1
|
Li M, Sui J, Wang X, Song C, Cao X, Sun X, Zhao R, Wang S, Qin L, Wang Y, Liu K, Zhao S, Huo N. Single-walled carbon nanotube-protein complex: A strategy to improve the immune response to protein in mice. Vaccine 2024; 42:126013. [PMID: 38834429 DOI: 10.1016/j.vaccine.2024.05.061] [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: 10/27/2023] [Revised: 04/30/2024] [Accepted: 05/25/2024] [Indexed: 06/06/2024]
Abstract
Vaccines represent an effective tool for controlling disease infection. As a key component of vaccines, many types of adjuvants have been developed and used today. This study is designed to investigate the efficacy of single-walled carbon nanotubes (SWCNTs) as a new adjuvant. The results showed that SWCNT could adsorb the antigen by intermolecular action, and the adsorption rate was significantly higher after dispersion of the SWCNTs in a sonic bath. The titer of specific antibody of mice in the SWCNTs group was higher than that of the mice in the antigen control group, confirming the adjuvant efficacy of SWCNTs. During immunisation, the specific antibody was detected earlier in the mice of the SWCNTs group, especially when the amount of antigen was reduced. And it was proved that the titer of antibodies was higher after subcutaneous and intraperitoneal injection compared to intramuscular injection. Most importantly, the mice immunised with SWCNTs showed almost the same level of immunity as the mice in the FCA (Freund's complete adjuvant) group, indicating that the SWCNTs were an effective adjuvant. In addition, the mice in the SWCNT group maintained antibody levels for 90 days after the last booster vaccination and showed a good state of health during the observed period. We also found that the SWCNTs were able to induce macrophages activation and enhance antigen uptake by mouse peritoneal macrophages.
Collapse
Affiliation(s)
- Muzi Li
- Laboratory of Quality and Safety Risk Assessment for Animal Products of Ministry of Agriculture, China Animal Health and Epidemiology Center, Qingdao, Shandong 266032, China
| | - Jinyu Sui
- Laboratory of Quality and Safety Risk Assessment for Animal Products of Ministry of Agriculture, China Animal Health and Epidemiology Center, Qingdao, Shandong 266032, China
| | - Xiaoyin Wang
- Laboratory of Quality and Safety Risk Assessment for Animal Products of Ministry of Agriculture, China Animal Health and Epidemiology Center, Qingdao, Shandong 266032, China
| | - Cuiping Song
- Laboratory of Quality and Safety Risk Assessment for Animal Products of Ministry of Agriculture, China Animal Health and Epidemiology Center, Qingdao, Shandong 266032, China
| | - Xumin Cao
- Laboratory of Quality and Safety Risk Assessment for Animal Products of Ministry of Agriculture, China Animal Health and Epidemiology Center, Qingdao, Shandong 266032, China
| | - Xiaoliang Sun
- Laboratory of Quality and Safety Risk Assessment for Animal Products of Ministry of Agriculture, China Animal Health and Epidemiology Center, Qingdao, Shandong 266032, China
| | - Ruimin Zhao
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030800, China
| | - Shuting Wang
- Laboratory of Quality and Safety Risk Assessment for Animal Products of Ministry of Agriculture, China Animal Health and Epidemiology Center, Qingdao, Shandong 266032, China
| | - Lide Qin
- Laboratory of Quality and Safety Risk Assessment for Animal Products of Ministry of Agriculture, China Animal Health and Epidemiology Center, Qingdao, Shandong 266032, China
| | - Yudong Wang
- Laboratory of Quality and Safety Risk Assessment for Animal Products of Ministry of Agriculture, China Animal Health and Epidemiology Center, Qingdao, Shandong 266032, China
| | - Kun Liu
- Laboratory of Quality and Safety Risk Assessment for Animal Products of Ministry of Agriculture, China Animal Health and Epidemiology Center, Qingdao, Shandong 266032, China
| | - Sijun Zhao
- Laboratory of Quality and Safety Risk Assessment for Animal Products of Ministry of Agriculture, China Animal Health and Epidemiology Center, Qingdao, Shandong 266032, China.
| | - Nairui Huo
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030800, China.
| |
Collapse
|
2
|
Zhong K, Chen X, Zhang J, Jiang X, Zhang J, Huang M, Bi S, Ju C, Luo Y. Recent Advances in Oral Vaccines for Animals. Vet Sci 2024; 11:353. [PMID: 39195807 PMCID: PMC11360704 DOI: 10.3390/vetsci11080353] [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: 07/01/2024] [Revised: 07/29/2024] [Accepted: 08/02/2024] [Indexed: 08/29/2024] Open
Abstract
Compared to traditional injected vaccines, oral vaccines offer significant advantages for the immunization of livestock and wildlife due to their ease of use, high compliance, improved safety, and potential to stimulate mucosal immune responses and induce systemic immunity against pathogens. This review provides an overview of the delivery methods for oral vaccines, and the factors that influence their immunogenicity. We also highlight the global progress and achievements in the development and use of oral vaccines for animals, shedding light on potential future applications in this field.
Collapse
Affiliation(s)
- Kaining Zhong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510640, China; (K.Z.); (X.C.); (J.Z.); (X.J.); (J.Z.); (M.H.)
| | - Xinting Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510640, China; (K.Z.); (X.C.); (J.Z.); (X.J.); (J.Z.); (M.H.)
| | - Junhao Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510640, China; (K.Z.); (X.C.); (J.Z.); (X.J.); (J.Z.); (M.H.)
| | - Xiaoyu Jiang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510640, China; (K.Z.); (X.C.); (J.Z.); (X.J.); (J.Z.); (M.H.)
| | - Junhui Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510640, China; (K.Z.); (X.C.); (J.Z.); (X.J.); (J.Z.); (M.H.)
| | - Minyi Huang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510640, China; (K.Z.); (X.C.); (J.Z.); (X.J.); (J.Z.); (M.H.)
| | - Shuilian Bi
- School of Food Science, Guangdong Pharmaceutical University, Zhongshan 528458, China;
| | - Chunmei Ju
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510640, China; (K.Z.); (X.C.); (J.Z.); (X.J.); (J.Z.); (M.H.)
- Key Laboratory of Animal Vaccine Development of the Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou 510640, China
| | - Yongwen Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510640, China; (K.Z.); (X.C.); (J.Z.); (X.J.); (J.Z.); (M.H.)
- Key Laboratory of Animal Vaccine Development of the Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou 510640, China
| |
Collapse
|
3
|
Jang JH, Lee JE, Kim KT, Ahn DU, Paik HD. Immunostimulatory Effect of Ovomucin Hydrolysates by Pancreatin in RAW 264.7 Macrophages via Mitogen-Activated Protein Kinase (MAPK) Signaling Pathway. Food Sci Anim Resour 2024; 44:885-898. [PMID: 38974730 PMCID: PMC11222692 DOI: 10.5851/kosfa.2024.e25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 02/26/2024] [Accepted: 03/17/2024] [Indexed: 07/09/2024] Open
Abstract
Ovomucin (OM), which has insoluble fractions is a viscous glycoprotein, found in egg albumin. Enzymatic hydrolysates of OM have water solubility and bioactive properties. This study investigated that the immunostimulatory effects of OM hydrolysates (OMHs) obtained by using various proteolytic enzymes (Alcalase®, bromelain, α-chymotrypsin, Neutrase®, pancreatin, papain, Protamax®, and trypsin) in RAW 264.7 cells. The results showed that OMH prepared with pancreatin (OMPA) produced the highest levels of nitrite oxide in RAW 264.7 cells, through upregulation of inducible nitric oxide synthase mRNA expression. The production of pro-inflammatory cytokines such as tumor necrosis factor-α and interleukin-6 were increased with the cytokines mRNA expression. The effect of OMPA on mitogen-activated protein kinase signaling pathway was increased the phosphorylation of p38, c-Jun NH2-terminal kinase, and extracellular signal-regulated kinase in a concentration-dependent manner. Therefore, OMPA could be used as a potential immune-stimulating agent in the functional food industry.
Collapse
Affiliation(s)
- Jin-Hong Jang
- Department of Food Science and
Biotechnology of Animal Resources, Konkuk University, Seoul
05029, Korea
| | - Ji-Eun Lee
- Department of Food Science and
Biotechnology of Animal Resources, Konkuk University, Seoul
05029, Korea
| | - Kee-Tae Kim
- Research Institute, WithBio
Inc, Seoul 05029, Korea
| | - Dong Uk Ahn
- Department of Animal Science, Iowa State
University, Ames, IA 50011, USA
| | - Hyun-Dong Paik
- Department of Food Science and
Biotechnology of Animal Resources, Konkuk University, Seoul
05029, Korea
| |
Collapse
|
4
|
Alonso-Cerda MJ, García-Soto MJ, Miranda-López A, Segura-Velázquez R, Sánchez-Betancourt JI, González-Ortega O, Rosales-Mendoza S. Layered Double Hydroxides (LDH) as Delivery Vehicles of a Chimeric Protein Carrying Epitopes from the Porcine Reproductive and Respiratory Syndrome Virus. Pharmaceutics 2024; 16:841. [PMID: 39065539 PMCID: PMC11279870 DOI: 10.3390/pharmaceutics16070841] [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: 05/18/2024] [Revised: 06/14/2024] [Accepted: 06/18/2024] [Indexed: 07/28/2024] Open
Abstract
The Porcine Reproductive and Respiratory Syndrome Virus (PRRSV) causes reproductive failure and respiratory symptoms, leading to huge economic losses for the pig farming industry. Although several vaccines against PRRSV are available in the market; they show an overall low efficacy, and several countries have the need for vaccines covering the local, circulating variants. This project aims at developing a new chimeric antigen targeting specific epitopes from PRRSV and evaluating two test adjuvants to formulate a vaccine candidate. The test antigen was called LTB-PRRSV, which was produced recombinantly in Escherichia coli and consisted of the heat labile enterotoxin B subunit from E. coli (LTB) and four epitopes from PRRSV. LTB-PRRSV was rescued as inclusion bodies and methods for its solubilization, IMAC-based purification, and refolding were standardized, leading to mean yields of 18 mg of pure protein per liter culture. Layered double hydroxides (LDH) have been used as vaccine adjuvants given their biocompatibility, low cost, and positive surface charge that allows an efficient adsorption of negatively charged biomolecules. Therefore, LDH were selected as delivery vehicles of LTB-PRRSV. Pure LTB-PRRSV was adsorbed onto LDH by incubation at different LDH:LTB-PRRSV mass ratios (1:0.25, 1:0.5, 1:1, and 1:2) and at pH 9.5. The best adsorption occurred with a 1:2 mass ratio, and in a sucrose-tween solution. The conjugates obtained had a polydispersity index of 0.26, a hydrodynamic diameter of 192 nm, and a final antigen concentration of 64.2 μg/mL. An immunogenicity assessment was performed by injecting mice with LDH:LTB-PRRSV, Alum/LTB-PRRSV, or LTB-PRRSV in a scheme comprising three immunizations at two-week intervals and two dose levels (1 and 5 μg). LTB-PRRSV was capable of inducing strong humoral responses, which lasted for a longer period when LDH was used as the delivery vehicle/adjuvant. The potential of LDH to serve as an attractive carrier for veterinary vaccines is discussed.
Collapse
Affiliation(s)
- María José Alonso-Cerda
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Manuel Nava 6, San Luis Potosí 78210, Mexico; (M.J.A.-C.); (M.J.G.-S.); (A.M.-L.)
- Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, San Luis Potosí 78210, Mexico
| | - Mariano J. García-Soto
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Manuel Nava 6, San Luis Potosí 78210, Mexico; (M.J.A.-C.); (M.J.G.-S.); (A.M.-L.)
| | - Arleth Miranda-López
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Manuel Nava 6, San Luis Potosí 78210, Mexico; (M.J.A.-C.); (M.J.G.-S.); (A.M.-L.)
- Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, San Luis Potosí 78210, Mexico
| | - René Segura-Velázquez
- Unidad de Investigación, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
| | - José Ivan Sánchez-Betancourt
- Departamento de Medicina y Zootecnia de Cerdos, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Omar González-Ortega
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Manuel Nava 6, San Luis Potosí 78210, Mexico; (M.J.A.-C.); (M.J.G.-S.); (A.M.-L.)
- Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, San Luis Potosí 78210, Mexico
| | - Sergio Rosales-Mendoza
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Manuel Nava 6, San Luis Potosí 78210, Mexico; (M.J.A.-C.); (M.J.G.-S.); (A.M.-L.)
- Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, San Luis Potosí 78210, Mexico
| |
Collapse
|
5
|
Thomazini VC, da Cunha GM, Guimarães NM, Saleme SD, de Melo RCG, de Paula GA, Carvalho SG, Chorilli M, Dos Santos Giuberti C, Villanova JCO. Impact of concerning excipients on animal safety: insights for veterinary pharmacotherapy and regulatory considerations. Daru 2024; 32:289-305. [PMID: 37903944 PMCID: PMC11087455 DOI: 10.1007/s40199-023-00486-8] [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: 06/21/2023] [Accepted: 10/09/2023] [Indexed: 11/01/2023] Open
Abstract
OBJECTIVES Veterinarians and pharmacists are familiar with the efficacy and safety aspects attributed to active pharmaceutical ingredients included in medicines, but they are rarely concerned with the safety of excipients present in medicines. Although generally recognized as safe, excipients are not chemically inert and may produce adverse events in certain animal populations. This review aims to present excipients of concern to these populations and highlight their relevance for rational veterinary pharmacotherapy. EVIDENCE ACQUISITION A comprehensive review of the literature about the existence of adverse reactions in animals caused by pharmaceutical excipients was carried out based on an exploratory study. An overview of the correct conditions of use and safety of these excipients has also been provided, with information about their function, the proportion in which they are included in the different pharmaceutical dosage forms and the usual routes of administration. RESULTS We identified 18 excipients considered of concern due to their potential to cause harm to the health of specific animal populations: bentonite, benzalkonium chloride, benzoic acid, benzyl alcohol, ethanol, lactose, mannitol, mineral oil, monosodium glutamate, polyethylene glycol, polysorbate, propylene glycol, sodium benzoate, sodium carboxymethylcellulose, sodium lauryl sulfate, sulfites, polyoxyethylene castor oil derivatives, and xylitol. Among the 135 manuscripts listed, only 24 referred to studies in which the substances were correctly evaluated as excipients. CONCLUSIONS Based on the information presented in this review, the authors hope to draw the attention of professionals involved in veterinary pharmacotherapy to the existence of excipients of concern in medicines. This information contributes to rational veterinary pharmacotherapy and supports veterinary pharmacovigilance actions. We hope to shed light on the subject and encourage studies and new manuscripts that address the safety of pharmaceutical excipients to the animal population.
Collapse
Affiliation(s)
- Vanessa Cola Thomazini
- Graduate Program in Veterinary Sciences, Center for Agricultural Sciences and Engineering - CCAE, Federal University of Espírito Santo - UFES, Alegre, ES, 29500-000, Brazil
| | - Gabriel Mendes da Cunha
- Pharmaceutical Product Development Laboratory, Center of Exact, Natural and Health Sciences - CCENS, Federal University of Espírito Santo - UFES, Alegre, ES, 29500-000, Brazil
| | - Nayhara Madeira Guimarães
- Graduate Program in Veterinary Sciences, Center for Agricultural Sciences and Engineering - CCAE, Federal University of Espírito Santo - UFES, Alegre, ES, 29500-000, Brazil
| | - Soraya Dias Saleme
- Graduate Program in Veterinary Sciences, Center for Agricultural Sciences and Engineering - CCAE, Federal University of Espírito Santo - UFES, Alegre, ES, 29500-000, Brazil
| | - Rita Cristina Gonçalves de Melo
- Graduate Program in Pharmaceutical Sciences, Center of Health Sciences - CCS, Federal University of Espírito Santo - UFES, Vitória, ES, 29047-105, Brazil
| | - Geanne Aparecida de Paula
- Department of Drugs and Pharmaceutics, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, SP, 14800-903, Brazil
| | - Suzana Gonçalves Carvalho
- Department of Drugs and Pharmaceutics, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, SP, 14800-903, Brazil
| | - Marlus Chorilli
- Department of Drugs and Pharmaceutics, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, SP, 14800-903, Brazil
| | - Cristiane Dos Santos Giuberti
- Pharmaceutical Product Development Laboratory, Center of Exact, Natural and Health Sciences - CCENS, Federal University of Espírito Santo - UFES, Alegre, ES, 29500-000, Brazil
| | - Janaina Cecília Oliveira Villanova
- Graduate Program in Veterinary Sciences, Center for Agricultural Sciences and Engineering - CCAE, Federal University of Espírito Santo - UFES, Alegre, ES, 29500-000, Brazil.
- Pharmaceutical Product Development Laboratory, Center of Exact, Natural and Health Sciences - CCENS, Federal University of Espírito Santo - UFES, Alegre, ES, 29500-000, Brazil.
- Graduate Program in Pharmaceutical Sciences, Center of Health Sciences - CCS, Federal University of Espírito Santo - UFES, Vitória, ES, 29047-105, Brazil.
| |
Collapse
|
6
|
Korangath P, Jin L, Yang CT, Healy S, Guo X, Ke S, Grüttner C, Hu C, Gabrielson K, Foote J, Clarke R, Ivkov R. Iron Oxide Nanoparticles Inhibit Tumor Progression and Suppress Lung Metastases in Mouse Models of Breast Cancer. ACS NANO 2024; 18:10509-10526. [PMID: 38564478 PMCID: PMC11025112 DOI: 10.1021/acsnano.3c12064] [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: 12/01/2023] [Revised: 03/07/2024] [Accepted: 03/13/2024] [Indexed: 04/04/2024]
Abstract
Systemic exposure to starch-coated iron oxide nanoparticles (IONPs) can stimulate antitumor T cell responses, even when little IONP is retained within the tumor. Here, we demonstrate in mouse models of metastatic breast cancer that IONPs can alter the host immune landscape, leading to systemic immune-mediated disease suppression. We report that a single intravenous injection of IONPs can inhibit primary tumor growth, suppress metastases, and extend survival. Gene expression analysis revealed the activation of Toll-like receptor (TLR) pathways involving signaling via Toll/Interleukin-1 receptor domain-containing adaptor-inducing IFN-β (TRIF), a TLR pathway adaptor protein. Requisite participation of TRIF in suppressing tumor progression was demonstrated with histopathologic evidence of upregulated IFN-regulatory factor 3 (IRF3), a downstream protein, and confirmed in a TRIF knockout syngeneic mouse model of metastatic breast cancer. Neither starch-coated polystyrene nanoparticles lacking iron, nor iron-containing dextran-coated parenteral iron replacement agent, induced significant antitumor effects, suggesting a dependence on the type of IONP formulation. Analysis of multiple independent clinical databases supports a hypothesis that upregulation of TLR3 and IRF3 correlates with increased overall survival among breast cancer patients. Taken together, these data support a compelling rationale to re-examine IONP formulations as harboring anticancer immune (nano)adjuvant properties to generate a therapeutic benefit without requiring uptake by cancer cells.
Collapse
Affiliation(s)
- Preethi Korangath
- Department
of Radiation Oncology and Molecular Radiation Sciences, School of
Medicine, Johns Hopkins University, Baltimore, Maryland 21231, United States
| | - Lu Jin
- The
Hormel Institute, University of Minnesota, Austin, Minnesota 55912, United States
| | - Chun-Ting Yang
- Department
of Radiation Oncology and Molecular Radiation Sciences, School of
Medicine, Johns Hopkins University, Baltimore, Maryland 21231, United States
| | - Sean Healy
- Department
of Radiation Oncology and Molecular Radiation Sciences, School of
Medicine, Johns Hopkins University, Baltimore, Maryland 21231, United States
| | - Xin Guo
- Department
of Molecular and Comparative Pathobiology, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205, United States
| | - Suqi Ke
- Department
of Biostatistics and Bioinformatics, Sidney Kimmel Comprehensive Cancer
Centre, School of Medicine, Johns Hopkins
University, Baltimore, Maryland 21231, United States
| | | | - Chen Hu
- Department
of Biostatistics and Bioinformatics, Sidney Kimmel Comprehensive Cancer
Centre, School of Medicine, Johns Hopkins
University, Baltimore, Maryland 21231, United States
| | - Kathleen Gabrielson
- Department
of Molecular and Comparative Pathobiology, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205, United States
| | - Jeremy Foote
- Department
of Microbiology, School of Medicine, University
of Alabama at Birmingham, Birmingham, Alabama 35294, United States
| | - Robert Clarke
- The
Hormel Institute, University of Minnesota, Austin, Minnesota 55912, United States
| | - Robert Ivkov
- Department
of Radiation Oncology and Molecular Radiation Sciences, School of
Medicine, Johns Hopkins University, Baltimore, Maryland 21231, United States
- Department
of Oncology, Sidney Kimmel Comprehensive Cancer Centre, School of
Medicine, Johns Hopkins University, Baltimore, Maryland 21231, United States
- Department
of Mechanical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Department
of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
| |
Collapse
|
7
|
Zhang R, Wei Y, Liu X, Wu Y. Development and efficacy evaluation of a novel water-in-oil-in-water adjuvant for an inactivated foot-and-mouth disease vaccine. Pharm Dev Technol 2024; 29:75-85. [PMID: 38217108 DOI: 10.1080/10837450.2024.2305107] [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: 08/29/2023] [Accepted: 01/10/2024] [Indexed: 01/15/2024]
Abstract
To develop a novel water-in-oil-in-water (W/O/W) adjuvant and evaluate the effect on foot-and-mouth disease (FMD) inactivated vaccine, in this study, we prepared the novel nano-emulsion adjuvant based on QS-21 (BEA) which is composed of the mixture of mineral oil Marcol52, surfactant Tween80, oleate polyoxyethylene ether ester, polyoxyethylene palmitic acid ester and span80, cosurfactant polyethylene glycol and QS-21. The two-step emulsification method formed the W/O/W nano-emulsion with two films and three-phase structures. The effective particle diameter of the BEA was about 184 nm, and it has good thermal stability. Then, BEA was emulsified as an adjuvant to prepare for the inactivated FMDV vaccine, and BALB/c mice and pigs were immunized to evaluate its safety and immunization effect. The results showed that the inactivated BEA-FMDV vaccine significantly increased BALB/c mice and pigs' antibodies and cytokine IFN-γ in serum. Meanwhile, the pig-neutralizing antibodies were higher than control group. Safety tests found no symptoms of FMD or significant toxic reactions. After 28 days of immunization, the protection rate can reach 93.3%. The BEA vaccine had good stability at 4 °C, no stratification after 180 days, and the content of 146S in the vaccine did not decrease. In conclusion, the BEA prepared in this study is suitable for FMDV inactivated vaccine and is an effective adjuvant.
Collapse
Affiliation(s)
- Rong Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
- China Agricultural Vet Biological Science and Technology Co., Ltd, Lanzhou, China
| | - Yanming Wei
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Xuerong Liu
- China Agricultural Vet Biological Science and Technology Co., Ltd, Lanzhou, China
| | - Yongshu Wu
- College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang, A&F University, Hangzhou, China
| |
Collapse
|
8
|
Zumuk CP, Jones MK, Navarro S, Gray DJ, You H. Transmission-Blocking Vaccines against Schistosomiasis Japonica. Int J Mol Sci 2024; 25:1707. [PMID: 38338980 PMCID: PMC10855202 DOI: 10.3390/ijms25031707] [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: 12/23/2023] [Revised: 01/19/2024] [Accepted: 01/21/2024] [Indexed: 02/12/2024] Open
Abstract
Control of schistosomiasis japonica, endemic in Asia, including the Philippines, China, and Indonesia, is extremely challenging. Schistosoma japonicum is a highly pathogenic helminth parasite, with disease arising predominantly from an immune reaction to entrapped parasite eggs in tissues. Females of this species can generate 1000-2200 eggs per day, which is about 3- to 15-fold greater than the egg output of other schistosome species. Bovines (water buffalo and cattle) are the predominant definitive hosts and are estimated to generate up to 90% of parasite eggs released into the environment in rural endemic areas where these hosts and humans are present. Here, we highlight the necessity of developing veterinary transmission-blocking vaccines for bovines to better control the disease and review potential vaccine candidates. We also point out that the approach to producing efficacious transmission-blocking animal-based vaccines before moving on to human vaccines is crucial. This will result in effective and feasible public health outcomes in agreement with the One Health concept to achieve optimum health for people, animals, and the environment. Indeed, incorporating a veterinary-based transmission vaccine, coupled with interventions such as human mass drug administration, improved sanitation and hygiene, health education, and snail control, would be invaluable to eliminating zoonotic schistosomiasis.
Collapse
Affiliation(s)
- Chika P. Zumuk
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (C.P.Z.); (M.K.J.); (S.N.)
- Faculty of Medicine, The University of Queensland, Herston, QLD 4006, Australia
| | - Malcolm K. Jones
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (C.P.Z.); (M.K.J.); (S.N.)
- School of Veterinary Science, The University of Queensland, Gatton, QLD 4343, Australia
| | - Severine Navarro
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (C.P.Z.); (M.K.J.); (S.N.)
- Faculty of Medicine, The University of Queensland, Herston, QLD 4006, Australia
- Centre for Childhood Nutrition Research, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Darren J. Gray
- Population Health Program, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia;
| | - Hong You
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (C.P.Z.); (M.K.J.); (S.N.)
- School of Veterinary Science, The University of Queensland, Gatton, QLD 4343, Australia
| |
Collapse
|
9
|
Abdelaziz K, Helmy YA, Yitbarek A, Hodgins DC, Sharafeldin TA, Selim MSH. Advances in Poultry Vaccines: Leveraging Biotechnology for Improving Vaccine Development, Stability, and Delivery. Vaccines (Basel) 2024; 12:134. [PMID: 38400118 PMCID: PMC10893217 DOI: 10.3390/vaccines12020134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 01/25/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
With the rapidly increasing demand for poultry products and the current challenges facing the poultry industry, the application of biotechnology to enhance poultry production has gained growing significance. Biotechnology encompasses all forms of technology that can be harnessed to improve poultry health and production efficiency. Notably, biotechnology-based approaches have fueled rapid advances in biological research, including (a) genetic manipulation in poultry breeding to improve the growth and egg production traits and disease resistance, (b) rapid identification of infectious agents using DNA-based approaches, (c) inclusion of natural and synthetic feed additives to poultry diets to enhance their nutritional value and maximize feed utilization by birds, and (d) production of biological products such as vaccines and various types of immunostimulants to increase the defensive activity of the immune system against pathogenic infection. Indeed, managing both existing and newly emerging infectious diseases presents a challenge for poultry production. However, recent strides in vaccine technology are demonstrating significant promise for disease prevention and control. This review focuses on the evolving applications of biotechnology aimed at enhancing vaccine immunogenicity, efficacy, stability, and delivery.
Collapse
Affiliation(s)
- Khaled Abdelaziz
- Department of Animal and Veterinary Science, College of Agriculture, Forestry and Life Sciences, Clemson University Poole Agricultural Center, Jersey Ln #129, Clemson, SC 29634, USA
- Clemson University School of Health Research (CUSHR), Clemson, SC 29634, USA
| | - Yosra A. Helmy
- Department of Veterinary Science, Martin-Gatton College of Agriculture, Food, and Environment, University of Kentucky, Lexington, KY 40546, USA;
| | - Alexander Yitbarek
- Department of Animal & Food Sciences, University of Delaware, 531 S College Ave, Newark, DE 19716, USA;
| | - Douglas C. Hodgins
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada;
| | - Tamer A. Sharafeldin
- Department of Veterinary Biomedical Science, Animal Disease Research and Diagnostic Laboratory, South Dakota State University, Brookings, SD 57007, USA; (T.A.S.); (M.S.H.S.)
| | - Mohamed S. H. Selim
- Department of Veterinary Biomedical Science, Animal Disease Research and Diagnostic Laboratory, South Dakota State University, Brookings, SD 57007, USA; (T.A.S.); (M.S.H.S.)
| |
Collapse
|
10
|
Domínguez-Odio A, Rodríguez Martínez E, Cala Delgado DL. Commercial vaccines used in poultry, cattle, and aquaculture: a multidirectional comparison. Front Vet Sci 2024; 10:1307585. [PMID: 38234985 PMCID: PMC10791835 DOI: 10.3389/fvets.2023.1307585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 12/08/2023] [Indexed: 01/19/2024] Open
|
11
|
Shahbazi S, Habibi M, Badmasti F, Sabzi S, Farokhi M, Asadi Karam MR. Design and fabrication of a vaccine candidate based on rOmpA from Klebsiella pneumoniae encapsulated in silk fibroin-sodium alginate nanoparticles against pneumonia infection. Int Immunopharmacol 2023; 125:111171. [PMID: 37948863 DOI: 10.1016/j.intimp.2023.111171] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/14/2023] [Accepted: 10/31/2023] [Indexed: 11/12/2023]
Abstract
The present study describes the design and fabrication of a novel vaccine candidate based on the outer membrane protein A (rOmpA) from Klebsiella pneumoniae (K. pneumoniae) encapsulated in silk fibroin-sodium alginate nanoparticles (SF-SANPs) against K. pneumoniae-mediated pneumonia. The physicochemical properties, toxicity, release profile, and in vivo potency of SF-SANPs encapsulated with rOmpA were evaluated. The spherical nano vaccine was created with an average particle size of 160 nm and an encapsulation efficiency of 80 %. Antigen release from SF-SANPs was 40 % after 22 days release assay. The SF-SANPs showed a zeta potential of -24.8 mV and had no toxic effect on the L929 cells in vitro. It was found that SF-SANPs in the vaccine formulation promoted systemic and mucosal antibodies and also stimulated cytokine responses, inducing both humoral (Th2) and cellular (Th1) immune responses, with a Th1-polarized response. The vaccine candidate was effective in protecting the mice lung against experimental pneumonia and reducing inflammation. These findings suggest that the rOmpA-based vaccine encapsulated in SF-SANPs could be a promising strategy for preventing pneumonia caused by K. pneumoniae.
Collapse
Affiliation(s)
- Shahla Shahbazi
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, Iran
| | - Mehri Habibi
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, Iran
| | - Farzad Badmasti
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran
| | - Samira Sabzi
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, Iran
| | - Mehdi Farokhi
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran.
| | | |
Collapse
|
12
|
Madkhali OA. Drug Delivery of Gelatin Nanoparticles as a Biodegradable Polymer for the Treatment of Infectious Diseases: Perspectives and Challenges. Polymers (Basel) 2023; 15:4327. [PMID: 37960007 PMCID: PMC10648051 DOI: 10.3390/polym15214327] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/31/2023] [Accepted: 11/03/2023] [Indexed: 11/15/2023] Open
Abstract
In recent years, there has been a growing interest in the use of gelatin nanoparticles (GNPs) for the treatment of infectious diseases. The inherent properties of these nanoparticles make them attractive options for drug delivery. Their biocompatibility ensures that they can interact with biological systems without causing adverse reactions, while their biodegradability ensures that they can break down harmlessly in the body once their function is performed. Furthermore, their capacity for controlled drug release ensures that therapeutic agents can be delivered over a sustained period, thereby enhancing treatment efficacy. This review examines the current landscape of GNP-based drug delivery, with a specific focus on its potential applications and challenges in the context of infectious diseases. Key challenges include controlling drug release rates, ensuring nanoparticle stability under physiological conditions, scaling up production while maintaining quality, mitigating potential immunogenic reactions, optimizing drug loading efficiency, and tracking the biodistribution and clearance of GNPs in the body. Despite these hurdles, GNPs hold promising potential in the realm of infectious disease treatment. Ongoing research and innovation are essential to overcome these obstacles and completely harness the potential of GNPs in clinical applications.
Collapse
Affiliation(s)
- Osama A Madkhali
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan 45124, Saudi Arabia
| |
Collapse
|
13
|
Mosadegh S, Abtahi H, Amani J, Karizi SZ, Salmanian AH. Protective immunization against Enterohemorrhagic Escherichia coli and Shigella dysenteriae Type 1 by chitosan nanoparticle loaded with recombinant chimeric antigens comprising EIT and STX1B-IpaD. Microb Pathog 2023; 184:106344. [PMID: 37704060 DOI: 10.1016/j.micpath.2023.106344] [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: 07/05/2023] [Revised: 09/09/2023] [Accepted: 09/10/2023] [Indexed: 09/15/2023]
Abstract
Increasing evidence demonstrated that Enterohemorrhagic Escherichia coli (EHEC) and Shigella dysenteriae type 1 (S. dysenteriae1) are considered pathogens, that are connected with diarrhea and are still the greatest cause of death in children under the age of five years, worldwide. EHEC and S. dysenteriae 1 infections can be prevented and managed using a vaccination strategy against pathogen attachment stages. In this study, the chitosan nanostructures were loaded with recombinant EIT and STX1B-IpaD polypeptides. The immunogenic properties of this nano-vaccine candidate were investigated. The EIT and STX1B-IpaD recombinant proteins were heterologous expressed, purified, and confirmed by western blotting. The chitosan nanoparticles, were used to encapsulate the purified proteins. The immunogenicity of recombinant nano vaccine candidate, was examined in three groups of BalB/c mice by injection, oral delivery, and combination of oral-injection. ELISA and antibody titer, evaluated the humoral immune response. Finally, all three mice groups were challenged by two pathogens to test the ability of the nano-vaccine candidate to protect against bacterial infection. The Sereny test in guinea pigs was used to confirm the neutralizing effect of immune sera in controlling S. dysenteriae 1, infections. SDS-PAGE and western blotting, confirmed the presence and specificity of 63 and 27 kDa recombinant EIT and STX1B-IpaD, respectively. The results show that the nanoparticles containing recombinant proteins could stimulate the systemic and mucosal immune systems by producing IgG and IgA, respectively. The challenge test showed that, the candidate nano-vaccine could protect the animal model from bacterial infection. The combination of multiple recombinant proteins, carrying several epitopes and natural nanoparticles could evocate remarkable humoral and mucosal responses and improve the protection properties of synthetic antigens. Furthermore, compared with other available antigen delivery methods, using oral delivery as immune priming and injection as a booster method, could act as combinatorial methods to achieve a higher level of immunity. This approach could present an appropriate vaccine candidate against both EHEC and S. dysenteriae 1.
Collapse
Affiliation(s)
- Shadi Mosadegh
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Hamid Abtahi
- Molecular and Medicine Research Center, Arak University of Medical Sciences, Arak, Iran
| | - Jafar Amani
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Shohreh Zare Karizi
- Department of Biology, Varamin Pishva Branch, Islamic Azad University, Pishva, Varamin, Iran
| | - Ali Hatef Salmanian
- Department of Agricultural Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran.
| |
Collapse
|
14
|
Rezvanirad A, Habibi M, Farokhi M, Asadi Karam MR. Immunogenic Potential and Therapeutic Efficacy of Multi-Epitope Encapsulated Silk Fibroin Nanoparticles against Pseudomonas aeruginosa-Mediated Urinary Tract Infections. Macromol Biosci 2023; 23:e2300074. [PMID: 37159936 DOI: 10.1002/mabi.202300074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 05/01/2023] [Indexed: 05/11/2023]
Abstract
Pseudomonas aeruginosa (P. aeruginosa) causing urinary tract infections (UTIs) are a major concern among hospital-acquired infections. The need for an effective vaccine that reduces the infections is imperative. This study aims to evaluate the efficacy of a multi-epitope vaccine encapsulated in silk fibroin nanoparticles (SFNPs) against P. aeruginosa-mediated UTIs. A multi-epitope is constructed from nine proteins of P. aeruginosa using immunoinformatic analysis, expressed, and purified in BL21 (DE3) cells. The encapsulation efficiency of the multi-epitope in SFNPs is 85% with a mean particle size of 130 nm and 24% of the encapsulated antigen is released after 35 days. The vaccine formulations adjuvanted with SFNPs or alum significantly improve systemic and mucosal humoral responses and the cytokine profile (IFN-γ, IL-4, and IL-17) in mice. Additionally, the longevity of the IgG response is maintained for at least 110 days in a steady state. In a bladder challenge, mice treated with the multi-epitope admixed with alum or encapsulated in SFNPs demonstrate significant protection of the bladder and kidneys against P. aeruginosa. This study highlights the promising therapeutic potential of a multi-epitope vaccine encapsulated in SFNPs or adjuvanted with alum against P. aeruginosa infections.
Collapse
Affiliation(s)
- Azam Rezvanirad
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Mehri Habibi
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Mehdi Farokhi
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | | |
Collapse
|
15
|
Al-Hawary SIS, Saleh EAM, Mamajanov NA, S Gilmanova N, Alsaab HO, Alghamdi A, Ansari SA, Alawady AHR, Alsaalamy AH, Ibrahim AJ. Breast cancer vaccines; A comprehensive and updated review. Pathol Res Pract 2023; 249:154735. [PMID: 37611432 DOI: 10.1016/j.prp.2023.154735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 07/30/2023] [Accepted: 08/02/2023] [Indexed: 08/25/2023]
Abstract
According to the International Agency for Research on Cancer, breast cancer is more common than lung cancer globally. By 2040, mortality from breast cancer will rise by 50% and 40%, respectively. Despite advances in chemotherapy, endocrine therapy, and HER2-targeted therapy, breast cancer metastases and recurrences remain challenging to treat. Cancer vaccines are an effective treatment option because they stimulate a long-lasting immune response that will eliminate tumor cells. In studies on the breast cancer vaccine, no appreciable advantages were discovered. A recent study claims that immune checkpoint inhibitors or anti-HER2 monoclonal antibodies may be used in vaccinations. This vaccination strengthens the immune system to fight off breast cancer cells. Clinical trials have been conducted on DNA, dendritic cells, and peptide-based breast cancer vaccines. Studies on the breast cancer vaccine have employed subcutaneous, intramuscular, and intradermal injections. Clinical studies have shown that these efforts have not been successful. Several factors might have slowed the development of a breast cancer vaccine. The complexity of the immune system makes it challenging to create cancer vaccines. Given the heterogeneity of breast cancer, there may be a need for different vaccination strategies. Despite these obstacles, research into breast cancer vaccines continues. Effective methods for creating vaccines include immune checkpoint inhibition and anti-HER2 monoclonal antibodies. Research is also being done on specialized tumor vaccinations.
Collapse
Affiliation(s)
| | - Ebraheem Abdu Musad Saleh
- Department of Chemistry, Prince Sattam Bin Abdulaziz University, College of Arts and Science, Wadi Al-Dawasir 11991, Saudi Arabia
| | - Nodirjon Akhmetovich Mamajanov
- Teaching Assistant, MD, Department of Public Health, Healthcare Management and Physical Culture, Tashkent State Dental Institute, Tashkent, Uzbekistan; Research scholar, Department of Scientific Affairs, Samarkand State Medical Institute, Samarkand, Uzbekistan
| | - Nataliya S Gilmanova
- Department of Prosthetic Dentistry, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia.
| | - Hashem O Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, Taif University, Taif 21944, Saudi Arabia
| | - Adel Alghamdi
- Preparatory Year Program, Batterjee Medical College, Jeddah 21442, Saudi Arabia
| | - Shakeel Ahmed Ansari
- Department of Biochemistry, General Medicine Practice Program, Batterjee Medical College, Jeddah 21442, Saudi Arabia
| | - Ahmed Hussien Radie Alawady
- College of technical engineering, the Islamic University, Najaf, Iraq; College of technical engineering, the Islamic University of Al Diwaniyah, Iraq; College of technical engineering, the Islamic University of Babylon, Iraq
| | - Ali Hashiem Alsaalamy
- College of technical engineering, Imam Ja'afar Al-Sadiq University, Al-Muthanna 66002, Iraq
| | | |
Collapse
|
16
|
Govea-Alonso DO, García-Soto MJ, Mendoza-Pérez ES, Farfán-Castro S, Fuente D, González-Ortega O, Rosales-Mendoza S. Assessing the Adjuvant Effect of Layered Double Hydroxides (LDH) on BALB/c Mice. MATERIALS (BASEL, SWITZERLAND) 2023; 16:5467. [PMID: 37570172 PMCID: PMC10419364 DOI: 10.3390/ma16155467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/23/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023]
Abstract
The discovery and validation of new adjuvants are critical areas for vaccinology. Mineral materials (e.g., alum microparticles) have been used for a long time as adjuvants in human vaccine formulations. Nonetheless, the use of nanosized materials is a promising approach to diversify the properties of adjuvants. Nanoclays are potential adjuvants proposed by some research groups. However, their adjuvant mechanisms and safety have not been fully elucidated. Herein, we aimed at expanding the knowledge on the potential adjuvanticity of layered double hydroxide (LDH) nanoparticles by reporting a detailed method for the synthesis and characterization of LDHs and the adsorption of a model antigen (bovine serum albumin, BSA). LDHs varying in diameter (from 56 to 88 nm) were obtained, and an in vitro evaluation revealed that the LDHs are not inherently toxic. BSA was passively adsorbed onto the LDHs, and the immunogenicity in mice of the conjugates obtained was compared to that of free BSA and BSA co-administered with alum (Alum-BSA). The LDH-BSA conjugates induced a higher humoral response that lasted for a longer period compared with that of free BSA and Alum-BSA, confirming that LDH exerts adjuvant effects. The 56 nm LDH particles were deemed as the more efficient carrier since they induced a higher and more balanced Th1/Th2 response than the 88 nm particles. This study is a contribution toward expanding the characterization and use of nanoclays in vaccinology and justifies further studies with pathogen-specific antigens.
Collapse
Affiliation(s)
- Dania O. Govea-Alonso
- Departamento de Biotecnológicas y Ambientales, Universidad Autónoma de Guadalajara, Zapopan 45129, Mexico;
| | - Mariano J. García-Soto
- Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, Lomas 2ª. Sección, San Luis Potosí 78210, Mexico; (M.J.G.-S.); (E.S.M.-P.); (S.F.-C.)
| | - Emilio Sebastián Mendoza-Pérez
- Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, Lomas 2ª. Sección, San Luis Potosí 78210, Mexico; (M.J.G.-S.); (E.S.M.-P.); (S.F.-C.)
| | - Susan Farfán-Castro
- Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, Lomas 2ª. Sección, San Luis Potosí 78210, Mexico; (M.J.G.-S.); (E.S.M.-P.); (S.F.-C.)
| | - Diana Fuente
- Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, Lomas 2ª. Sección, San Luis Potosí 78210, Mexico; (M.J.G.-S.); (E.S.M.-P.); (S.F.-C.)
| | - Omar González-Ortega
- Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, Lomas 2ª. Sección, San Luis Potosí 78210, Mexico; (M.J.G.-S.); (E.S.M.-P.); (S.F.-C.)
| | - Sergio Rosales-Mendoza
- Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, Lomas 2ª. Sección, San Luis Potosí 78210, Mexico; (M.J.G.-S.); (E.S.M.-P.); (S.F.-C.)
| |
Collapse
|
17
|
Stiefel J, Zimmer J, Schloßhauer JL, Vosen A, Kilz S, Balakin S. Just Keep Rolling?-An Encompassing Review towards Accelerated Vaccine Product Life Cycles. Vaccines (Basel) 2023; 11:1287. [PMID: 37631855 PMCID: PMC10459022 DOI: 10.3390/vaccines11081287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/20/2023] [Accepted: 07/24/2023] [Indexed: 08/27/2023] Open
Abstract
In light of the recent pandemic, several COVID-19 vaccines were developed, tested and approved in a very short time, a process that otherwise takes many years. Above all, these efforts have also unmistakably revealed the capacity limits and potential for improvement in vaccine production. This review aims to emphasize recent approaches for the targeted rapid adaptation and production of vaccines from an interdisciplinary, multifaceted perspective. Using research from the literature, stakeholder analysis and a value proposition canvas, we reviewed technological innovations on the pharmacological level, formulation, validation and resilient vaccine production to supply bottlenecks and logistic networks. We identified four main drivers to accelerate the vaccine product life cycle: computerized candidate screening, modular production, digitized quality management and a resilient business model with corresponding transparent supply chains. In summary, the results presented here can serve as a guide and implementation tool for flexible, scalable vaccine production to swiftly respond to pandemic situations in the future.
Collapse
Affiliation(s)
- Janis Stiefel
- Fraunhofer Institute for Microengineering and Microsystems IMM, Carl-Zeiss-Straße 18-20, 55129 Mainz, Germany
| | - Jan Zimmer
- Fraunhofer Institute for Microengineering and Microsystems IMM, Carl-Zeiss-Straße 18-20, 55129 Mainz, Germany
| | - Jeffrey L. Schloßhauer
- Fraunhofer Institute for Cell Therapy and Immunology, Branch Bioanalytics and Bioprocesses IZI-BB, Am Mühlenberg 13, 14476 Potsdam, Germany
| | - Agnes Vosen
- Fraunhofer Center for International Management and Knowledge Economy IMW, Neumarkt 20, 04109 Leipzig, Germany
| | - Sarah Kilz
- Fraunhofer Center for International Management and Knowledge Economy IMW, Neumarkt 20, 04109 Leipzig, Germany
| | - Sascha Balakin
- Fraunhofer Institute for Ceramic Technologies and Systems IKTS Material Diagnostics, Bio- and Nanotechnology, Maria-Reiche-Straße 2, 01109 Dresden, Germany
- Max Bergmann Center of Biomaterials (MBC), Technical University of Dresden, Budapester Strasse 27, 01069 Dresden, Germany
| |
Collapse
|
18
|
Shah SM, Joshi D, Chbib C, Roni MA, Uddin MN. The Autoinducer N-Octanoyl-L-Homoserine Lactone (C8-HSL) as a Potential Adjuvant in Vaccine Formulations. Pharmaceuticals (Basel) 2023; 16:ph16050713. [PMID: 37242496 DOI: 10.3390/ph16050713] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/24/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
Autoinducers AI-1 and AI-2 play an important role in bacterial quorum sensing (QS), a form of chemical communication between bacteria. The autoinducer N-octanoyl-L-Homoserinehomoserine lactone (C8-HSL) serves as a major inter- and intraspecies communicator or 'signal', mainly for Gram-negative bacteria. C8-HSL is proposed to have immunogenic properties. The aim of this project is to evaluate C8-HSL as a potential vaccine adjuvant. For this purpose, a microparticulate formulation was developed. The C8-HSL microparticles (MPs) were formulated by a water/oil/water (W/O/W) double-emulsion solvent evaporation method using PLGA (poly (lactic-co-glycolic acid)) polymer. We tested C8-HSL MPs with two spray-dried bovine serum albumin (BSA)-encapsulated bacterial antigens: colonization factor antigen I (CFA/I) from Escherichia coli (E. coli.) and the inactive protective antigen (PA) from Bacillus anthracis (B. anthracis). We formulated and tested C8-HSL MP to determine its immunogenicity potential and its ability to serve as an adjuvant with particulate vaccine formulations. An in vitro immunogenicity assessment was performed using Griess's assay, which indirectly measures the nitric oxide radical (NOˑ) released by dendritic cells (DCs). The C8-HSL MP adjuvant was compared with FDA-approved adjuvants to determine its immunogenicity potential. C8-HSL MP was combined with particulate vaccines for measles, Zika and the marketed influenza vaccine. The cytotoxicity study showed that MPs were non-cytotoxic toward DCs. Griess's assay showed a comparable release of NOˑ from DCs when exposed to CFA and PA bacterial antigens. Nitric oxide radical (NOˑ) release was significantly higher when C8-HSL MPs were combined with particulate vaccines for measles and Zika. C8-HSL MPs showed immunostimulatory potential when combined with the influenza vaccine. The results showed that C8-HSL MPs were as immunogenic as FDA-approved adjuvants such as alum, MF59, and CpG. This proof-of-concept study showed that C8-HSL MP displayed adjuvant potential when combined with several particulate vaccines, indicating that C8-HSL MPs can increase the immunogenicity of both bacterial and viral vaccines.
Collapse
Affiliation(s)
- Sarthak M Shah
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Devyani Joshi
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Christiane Chbib
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin University, Miami, FL 33169, USA
| | - Monzurul A Roni
- Department of Health Sciences Education and Pathology, University of Illinois College of Medicine, Peoria, IL 61605, USA
| | - Mohammad N Uddin
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| |
Collapse
|
19
|
Domínguez-Odio A, Delgado DLC. Global commercialization and research of veterinary vaccines against Pasteurella multocida: 2015-2022 technological surveillance. Vet World 2023; 16:946-956. [PMID: 37576757 PMCID: PMC10420726 DOI: 10.14202/vetworld.2023.946-956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 03/31/2023] [Indexed: 08/15/2023] Open
Abstract
Background and Aim Pasteurella multocida can infect a multitude of wild and domesticated animals, bacterial vaccines have become a crucial tool in combating antimicrobial resistance (AMR) in animal production. The study aimed to evaluate the current status and scientific trends related to veterinary vaccines against Pasteurella multocida during the 2015-2022 period. Material and Methods The characteristics of globally marketed vaccines were investigated based on the official websites of 22 pharmaceutical companies. VOSviewer® 1.6.18 was used to visualize networks of coauthorship and cooccurrence of keywords from papers published in English and available in Scopus. Results Current commercial vaccines are mostly inactivated (81.7%), adjuvanted in aluminum hydroxide (57.8%), and designed to immunize cattle (33.0%). Investigational vaccines prioritize the inclusion of attenuated strains, peptide fragments, recombinant proteins, DNA as antigens, aluminum compounds as adjuvants and poultry as the target species. Conclusion Despite advances in genetic engineering and biotechnology, there will be no changes in the commercial dominance of inactivated and aluminum hydroxide-adjuvanted vaccines in the short term (3-5 years). The future prospects for bacterial vaccines in animal production are promising, with advancements in vaccine formulation and genetic engineering, they have the potential to improve the sustainability of the industry. It is necessary to continue with the studies to improve the efficacy of the vaccines and their availability.
Collapse
Affiliation(s)
- Aníbal Domínguez-Odio
- Dirección de Ciencia e Innovación. Grupo Empresarial LABIOFAM. Avenida Independencia km 16½, Boyeros, La Habana, Cuba
| | - Daniel Leonardo Cala Delgado
- Animal Science Research Group, Universidad Cooperativa de Colombia, Sede Bucaramanga, Carrera 33 N°, 30ª-05 (4.162,49 km) 68000, Bucaramanga, Colombia
| |
Collapse
|