine/pharmacology
- Epitopes, B-Lymphocyte/immunology
- Humans
- Immunoglobulin G/blood
- Immunoglobulin G/immunology
- CD8-Positive T-Lymphocytes/immunology
- Cytokines/metabolism
- Spike Glycoprotein, Coronavirus/immunology
Collapse
Affiliation(s)
- Lineth Juliana Vega Rojas
- Immunology and Vaccines Laboratory, Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Campus Aeropuerto, Carretera a Chichimequillas, Ejido Bolaños, Querétaro 76140, Mexico; (L.J.V.R.); (R.A.R.-M.); (M.A.V.-E.); (D.J.H.-S.); (M.R.-S.)
- Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT), Av. Insurgentes Sur 1582, Alcaldía Benito Juárez, Crédito Constructor, Ciudad de México 03940, Mexico
| | - Rocío Alejandra Ruíz-Manzano
- Immunology and Vaccines Laboratory, Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Campus Aeropuerto, Carretera a Chichimequillas, Ejido Bolaños, Querétaro 76140, Mexico; (L.J.V.R.); (R.A.R.-M.); (M.A.V.-E.); (D.J.H.-S.); (M.R.-S.)
| | - Miguel Andrés Velasco-Elizondo
- Immunology and Vaccines Laboratory, Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Campus Aeropuerto, Carretera a Chichimequillas, Ejido Bolaños, Querétaro 76140, Mexico; (L.J.V.R.); (R.A.R.-M.); (M.A.V.-E.); (D.J.H.-S.); (M.R.-S.)
| | - María Antonieta Carbajo-Mata
- Instituto de Neurobiología UNAM, Laboratorio Universitario del Bioterio, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico
| | - Diego Josimar Hernández-Silva
- Immunology and Vaccines Laboratory, Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Campus Aeropuerto, Carretera a Chichimequillas, Ejido Bolaños, Querétaro 76140, Mexico; (L.J.V.R.); (R.A.R.-M.); (M.A.V.-E.); (D.J.H.-S.); (M.R.-S.)
| | - Mariana Rocha-Solache
- Immunology and Vaccines Laboratory, Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Campus Aeropuerto, Carretera a Chichimequillas, Ejido Bolaños, Querétaro 76140, Mexico; (L.J.V.R.); (R.A.R.-M.); (M.A.V.-E.); (D.J.H.-S.); (M.R.-S.)
| | - Jesús Hernández
- Laboratorio de Inmunología, Centro de Investigación en Alimentación y Desarrollo, A.C, Hermosillo 83304, Mexico;
| | - Rosa Martha Pérez-Serrano
- Advanced Biomedical Research Center, School of Medicine, Universidad Autónoma de Querétaro, Querétaro 76176, Mexico; (R.M.P.-S.); (G.Z.-L.d.L.)
| | - Guadalupe Zaldívar-Lelo de Larrea
- Advanced Biomedical Research Center, School of Medicine, Universidad Autónoma de Querétaro, Querétaro 76176, Mexico; (R.M.P.-S.); (G.Z.-L.d.L.)
| | - Teresa García-Gasca
- Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Av. de las Ciencias s/n, Juriquilla, Querétaro 76230, Mexico
| | - Juan Mosqueda
- Immunology and Vaccines Laboratory, Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Campus Aeropuerto, Carretera a Chichimequillas, Ejido Bolaños, Querétaro 76140, Mexico; (L.J.V.R.); (R.A.R.-M.); (M.A.V.-E.); (D.J.H.-S.); (M.R.-S.)
| |
Collapse
|
3
|
Wang N, Wang C, Wei C, Chen M, Gao Y, Zhang Y, Wang T. Constructing the cGAMP-Aluminum Nanoparticles as a Vaccine Adjuvant-Delivery System (VADS) for Developing the Efficient Pulmonary COVID-19 Subunit Vaccines. Adv Healthc Mater 2024; 13:e2401650. [PMID: 39319481 DOI: 10.1002/adhm.202401650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 09/05/2024] [Indexed: 09/26/2024]
Abstract
The cGAMP-aluminum nanoparticles (CAN) are engineered as a vaccine adjuvant-delivery system to carry mixed RBD (receptor-binding domain) of the original severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its new variant for developing bivalent pulmonary coronavirus disease 2019 (COVID-19) vaccines (biRBD-CAN). High phosphophilicity/adsorptivity made intrapulmonary CAN instantly form the pulmonary ingredient-coated CAN (piCAN) to possess biomimetic features enhancing biocompatibility. In vitro biRBD-CAN sparked APCs (antigen-presenting cells) to mature and make extra reactive oxygen species, engendered lysosome escape effects and enhanced proteasome activities. Through activating the intracellular stimulator of interferon genes (STING) and nucleotide-binding domain and leucine-rich repeat and pyrin domain containing proteins 3 (NALP3) inflammasome pathways to exert synergy between cGAMP and AN, biRBD-CAN stimulated APCs to secret cytokines favoring mixed Th1/Th2 immunoresponses. Mice bearing twice intrapulmonary biRBD-CAN produced high levels of mucosal antibodies, the long-lasting systemic antibodies, and potent cytotoxic T lymphocytes which efficiently erased cells displaying cognate epitopes. Notably, biRBD-CAN existed in mouse lungs and different lymph nodes for at least 48 h, unveiling their sustained immunostimulatory activity as the main mechanism underlying the long-lasting immunity and memory. Hamsters bearing twice intrapulmonary biRBD-CAN developed high resistance to pseudoviral challenges performed using different recombinant strains including the ones with distinct SARS-CoV-2-spike mutations. Thus, biRBD-CAN as a broad-spectrum pulmonary COVID-19 vaccine candidate may provide a tool for controlling the emerging SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Ning Wang
- School of Food and Bioengineering, Hefei University of Technology, 420 Jade Road, Hefei, Anhui Province, 230601, China
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province, 230032, China
| | - Can Wang
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province, 230032, China
- Department of Pharmacy, The Second People's Hospital of Lianyungang, 41 Hailian East Road, Lianyungang, Jiangsu Province, 222006, China
| | - Chunliu Wei
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province, 230032, China
| | - Minnan Chen
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province, 230032, China
| | - Yuhao Gao
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province, 230032, China
| | - Yuxi Zhang
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province, 230032, China
| | - Ting Wang
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province, 230032, China
| |
Collapse
|
4
|
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
|
5
|
Hellfritzsch M, Christensen D, Foged C, Scherließ R, Thakur A. Reconstituted dry powder formulations of ZnO-adjuvanted ovalbumin induce equivalent antigen specific antibodies but lower T cell responses than ovalbumin adjuvanted with Alhydrogel® or cationic adjuvant formulation 01 (CAF®01). Int J Pharm 2023; 648:123581. [PMID: 37931728 DOI: 10.1016/j.ijpharm.2023.123581] [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: 08/11/2023] [Revised: 10/28/2023] [Accepted: 11/03/2023] [Indexed: 11/08/2023]
Abstract
Most licensed human vaccines are based on liquid dosage forms but have poor storage stability and require continuous and expensive cold-chain storage. In contrast, the use of solid vaccine dosage forms produced by for example spray drying, extends shelf life and eliminates the need for a cold chain. Zinc oxide (ZnO)-based nanoparticles display immunomodulatory properties, but their adjuvant effect as a dry powder formulation is unknown. Here, we show that reconstituted dry powder formulations of ZnO particles containing the model antigen ovalbumin (OVA) induce antigen-specific CD8+ T-cell and humoral responses. By systematically varying the ratio between ZnO and mannitol during spray drying, we manufactured dry powder formulations of OVA-containing ZnO particles that displayed: (i) a spherical or wrinkled surface morphology, (ii) an aerodynamic diameter and particle size distribution optimal for deep lung deposition, and (iii) aerosolization properties suitable for lung delivery. Reconstituted dry powder formulations of ZnO particles were well-tolerated by Calu-3 lung epithelial cells. Furthermore, almost equivalent OVA-specific serum antibody responses were stimulated by reconstituted ZnO particles, OVA adjuvanted with Alhydrogel®, and OVA adjuvanted with the cationic adjuvant formulation 01 (CAF®01). However, reconstituted dry powder ZnO particles and OVA adjuvanted with Alhydrogel® induced significantly lower OVA-specific CD8+CD44+ T-cell responses in the spleen than OVA adjuvanted with CAF®01. Similarly, reconstituted dry powder ZnO particles activated significantly lower percentages of follicular helper T cells and germinal center B cells in the draining lymph nodes than OVA adjuvanted with CAF®01. Overall, our results show that reconstituted dry powder formulations of ZnO nanoparticles can induce antigen-specific antibodies and can be used in vaccines to enhance antigen-specific humoral immune responses against subunit protein antigens.
Collapse
Affiliation(s)
- Marie Hellfritzsch
- Department of Pharmaceutics and Biopharmaceutics, Kiel University, Grasweg 9a, 24118 Kiel, Germany
| | - Dennis Christensen
- Department of Infectious Disease Immunology, Statens Serum Institut, Artillerivej 5, 2300 Copenhagen S, Denmark
| | - Camilla Foged
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Regina Scherließ
- Department of Pharmaceutics and Biopharmaceutics, Kiel University, Grasweg 9a, 24118 Kiel, Germany.
| | - Aneesh Thakur
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark.
| |
Collapse
|
6
|
Nguyen TP, Do Q, Phan LT, Dinh DV, Khong H, Hoang LV, Nguyen TV, Pham HN, Chu MV, Nguyen TT, Pham QD, Le TM, Trang TN, Dinh TT, Vo TV, Vu TT, Nguyen QB, Phan VT, Nguyen LV, Nguyen GT, Tran PM, Nghiem TD, Tran TV, Nguyen TG, Tran TQ, Nguyen LT, Do AT, Nguyen DD, Ho SA, Nguyen VT, Pham DT, Tran HB, Vu ST, Hoang SX, Do TM, Nguyen XT, Le GQ, Tran T, Cao TM, Dao HM, Nguyen TT, Doan UY, Le VT, Tran LP, Nguyen NM, Nguyen NT, Pham HT, Nguyen QH, Nguyen HT, Nguyen HL, Tran VT, Tran MT, Nguyen TT, Ha PT, Huynh HT, Nguyen KD, Thuan UT, Doan CC, Do SM. Safety and immunogenicity of Nanocovax, a SARS-CoV-2 recombinant spike protein vaccine: Interim results of a double-blind, randomised controlled phase 1 and 2 trial. THE LANCET REGIONAL HEALTH. WESTERN PACIFIC 2022; 24:100474. [PMID: 35602004 PMCID: PMC9108376 DOI: 10.1016/j.lanwpc.2022.100474] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
BACKGROUND Nanocovax is a recombinant severe acute respiratory syndrome coronavirus 2 subunit vaccine composed of full-length prefusion stabilized recombinant SARS-CoV-2 spike glycoproteins (S-2P) and aluminium hydroxide adjuvant. METHODS We conducted a dose-escalation, open label trial (phase 1) and a randomized, double-blind, placebo-controlled trial (phase 2) to evaluate the safety and immunogenicity of the Nanocovax vaccine (in 25 mcg, 50 mcg, and 75 mcg doses, aluminium hydroxide adjuvanted (0·5 mg/dose) in 2-dose regime, 28 days apart (ClinicalTrials.gov number, NCT04683484). In phase 1, 60 participants received two intramuscular injection of the vaccine following dose-escalation procedure. The primary outcomes were reactogenicity and laboratory tests to evaluate the vaccine safety. In phase 2, 560 healthy adults received either vaccine doses similar in phase 1 (25 or 50 or 75 mcg S antigen in 0·5 mg aluminium per dose) or adjuvant (0·5 mg aluminium) in a ratio of 2:2:2:1. One primary outcome was the vaccine safety, including solicited adverse events for 7 day and unsolicited adverse events for 28 days after each injection as well as serious adverse event or adverse events of special interest throughout the study period. Another primary outcome was anti-S IgG antibody response (Index unit/ml). Secondary outcomes were surrogate virus neutralisation (inhibition percentage), wild-type SARS-CoV-2 neutralisation (dilution fold), and T-cell responses by intracellular staining for interferon gamma (IFNg). Anti-S IgG and neutralising antibody levels were compared with convalescent serum samples from symptomatic Covid-19 patients. FINDINGS For phase 1 study, no serious adverse events were observed for all 60 participants. Most adverse events were grade 1 and disappeared shortly after injection. For phase 2 study, after randomisation, 480 participants were assigned to receive the vaccine with adjuvant, and 80 participants were assigned to receive the placebo (adjuvant only). Reactogenicity was absent or mild in the majority of participants and of short duration (mean ≤3 days). Unsolicited adverse events were mild in most participants. There were no serious adverse events related to Nanocovax. Regarding the immunogenicity, Nanocovax induced robust anti-S antibody responses. In general, there humoral responses were similar among vaccine groups which reached their peaks at day 42 and declined afterward. At day 42, IgG levels of vaccine groups were 60·48 [CI95%: 51·12-71·55], 49·11 [41·26-58·46], 57·18 [48·4-67·5] compared to 7·10 [6·32-13·92] of convalescent samples. IgG levels reported here can be converted to WHO international standard binding antibody unit (BAU/ml) by multiplying them to a conversion factor of 21·8. Neutralising antibody titre of vaccine groups at day 42 were 89·2 [52·2-152·3], 80·0 [50·8-125.9] and 95·1 [63·1-143·6], compared to 55·1 [33·4-91·0] of the convalescent group. INTERPRETATION Up to day 90, Nanocovax was found to be safe, well tolerated, and induced robust immune responses. FUNDING This work was funded by the Coalition for Epidemic Preparedness Innovations (CEPI), the Ministry of Science and Technology of Vietnam, and Nanogen Pharmaceutical Biotechnology JSC.
Collapse
Affiliation(s)
- Thuy P. Nguyen
- Nanogen Pharmaceutical Biotechnology JSC, Lot I-5C Saigon Hitech Park, Ho Chi Minh City, Viet Nam
| | - Quyet Do
- Vietnam Military Medical University, 160 Phung Hung, Ha Dong, Ha Noi, Viet Nam
| | - Lan T. Phan
- Pasteur Institute, 167 Pasteur, District 3, Ho Chi Minh City, Viet Nam
| | - Duc V. Dinh
- Vietnam Military Medical University, 160 Phung Hung, Ha Dong, Ha Noi, Viet Nam
| | - Hiep Khong
- Nanogen Pharmaceutical Biotechnology JSC, Lot I-5C Saigon Hitech Park, Ho Chi Minh City, Viet Nam
| | - Luong V. Hoang
- Vietnam Military Medical University, 160 Phung Hung, Ha Dong, Ha Noi, Viet Nam
| | - Thuong V. Nguyen
- Pasteur Institute, 167 Pasteur, District 3, Ho Chi Minh City, Viet Nam
| | - Hung N. Pham
- Vietnam Military Medical University, 160 Phung Hung, Ha Dong, Ha Noi, Viet Nam
| | - Men V. Chu
- Vietnam Military Medical University, 160 Phung Hung, Ha Dong, Ha Noi, Viet Nam
| | - Toan T. Nguyen
- Pasteur Institute, 167 Pasteur, District 3, Ho Chi Minh City, Viet Nam
| | - Quang D. Pham
- Pasteur Institute, 167 Pasteur, District 3, Ho Chi Minh City, Viet Nam
| | - Tri M. Le
- Nanogen Pharmaceutical Biotechnology JSC, Lot I-5C Saigon Hitech Park, Ho Chi Minh City, Viet Nam
| | - Tuyen N.T. Trang
- Nanogen Pharmaceutical Biotechnology JSC, Lot I-5C Saigon Hitech Park, Ho Chi Minh City, Viet Nam
| | - Thanh T. Dinh
- Nanogen Pharmaceutical Biotechnology JSC, Lot I-5C Saigon Hitech Park, Ho Chi Minh City, Viet Nam
| | - Thuong V. Vo
- Nanogen Pharmaceutical Biotechnology JSC, Lot I-5C Saigon Hitech Park, Ho Chi Minh City, Viet Nam
| | - Thao T. Vu
- Nanogen Pharmaceutical Biotechnology JSC, Lot I-5C Saigon Hitech Park, Ho Chi Minh City, Viet Nam
| | - Quynh B.P. Nguyen
- Nanogen Pharmaceutical Biotechnology JSC, Lot I-5C Saigon Hitech Park, Ho Chi Minh City, Viet Nam
| | - Vuong T. Phan
- Nanogen Pharmaceutical Biotechnology JSC, Lot I-5C Saigon Hitech Park, Ho Chi Minh City, Viet Nam
| | - Luong V. Nguyen
- Vietnam Military Medical University, 160 Phung Hung, Ha Dong, Ha Noi, Viet Nam
| | - Giang T. Nguyen
- Vietnam Military Medical University, 160 Phung Hung, Ha Dong, Ha Noi, Viet Nam
| | - Phong M. Tran
- Vietnam Military Medical University, 160 Phung Hung, Ha Dong, Ha Noi, Viet Nam
| | - Thuan D. Nghiem
- Vietnam Military Medical University, 160 Phung Hung, Ha Dong, Ha Noi, Viet Nam
| | - Tien V. Tran
- Vietnam Military Medical University, 160 Phung Hung, Ha Dong, Ha Noi, Viet Nam
| | - Tien G. Nguyen
- Vietnam Military Medical University, 160 Phung Hung, Ha Dong, Ha Noi, Viet Nam
| | - Tuynh Q. Tran
- Vietnam Military Medical University, 160 Phung Hung, Ha Dong, Ha Noi, Viet Nam
| | - Linh T. Nguyen
- Vietnam Military Medical University, 160 Phung Hung, Ha Dong, Ha Noi, Viet Nam
| | - Anh T. Do
- Vietnam Military Medical University, 160 Phung Hung, Ha Dong, Ha Noi, Viet Nam
| | - Dung D. Nguyen
- Vietnam Military Medical University, 160 Phung Hung, Ha Dong, Ha Noi, Viet Nam
| | - Son A. Ho
- Vietnam Military Medical University, 160 Phung Hung, Ha Dong, Ha Noi, Viet Nam
| | - Viet T. Nguyen
- Vietnam Military Medical University, 160 Phung Hung, Ha Dong, Ha Noi, Viet Nam
| | - Dung T. Pham
- Vietnam Military Medical University, 160 Phung Hung, Ha Dong, Ha Noi, Viet Nam
| | - Hieu B. Tran
- Vietnam Military Medical University, 160 Phung Hung, Ha Dong, Ha Noi, Viet Nam
| | - Son T. Vu
- Vietnam Military Medical University, 160 Phung Hung, Ha Dong, Ha Noi, Viet Nam
| | - Su X. Hoang
- Vietnam Military Medical University, 160 Phung Hung, Ha Dong, Ha Noi, Viet Nam
| | - Trung M. Do
- Vietnam Military Medical University, 160 Phung Hung, Ha Dong, Ha Noi, Viet Nam
| | - Xuan T. Nguyen
- Vietnam Military Medical University, 160 Phung Hung, Ha Dong, Ha Noi, Viet Nam
| | - Giang Q. Le
- Vietnam Military Medical University, 160 Phung Hung, Ha Dong, Ha Noi, Viet Nam
| | - Ton Tran
- Pasteur Institute, 167 Pasteur, District 3, Ho Chi Minh City, Viet Nam
| | - Thang M. Cao
- Pasteur Institute, 167 Pasteur, District 3, Ho Chi Minh City, Viet Nam
| | - Huy M. Dao
- Pasteur Institute, 167 Pasteur, District 3, Ho Chi Minh City, Viet Nam
| | - Thao T.T. Nguyen
- Pasteur Institute, 167 Pasteur, District 3, Ho Chi Minh City, Viet Nam
| | - Uyen Y Doan
- Pasteur Institute, 167 Pasteur, District 3, Ho Chi Minh City, Viet Nam
| | - Vy T.T. Le
- Pasteur Institute, 167 Pasteur, District 3, Ho Chi Minh City, Viet Nam
| | - Linh P. Tran
- Pasteur Institute, 167 Pasteur, District 3, Ho Chi Minh City, Viet Nam
| | - Ngoc M. Nguyen
- Pasteur Institute, 167 Pasteur, District 3, Ho Chi Minh City, Viet Nam
| | - Ngoc T. Nguyen
- Pasteur Institute, 167 Pasteur, District 3, Ho Chi Minh City, Viet Nam
| | - Hang T.T. Pham
- Pasteur Institute, 167 Pasteur, District 3, Ho Chi Minh City, Viet Nam
| | - Quan H. Nguyen
- Pasteur Institute, 167 Pasteur, District 3, Ho Chi Minh City, Viet Nam
| | - Hieu T. Nguyen
- Pasteur Institute, 167 Pasteur, District 3, Ho Chi Minh City, Viet Nam
| | - Hang L.K. Nguyen
- National Institute of Hygiene and Epidemiology (NIHE), Ha Noi, Viet Nam
| | - Vinh T. Tran
- Nanogen Pharmaceutical Biotechnology JSC, Lot I-5C Saigon Hitech Park, Ho Chi Minh City, Viet Nam
| | - Mai T.N. Tran
- Nanogen Pharmaceutical Biotechnology JSC, Lot I-5C Saigon Hitech Park, Ho Chi Minh City, Viet Nam
| | - Truc T.T. Nguyen
- Nanogen Pharmaceutical Biotechnology JSC, Lot I-5C Saigon Hitech Park, Ho Chi Minh City, Viet Nam
| | - Phat T. Ha
- Nanogen Pharmaceutical Biotechnology JSC, Lot I-5C Saigon Hitech Park, Ho Chi Minh City, Viet Nam
| | - Hieu T. Huynh
- Nanogen Pharmaceutical Biotechnology JSC, Lot I-5C Saigon Hitech Park, Ho Chi Minh City, Viet Nam
| | - Khanh D. Nguyen
- Nanogen Pharmaceutical Biotechnology JSC, Lot I-5C Saigon Hitech Park, Ho Chi Minh City, Viet Nam
| | - Ung T. Thuan
- Nanogen Pharmaceutical Biotechnology JSC, Lot I-5C Saigon Hitech Park, Ho Chi Minh City, Viet Nam
| | - Chung C. Doan
- Nanogen Pharmaceutical Biotechnology JSC, Lot I-5C Saigon Hitech Park, Ho Chi Minh City, Viet Nam
| | - Si M. Do
- Nanogen Pharmaceutical Biotechnology JSC, Lot I-5C Saigon Hitech Park, Ho Chi Minh City, Viet Nam
- Corresponding author.
| |
Collapse
|
7
|
Forsyth VS, Himpsl SD, Smith SN, Sarkissian CA, Mike LA, Stocki JA, Sintsova A, Alteri CJ, Mobley HLT. Optimization of an Experimental Vaccine To Prevent Escherichia coli Urinary Tract Infection. mBio 2020; 11:e00555-20. [PMID: 32345645 PMCID: PMC7188996 DOI: 10.1128/mbio.00555-20] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 03/30/2020] [Indexed: 12/12/2022] Open
Abstract
Urinary tract infections (UTI) affect half of all women at least once during their lifetime. The rise in the numbers of extended-spectrum beta-lactamase-producing strains and the potential for carbapenem resistance within uropathogenic Escherichia coli (UPEC), the most common causative agent of UTI, create an urgent need for vaccine development. Intranasal immunization of mice with UPEC outer membrane iron receptors FyuA, Hma, IreA, and IutA, conjugated to cholera toxin, provides protection in the bladder or kidneys under conditions of challenge with UPEC strain CFT073 or strain 536. On the basis of these data, we sought to optimize the vaccination route (intramuscular, intranasal, or subcutaneous) in combination with adjuvants suitable for human use, including aluminum hydroxide gel (alum), monophosphoryl lipid A (MPLA), unmethylated CpG synthetic oligodeoxynucleotides (CpG), polyinosinic:polycytidylic acid (polyIC), and mutated heat-labile E. coli enterotoxin (dmLT). Mice intranasally vaccinated with dmLT-IutA and dmLT-Hma displayed significant reductions in bladder colonization (86-fold and 32-fold, respectively), with 40% to 42% of mice having no detectable CFU. Intranasal vaccination of mice with CpG-IutA and polyIC-IutA significantly reduced kidney colonization (131-fold) and urine CFU (22-fold), respectively. dmLT generated the most consistently robust antibody response in intranasally immunized mice, while MPLA and alum produced greater concentrations of antigen-specific serum IgG with intramuscular immunization. On the basis of these results, we conclude that intranasal administration of Hma or IutA formulated with dmLT adjuvant provides the greatest protection from UPEC UTI. This report advances our progress toward a vaccine against uncomplicated UTI, which will significantly improve the quality of life for women burdened by recurrent UTI and enable better antibiotic stewardship.IMPORTANCE Urinary tract infections (UTI) are among the most common bacterial infection in humans, affecting half of all women at least once during their lifetimes. The rise in antibiotic resistance and health care costs emphasizes the need to develop a vaccine against the most common UTI pathogen, Escherichia coli Vaccinating mice intranasally with a detoxified heat-labile enterotoxin and two surface-exposed receptors, Hma or IutA, significantly reduced bacterial burden in the bladder. This work highlights progress in the development of a UTI vaccine formulated with adjuvants suitable for human use and antigens that encode outer membrane iron receptors required for infection in the iron-limited urinary tract.
Collapse
Affiliation(s)
- Valerie S Forsyth
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Stephanie D Himpsl
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Sara N Smith
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Christina A Sarkissian
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Laura A Mike
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Jolie A Stocki
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Anna Sintsova
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Christopher J Alteri
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Natural Sciences, University of Michigan-Dearborn, Dearborn, Michigan, USA
| | - Harry L T Mobley
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
8
|
Reyna-Margarita HR, Irais CM, Mario-Alberto RG, Agustina RM, Luis-Benjamín SG, David PE. Plant Phenolics and Lectins as Vaccine Adjuvants. Curr Pharm Biotechnol 2019; 20:1236-1243. [DOI: 10.2174/1389201020666190716110705] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 06/24/2019] [Accepted: 07/04/2019] [Indexed: 11/22/2022]
Abstract
Background:
The immune system is responsible for providing protection to the body
against foreign substances. The immune system divides into two types of immune responses to study
its mechanisms of protection: 1) Innate and 2) Adaptive. The innate immune response represents the
first protective barrier of the organism that also works as a regulator of the adaptive immune response,
if evaded the mechanisms of the innate immune response by the foreign substance the adaptive immune
response takes action with the consequent antigen neutralization or elimination. The adaptive
immune response objective is developing a specific humoral response that consists in the production of
soluble proteins known as antibodies capable of specifically recognizing the foreign agent; such protective
mechanism is induced artificially through an immunization or vaccination. Unfortunately, the
immunogenicity of the antigens is an intrinsic characteristic of the same antigen dependent on several
factors.
Conclusion:
Vaccine adjuvants are chemical substances of very varied structure that seek to improve
the immunogenicity of antigens. The main four types of adjuvants under investigation are the following:
1) Oil emulsions with an antigen in solution, 2) Pattern recognition receptors activating molecules,
3) Inflammatory stimulatory molecules or activators of the inflammasome complex, and 4) Cytokines.
However, this paper addresses the biological plausibility of two phytochemical compounds as vaccine
adjuvants: 5) Lectins, and 6) Plant phenolics whose characteristics, mechanisms of action and disadvantages
are addressed. Finally, the immunological usefulness of these molecules is discussed through
immunological data to estimate effects of plant phenolics and lectins as vaccine adjuvants, and current
studies that have implanted these molecules as vaccine adjuvants, demonstrating the results of this
immunization.
Collapse
Affiliation(s)
- Hernández-Ramos Reyna-Margarita
- Departamento de Bioquimica, Centro de Investigacion Biomedica de la Facultad de Medicina Unidad Torreon. Universidad Autonoma de Coahuila (UA de C), Torreon, Mexico
| | - Castillo-Maldonado Irais
- Departamento de Bioquimica, Centro de Investigacion Biomedica de la Facultad de Medicina Unidad Torreon. Universidad Autonoma de Coahuila (UA de C), Torreon, Mexico
| | - Rivera-Guillén Mario-Alberto
- Departamento de Bioquimica, Centro de Investigacion Biomedica de la Facultad de Medicina Unidad Torreon. Universidad Autonoma de Coahuila (UA de C), Torreon, Mexico
| | - Ramírez-Moreno Agustina
- Facultad de Ciencias Biologicas Unidad Torreon, Universidad Autonoma de Coahuila (UA de C), Torreon, Mexico
| | - Serrano-Gallardo Luis-Benjamín
- Departamento de Bioquimica, Centro de Investigacion Biomedica de la Facultad de Medicina Unidad Torreon. Universidad Autonoma de Coahuila (UA de C), Torreon, Mexico
| | - Pedroza-Escobar David
- Departamento de Bioquimica, Centro de Investigacion Biomedica de la Facultad de Medicina Unidad Torreon. Universidad Autonoma de Coahuila (UA de C), Torreon, Mexico
| |
Collapse
|
9
|
HogenEsch H, O'Hagan DT, Fox CB. Optimizing the utilization of aluminum adjuvants in vaccines: you might just get what you want. NPJ Vaccines 2018; 3:51. [PMID: 30323958 PMCID: PMC6180056 DOI: 10.1038/s41541-018-0089-x] [Citation(s) in RCA: 278] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 09/06/2018] [Accepted: 09/11/2018] [Indexed: 02/01/2023] Open
Abstract
Aluminum-containing adjuvants have been used for over 90 years to enhance the immune response to vaccines. Recent work has significantly advanced our understanding of the physical, chemical, and biological properties of these adjuvants, offering key insights on underlying mechanisms. Given the long-term success of aluminum adjuvants, we believe that they should continue to represent the “gold standard” against which all new adjuvants should be compared. New vaccine candidates that require adjuvants to induce a protective immune responses should first be evaluated with aluminum adjuvants before other more experimental approaches are considered, since use of established adjuvants would facilitate both clinical development and the regulatory pathway. However, the continued use of aluminum adjuvants requires an appreciation of their complexities, in combination with access to the necessary expertise to optimize vaccine formulations. In this article, we will review the properties of aluminum adjuvants and highlight those elements that are critical to optimize vaccine performance. We will discuss how other components (excipients, TLR ligands, etc.) can affect the interaction between adjuvants and antigens, and impact the potency of vaccines. This review provides a resource and guide, which will ultimately contribute to the successful development of newer, more effective and safer vaccines.
Collapse
Affiliation(s)
- Harm HogenEsch
- 1Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN USA.,2Purdue Institute of Inflammation, Immunology and Infectious Diseases, Purdue University, West Lafayette, IN USA
| | | | - Christopher B Fox
- 4IDRI, Seattle, WA USA.,5Department of Global Health, University of Washington, Seattle, WA USA
| |
Collapse
|
10
|
Khong H, Overwijk WW. Adjuvants for peptide-based cancer vaccines. J Immunother Cancer 2016; 4:56. [PMID: 27660710 PMCID: PMC5028954 DOI: 10.1186/s40425-016-0160-y] [Citation(s) in RCA: 129] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 08/12/2016] [Indexed: 12/12/2022] Open
Abstract
Cancer therapies based on T cells have shown impressive clinical benefit. In particular, immune checkpoint blockade therapies with anti-CTLA-4 and anti-PD-1/PD-L1 are causing dramatic tumor shrinkage and prolonged patient survival in a variety of cancers. However, many patients do not benefit, possibly due to insufficient spontaneous T cell reactivity against their tumors and/or lacking immune cell infiltration to tumor site. Such tumor-specific T cell responses could be induced through anti-cancer vaccination; but despite great success in animal models, only a few of many cancer vaccine trials have demonstrated robust clinical benefit. One reason for this difference may be the use of potent, effective vaccine adjuvants in animal models, vs. the use of safe, but very weak, vaccine adjuvants in clinical trials. As vaccine adjuvants dictate the type and magnitude of the T cell response after vaccination, it is critical to understand how they work to design safe, but also effective, cancer vaccines for clinical use. Here we discuss current insights into the mechanism of action and practical application of vaccine adjuvants, with a focus on peptide-based cancer vaccines.
Collapse
Affiliation(s)
- Hiep Khong
- Department of Melanoma Medical Oncology, University of Texas - MD Anderson Cancer Center, South Campus Research Building 1, 1515 Holcombe Blvd, Houston, TX 77030 USA ; Immunology program - University of Texas - Graduate School of Biomedical Sciences at Houston, 6767 Bertner Ave, Houston, TX 77030 USA
| | - Willem W Overwijk
- Department of Melanoma Medical Oncology, University of Texas - MD Anderson Cancer Center, South Campus Research Building 1, 1515 Holcombe Blvd, Houston, TX 77030 USA ; Immunology program - University of Texas - Graduate School of Biomedical Sciences at Houston, 6767 Bertner Ave, Houston, TX 77030 USA
| |
Collapse
|
11
|
Portuondo DL, Batista-Duharte A, Ferreira LS, Martínez DT, Polesi MC, Duarte RA, de Paula E Silva ACA, Marcos CM, Almeida AMFD, Carlos IZ. A cell wall protein-based vaccine candidate induce protective immune response against Sporothrix schenckii infection. Immunobiology 2015; 221:300-9. [PMID: 26547105 DOI: 10.1016/j.imbio.2015.10.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 10/03/2015] [Accepted: 10/23/2015] [Indexed: 11/24/2022]
Abstract
Sporotrichosis is a subcutaneous mycosis caused by several closely related thermo-dimorphic fungi of the Sporothrix schenckii species complex, affecting humans and other mammals. In the last few years, new strategies have been proposed for controlling sporotrichosis owning to concerns about its growing incidence in humans, cats, and dogs in Brazil, as well as the toxicity and limited efficacy of conventional antifungal drugs. In this study, we assessed the immunogenicity and protective properties of two aluminum hydroxide (AH)-adsorbed S. schenckii cell wall protein (ssCWP)-based vaccine formulations in a mouse model of systemic S. schenckii infection. Fractioning by SDS-PAGE revealed nine protein bands, two of which were functionally characterized: a 44kDa peptide hydrolase and a 47kDa enolase, which was predicted to be an adhesin. Sera from immunized mice recognized the 47kDa enolase and another unidentified 71kDa protein, whereas serum from S. schenckii-infected mice recognized both these proteins plus another unidentified 9.4kDa protein. Furthermore, opsonization with the anti-ssCWP sera led to markedly increased phagocytosis and was able to strongly inhibit the fungus' adhesion to fibroblasts. Immunization with the higher-dose AH-adjuvanted formulation led to increased ex vivo release of IL-12, IFN-γ, IL-4, and IL-17, whereas only IL-12 and IFN-γ were induced by the higher-dose non-adjuvanted formulation. Lastly, passive transference of the higher-dose AH-adjuvanted formulation's anti-ssCWP serum was able to afford in vivo protection in a subsequent challenge with S. schenckii, becoming a viable vaccine candidate for further testing.
Collapse
Affiliation(s)
- Deivys Leandro Portuondo
- Department of Clinical Analysis, Araraquara's School of Pharmaceutical Sciences, Universidade Estadual Paulista-UNESP, Júlio Mesquita Filho, Rua Expedicionários do Brasil, 1621, Postal Code: 14801-902, Araraquara, SP, Brazil.
| | - Alexander Batista-Duharte
- Immunotoxicology Laboratory, Toxicology and Biomedicine Center (TOXIMED), Medical Science University, Autopista Nacional Km. 1 1/2CP 90400, AP 4033 Santiago de Cuba, Cuba.
| | - Lucas Souza Ferreira
- Department of Clinical Analysis, Araraquara's School of Pharmaceutical Sciences, Universidade Estadual Paulista-UNESP, Júlio Mesquita Filho, Rua Expedicionários do Brasil, 1621, Postal Code: 14801-902, Araraquara, SP, Brazil.
| | - Damiana Téllez Martínez
- Department of Clinical Analysis, Araraquara's School of Pharmaceutical Sciences, Universidade Estadual Paulista-UNESP, Júlio Mesquita Filho, Rua Expedicionários do Brasil, 1621, Postal Code: 14801-902, Araraquara, SP, Brazil.
| | - Marisa Campos Polesi
- Department of Clinical Analysis, Araraquara's School of Pharmaceutical Sciences, Universidade Estadual Paulista-UNESP, Júlio Mesquita Filho, Rua Expedicionários do Brasil, 1621, Postal Code: 14801-902, Araraquara, SP, Brazil.
| | - Roberta Aparecida Duarte
- Department of Clinical Analysis, Araraquara's School of Pharmaceutical Sciences, Universidade Estadual Paulista-UNESP, Júlio Mesquita Filho, Rua Expedicionários do Brasil, 1621, Postal Code: 14801-902, Araraquara, SP, Brazil.
| | - Ana Carolina Alves de Paula E Silva
- Department of Clinical Analysis, Araraquara's School of Pharmaceutical Sciences, Universidade Estadual Paulista-UNESP, Júlio Mesquita Filho, Rua Expedicionários do Brasil, 1621, Postal Code: 14801-902, Araraquara, SP, Brazil.
| | - Caroline Maria Marcos
- Department of Clinical Analysis, Araraquara's School of Pharmaceutical Sciences, Universidade Estadual Paulista-UNESP, Júlio Mesquita Filho, Rua Expedicionários do Brasil, 1621, Postal Code: 14801-902, Araraquara, SP, Brazil.
| | - Ana Marisa Fusco de Almeida
- Department of Clinical Analysis, Araraquara's School of Pharmaceutical Sciences, Universidade Estadual Paulista-UNESP, Júlio Mesquita Filho, Rua Expedicionários do Brasil, 1621, Postal Code: 14801-902, Araraquara, SP, Brazil.
| | - Iracilda Zeppone Carlos
- Department of Clinical Analysis, Araraquara's School of Pharmaceutical Sciences, Universidade Estadual Paulista-UNESP, Júlio Mesquita Filho, Rua Expedicionários do Brasil, 1621, Postal Code: 14801-902, Araraquara, SP, Brazil.
| |
Collapse
|
12
|
The mechanisms of action of vaccines containing aluminum adjuvants: an in vitro vs in vivo paradigm. SPRINGERPLUS 2015; 4:181. [PMID: 25932368 PMCID: PMC4406982 DOI: 10.1186/s40064-015-0972-0] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 04/08/2015] [Indexed: 12/23/2022]
Abstract
Adjuvants such as the aluminum compounds (alum) have been dominantly used in many vaccines due to their immunopotentiation and safety records since 1920s. However, how these mineral agents influence the immune response to vaccination remains elusive. Many hypotheses exist as to the mode of action of these adjuvants, such as depot formation, antigen (Ag) targeting, and the induction of inflammation. These hypotheses are based on many in vitro and few in vivo studies. Understanding how cells interact with adjuvants in vivo will be crucial to fully understanding the mechanisms of action of these adjuvants. Interestingly, how alum influences the target cell at both the cellular and molecular level, and the consequent innate and adaptive responses, will be critical in the rational design of effective vaccines against many diseases. Thus, in this review, mechanisms of action of alum have been discussed based on available in vitro vs in vivo evidences to date.
Collapse
|
13
|
Portuondo DLF, Ferreira LS, Urbaczek AC, Batista-Duharte A, Carlos IZ. Adjuvants and delivery systems for antifungal vaccines: Current state and future developments. Med Mycol 2014; 53:69-89. [DOI: 10.1093/mmy/myu045] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
|
14
|
|
15
|
Vecchi S, Bufali S, Skibinski DAG, O'Hagan DT, Singh M. Aluminum adjuvant dose guidelines in vaccine formulation for preclinical evaluations. J Pharm Sci 2011; 101:17-20. [PMID: 21918987 DOI: 10.1002/jps.22759] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Revised: 08/13/2011] [Accepted: 08/23/2011] [Indexed: 11/07/2022]
Abstract
Aluminum (Al) salt-based adjuvants are present in a large variety of licensed vaccines and their use is widely considered for formulations in clinical trials. Although the regulatory agencies have clearly stated the acceptable levels of Al salts in vaccines for human use, there are no general indications for preclinical research. This brief commentary reviews the current status of Al concentrations in licensed vaccines, the related potential toxicity in preclinical species, and proposes a general guideline for selection of suitable Al salt levels in preclinical models, focusing on the formulation development for recombinant protein antigens. A table with conversion factors is included in order to provide a tool for calculation of doses with different Al salts.
Collapse
Affiliation(s)
- Simone Vecchi
- Novartis Vaccines and Diagnostics, Siena 53100, Italy.
| | | | | | | | | |
Collapse
|