1
|
Adediran E, Arte T, Pasupuleti D, Vijayanand S, Singh R, Patel P, Gulani M, Ferguson A, Uddin M, Zughaier SM, D’Souza MJ. Delivery of PLGA-Loaded Influenza Vaccine Microparticles Using Dissolving Microneedles Induces a Robust Immune Response. Pharmaceutics 2025; 17:510. [PMID: 40284505 PMCID: PMC12030082 DOI: 10.3390/pharmaceutics17040510] [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: 03/11/2025] [Revised: 04/04/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Influenza virus is one of the major respiratory virus infections that is a global health concern. Although there are already approved vaccines, most are administered via the intramuscular route, which is usually painful, leading to vaccine hesitancy. To this end, exploring the non-invasive, transdermal vaccination route using dissolving microneedles would significantly improve vaccine compliance. Research on innovative vaccine delivery systems, such as antigen-loaded PLGA microparticles, has the potential to pave the way for a broader range of vaccine candidates. Methods: In this proof-of-concept study, a combination of the inactivated influenza A H1N1 virus and inactivated influenza A H3N2 virus were encapsulated in a biodegradable poly (lactic-co-glycolic acid) (PLGA) polymeric matrix within microparticles, which enhanced antigen presentation. The antigen PLGA microparticles were prepared separately using a double emulsion (w/o/w), lyophilized, and characterized. Next, the vaccine microparticles were assessed in vitro in dendritic cells (DC 2.4) for immunogenicity. To explore pain-free transdermal vaccination, the vaccine microparticles were loaded into dissolving microneedles and administered in mice (n = 5). Results: Our vaccination study demonstrated that the microneedle-based vaccine elicited strong humoral responses as demonstrated by high antigen-specific IgA, IgG, IgG1, and IgG2a antibodies in serum samples and IgA in lung supernatant. Further, the vaccine also elicited a strong cellular response as evidenced by high levels of CD4+ and CD8a+ T cells in lymphoid organs such as the lymph nodes and spleen. Conclusion: The delivery of influenza vaccine-loaded PLGA microparticles using microneedles would be beneficial to individuals experiencing needle-phobia, as well as the geriatric and pediatric population.
Collapse
Affiliation(s)
- Emmanuel Adediran
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA; (E.A.); (T.A.); (D.P.); (S.V.); (R.S.); (P.P.); (M.G.); (A.F.); (M.U.)
| | - Tanisha Arte
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA; (E.A.); (T.A.); (D.P.); (S.V.); (R.S.); (P.P.); (M.G.); (A.F.); (M.U.)
| | - Dedeepya Pasupuleti
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA; (E.A.); (T.A.); (D.P.); (S.V.); (R.S.); (P.P.); (M.G.); (A.F.); (M.U.)
| | - Sharon Vijayanand
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA; (E.A.); (T.A.); (D.P.); (S.V.); (R.S.); (P.P.); (M.G.); (A.F.); (M.U.)
| | - Revanth Singh
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA; (E.A.); (T.A.); (D.P.); (S.V.); (R.S.); (P.P.); (M.G.); (A.F.); (M.U.)
| | - Parth Patel
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA; (E.A.); (T.A.); (D.P.); (S.V.); (R.S.); (P.P.); (M.G.); (A.F.); (M.U.)
| | - Mahek Gulani
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA; (E.A.); (T.A.); (D.P.); (S.V.); (R.S.); (P.P.); (M.G.); (A.F.); (M.U.)
| | - Amarae Ferguson
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA; (E.A.); (T.A.); (D.P.); (S.V.); (R.S.); (P.P.); (M.G.); (A.F.); (M.U.)
| | - Mohammad Uddin
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA; (E.A.); (T.A.); (D.P.); (S.V.); (R.S.); (P.P.); (M.G.); (A.F.); (M.U.)
| | - Susu M. Zughaier
- College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Martin J. D’Souza
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA; (E.A.); (T.A.); (D.P.); (S.V.); (R.S.); (P.P.); (M.G.); (A.F.); (M.U.)
| |
Collapse
|
2
|
Zhang H, Song M, Zhuang S, Wang Z, Shi H, Song Z, Song C, Cen L. Development of α-Tocopherol Loaded PLGA Nanoparticles and Its Evaluation as a Novel Immune Adjuvant. Macromol Rapid Commun 2025; 46:e2400400. [PMID: 38981020 DOI: 10.1002/marc.202400400] [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: 05/30/2024] [Revised: 06/26/2024] [Indexed: 07/11/2024]
Abstract
With the continuous development of preventive and therapeutic vaccines, traditional adjuvants cannot provide sufficient immune efficacy and it is of high necessity to develop safe and effective novel nanoparticle-based vaccine adjuvants. α-Tocopherol (TOC) is commonly used in oil-emulsion adjuvant systems as an immune enhancer, yet its bioavailability is limited by poor water solubility. This study aims to develop TOC-loaded poly(lactic-co-glycolic acid) (PLGA) nanoparticles (TOC-PLGA NPs) to explore the potential of TOC-PLGA NPs as a novel nanoparticle-immune adjuvant. TOC-PLGA NPs are prepared by a nanoprecipitation method and their physicochemical properties are characterized. It is shown that TOC-PLGA NPs are 110.8 nm, polydispersity index value of 0.042, and Zeta potential of -13.26 mV. The encapsulation efficiency and drug loading of NPs are 82.57% and 11.80%, respectively, and the cumulative release after 35 days of in vitro testing reaches 47%. Furthermore, TOC-PLGA NPs demonstrate a superior promotion effect on RAW 264.7 cell proliferation compared to PLGA NPs, being well phagocytosed and also promoting antigen uptake by macrophages. TOC-PLGA NPs can strongly upregulate the expression of co-stimulatory surface molecules and the secretion of cytokines. In conclusion, TOC-PLGA NPs can be a novel vaccine adjuvant with excellent biocompatibility and significant immune-enhancing activity.
Collapse
Affiliation(s)
- Huan Zhang
- Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, Department of Product Engineering, School of Chemical Engineering, East China University of Science and Technology, No.130 Mei Long Road, Shanghai, 200237, China
| | - Meng Song
- Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, Department of Product Engineering, School of Chemical Engineering, East China University of Science and Technology, No.130 Mei Long Road, Shanghai, 200237, China
| | - Shiya Zhuang
- Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, Department of Product Engineering, School of Chemical Engineering, East China University of Science and Technology, No.130 Mei Long Road, Shanghai, 200237, China
| | - Zining Wang
- Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, Department of Product Engineering, School of Chemical Engineering, East China University of Science and Technology, No.130 Mei Long Road, Shanghai, 200237, China
| | - Hui Shi
- Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, Department of Product Engineering, School of Chemical Engineering, East China University of Science and Technology, No.130 Mei Long Road, Shanghai, 200237, China
| | - Zhuolang Song
- Shanghai Mingqi Energy Technology Co., Ltd, No. 29, Lane 155, Baocheng Road, Shanghai, 201199, China
| | - Chuanhe Song
- Shanghai Mingqi Energy Technology Co., Ltd, No. 29, Lane 155, Baocheng Road, Shanghai, 201199, China
| | - Lian Cen
- Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, Department of Product Engineering, School of Chemical Engineering, East China University of Science and Technology, No.130 Mei Long Road, Shanghai, 200237, China
| |
Collapse
|
3
|
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
|
4
|
Hu Q, Ren A, Zhang X, Tang Z, Wang R, Wang DY, Huang T, Liu J, Ming J. Manganese-Loaded pH-Responsive DNA Hydrogels Enable Tg-Guided Thyroid Tumor Targeted Magnetic Resonance Imaging. ACS APPLIED MATERIALS & INTERFACES 2025; 17:13403-13414. [PMID: 39996966 DOI: 10.1021/acsami.4c19676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/26/2025]
Abstract
The diagnosis of metastatic and recurrent occult thyroid cancer presents a significant challenge. This study introduces a DNA-Mn hydrogel (M-TDH) that specifically targets thyroglobulin (Tg). This nanogel is loaded with paramagnetic Mn2+ for facilitating magnetic resonance (MR) imaging. As a cofactor of DNA polymerase, Mn2+ promotes the extension of long-strand DNA and forms Mn2PPi nuclei with PPi4- in the system. The synthesis of M-TDH is achieved through Mn2PPi nucleation and growth with long-strand DNA acting as the structural framework. The X-scaffold functions as a junction point, thereby enhancing structural stability. The Tg aptamer sequence is incorporated into M-TDH, ensuring specific targeting of thyroid cancer cells. Furthermore, M-TDH demonstrates an extended residence time at the thyroid tumor site, thus increasing the duration of enhanced MR imaging. Overall, this study introduces an aptamer-based, thyroid tumor-targeted DNA nanogel for MR imaging diagnostic applications, with the potential to advance a multifunctional magnetic nanosystem toward clinical application.
Collapse
Affiliation(s)
- Qingyi Hu
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Anwen Ren
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Ximeng Zhang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Zimei Tang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Rong Wang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Dong-Yuan Wang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Tao Huang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Jie Liu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, 430022 Wuhan, China
- Hubei Key Laboratory of Molecular Imaging, 430022 Wuhan, China
| | - Jie Ming
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| |
Collapse
|
5
|
Yahyaei S, Abdoli A, Jamali A, Teimoori A, Arefian E, Eftekhari Z, Jamur P. Targeting Respiratory Viruses: The Efficacy of Intranasal mRNA Vaccination in Generating Protective Mucosal and Systemic Immunity Against Influenza A (H1N1). Influenza Other Respir Viruses 2025; 19:e70093. [PMID: 40127967 PMCID: PMC11932742 DOI: 10.1111/irv.70093] [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: 08/24/2024] [Revised: 02/20/2025] [Accepted: 03/10/2025] [Indexed: 03/26/2025] Open
Abstract
Four significant influenza outbreaks have occurred over the past 100 years, and the 1918 influenza pandemic is the most severe. Since influenza viruses undergo antigenic evolution, they are the pathogens most likely to trigger a new pandemic shortly. Intranasal vaccination offers a promising strategy for preventing diseases triggered by respiratory viruses by eliciting an immunoglobulin A (IgA) response, limiting virus replication and transmission from the respiratory tract more efficiently than intramuscular vaccines. Combining intranasal administration and mRNA-lipid nanoparticles can be an ideal strategy for limiting the extent of the next flu pandemic. This study explored the immunogenicity of intranasally delivered mRNA encapsulated in mannose-histidine-conjugated chitosan lipid nanoparticles (MHCS-LNPs) as a vaccine against influenza A (H1N1) in BALB/c mice. Intranasal administration of mRNA-MHCS-LNPs resulted in the generation of influenza A (H1N1) hemagglutinin-specific neutralizing antibodies in vaccinated animals. The enzyme-linked immunosorbent assay (ELISA) results indicated a notable increase in the quantity of immunoglobulin G (IgG) and IgA antibodies in serum and the bronchoalveolar lavage fluid (BALF), respectively, and exhibited influenza A-specific IFN-γ secretion in vaccinated mice, as well as a noticeable alteration in IL-5 production. Overall, this study demonstrated an effective immunogenic response against respiratory viral infections through intranasal delivery of an mRNA-MHCS-LNP vaccine.
Collapse
MESH Headings
- Animals
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A Virus, H1N1 Subtype/genetics
- Administration, Intranasal
- Mice, Inbred BALB C
- Influenza Vaccines/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/genetics
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Mice
- Orthomyxoviridae Infections/prevention & control
- Orthomyxoviridae Infections/immunology
- Female
- Nanoparticles/administration & dosage
- Immunity, Mucosal
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Immunoglobulin A/analysis
- Chitosan/administration & dosage
- Chitosan/chemistry
- Vaccination/methods
- Immunoglobulin G/blood
- RNA, Messenger/genetics
- RNA, Messenger/immunology
- mRNA Vaccines
- Bronchoalveolar Lavage Fluid/immunology
- Liposomes
Collapse
Affiliation(s)
- Sara Yahyaei
- Hepatitis and AIDS DepartmentPasteur Institute of IranTehranIran
- Student Research CommitteePasteur Institute of IranTehranIran
| | - Asghar Abdoli
- Hepatitis and AIDS DepartmentPasteur Institute of IranTehranIran
| | - Abbas Jamali
- Department of Influenza and Other Respiratory VirusesPasteur Institute of IranTehranIran
| | - Ali Teimoori
- Department of Virology, Faculty of MedicineHamadan University of Medical SciencesHamadanIran
| | - Ehsan Arefian
- Department of Microbiology, School of Biology, College of ScienceUniversity of TehranTehranIran
| | | | - Parisa Jamur
- Hepatitis and AIDS DepartmentPasteur Institute of IranTehranIran
| |
Collapse
|
6
|
Lim S, Chung HJ, Oh YJ, Hinterdorfer P, Myung SC, Seo Y, Ko K. Modification of Fc-fusion protein structures to enhance efficacy of cancer vaccine in plant expression system. PLANT BIOTECHNOLOGY JOURNAL 2025; 23:960-982. [PMID: 39724301 PMCID: PMC11869200 DOI: 10.1111/pbi.14552] [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: 10/29/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024]
Abstract
Epithelial cell adhesion molecule (EpCAM) fused to IgG, IgA and IgM Fc domains was expressed to create IgG, IgA and IgM-like structures as anti-cancer vaccines in Nicotiana tabacum. High-mannose glycan structures were generated by adding a C-terminal endoplasmic reticulum (ER) retention motif (KDEL) to the Fc domain (FcK) to produce EpCAM-Fc and EpCAM-FcK proteins in transgenic plants via Agrobacterium-mediated transformation. Cross-fertilization of EpCAM-Fc (FcK) transgenic plants with Joining chain (J-chain, J and JK) transgenic plants led to stable expression of large quaternary EpCAM-IgA Fc (EpCAM-A) and IgM-like (EpCAM-M) proteins. Immunoblotting, SDS-PAGE and ELISA analyses demonstrated that proteins with KDEL had higher expression levels and binding activity to anti-EpCAM IgGs. IgM showed the strongest binding among the fusion proteins, followed by IgA and IgG. Sera from BALB/c mice immunized with these vaccines produced anti-EpCAM IgGs. Flow cytometry indicated that the EpCAM-Fc fusion proteins significantly activated CD8+ cytotoxic T cells, CD4+ helper T cells and B cells, particularly with EpCAM-FcKP and EpCAM-FcP (FcKP) × JP (JKP). The induced anti-EpCAM IgGs captured human prostate cancer PC-3 and colorectal cancer SW620 cells. Sera from immunized mice inhibited cancer cell proliferation, migration and invasion; down-regulated proliferation markers (PCNA, Ki-67) and epithelial-mesenchymal transition markers (Vimentin); and up-regulated E-cadherin. These findings suggest that N. tabacum can produce effective vaccine candidates to induce anti-cancer immune responses.
Collapse
Affiliation(s)
- Sohee Lim
- BioSystems Design Lab, Department of Medicine, College of MedicineChung‐Ang UniversitySeoulKorea
| | - Hyun Joo Chung
- Department of Urology, College of MedicineChung‐Ang UniversitySeoulKorea
| | - Yoo Jin Oh
- Department of Applied Experimental BiophysicsJohannes Kepler UniversityLinzAustria
| | - Peter Hinterdorfer
- Department of Applied Experimental BiophysicsJohannes Kepler UniversityLinzAustria
| | - Soon Chul Myung
- Department of Urology, College of MedicineChung‐Ang UniversitySeoulKorea
| | - Young‐Jin Seo
- Department of Life ScienceChung‐Ang UniversitySeoulKorea
| | - Kisung Ko
- BioSystems Design Lab, Department of Medicine, College of MedicineChung‐Ang UniversitySeoulKorea
| |
Collapse
|
7
|
Suresh R, Olaitan Comfort S, Dolatyabi S, Schrock J, Singh M, Renukaradhya GJ. Evaluation of mucosal adjuvants to chitosan-nanoparticle-based oral subunit vaccine for controlling salmonellosis in broilers. Front Immunol 2025; 16:1509990. [PMID: 39981235 PMCID: PMC11840259 DOI: 10.3389/fimmu.2025.1509990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 01/06/2025] [Indexed: 02/22/2025] Open
Abstract
Salmonellosis, a gastrointestinal disease, continues to be one of the major public health concerns worldwide. Poultry meat and eggs are recognized as the major source of Salmonella food poisoning in humans. Our study evaluated the protective efficacy of mannose-conjugated chitosan-nanoparticle (mChitosan-NP)-based subunit vaccine, consisting of immunogenic outer membrane proteins and flagella of Salmonella Enteritidis [mChitosan (OMP+FLA)/FLA-NP], coadministered orally with potent mucosal adjuvants to reduce the colonization of S. Enteritidis in the intestines of broiler chickens. We evaluated the adjuvant effects of three different doses of two well-known mucosal adjuvants, c-di-GMP (stimulator of interferon gene agonist) and whole cell lysate (WCL) of Mycobacterium smegmatis, to improve the efficacy of mChitosan (OMP+FLA)/FLA-NP vaccine. Our data reaffirmed the potent adjuvanticity of both of these adjuvants and identified their optimal dose when entrapped in mChitosan-NP to potentiate the immunogenicity and efficacy of orally delivered mChitosan (OMP+FLA)/FLA-NP vaccine. The physical characteristics of mChitosan (OMP+FLA)/FLA-NP, mChitosan-GMP/FLA-NP, and mChitosan-WCL/FLA-NP formulations revealed a high positive charge (Zeta potential +20-25 mV), size 235-260 nm, and polydispersity index 0.35-0.52, which are conducive for oral delivery. The efficacy in chickens that received oral administration with a combination of the vaccine-adjuvant formulations was evaluated by challenging with Salmonella Enteritidis. Our data showed that mChitosan (OMP+FLA)/FLA-NP WCL at 10 µg/dose formulation consistently reduced the S. Enteritidis load by over 0.5 log10 comparable to a commercial live vaccine at post-challenge days 4 and 10. Immunologically, we observed enhanced systemic and mucosal antibody and cellular (B cells and T-helper cells) immune responses as well as upregulation of expression of immune cytokine genes IFN-γ, TGF-β, and IL-17 in the cecal tonsils of adjuvanted mChitosan-NP Salmonella-subunit-vaccinated birds. Overall, particularly the mucosal adjuvant WCL consistently enhanced the efficacy of mChitosan (OMP+FLA)/FLA-NP vaccine by inducing effective immune responses.
Collapse
Affiliation(s)
| | | | | | | | | | - Gourapura J. Renukaradhya
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agricultural, and Environmental Sciences, The Ohio State University, Wooster, OH, United States
| |
Collapse
|
8
|
Lei W, Zhou K, Lei Y, Li Q, Zhu H. Cancer vaccines: platforms and current progress. MOLECULAR BIOMEDICINE 2025; 6:3. [PMID: 39789208 PMCID: PMC11717780 DOI: 10.1186/s43556-024-00241-8] [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: 08/02/2024] [Revised: 12/03/2024] [Accepted: 12/05/2024] [Indexed: 01/12/2025] Open
Abstract
Cancer vaccines, crucial in the immunotherapeutic landscape, are bifurcated into preventive and therapeutic types, both integral to combating oncogenesis. Preventive cancer vaccines, like those against HPV and HBV, reduce the incidence of virus-associated cancers, while therapeutic cancer vaccines aim to activate dendritic cells and cytotoxic T lymphocytes for durable anti-tumor immunity. Recent advancements in vaccine platforms, such as synthetic peptides, mRNA, DNA, cellular, and nano-vaccines, have enhanced antigen presentation and immune activation. Despite the US Food and Drug Administration approval for several vaccines, the full therapeutic potential remains unrealized due to challenges such as antigen selection, tumor-mediated immunosuppression, and optimization of delivery systems. This review provides a comprehensive analysis of the aims and implications of preventive and therapeutic cancer vaccine, the innovative discovery of neoantigens enhancing vaccine specificity, and the latest strides in vaccine delivery platforms. It also critically evaluates the role of adjuvants in enhancing immunogenicity and mitigating the immunosuppressive tumor microenvironment. The review further examines the synergistic potential of combining cancer vaccines with other therapies, such as chemotherapy, radiotherapy, and immune checkpoint inhibitors, to improve therapeutic outcomes. Overcoming barriers such as effective antigen identification, immunosuppressive microenvironments, and adverse effects is critical for advancing vaccine development. By addressing these challenges, cancer vaccines can offer significant improvements in patient outcomes and broaden the scope of personalized cancer immunotherapy.
Collapse
Affiliation(s)
- Wanting Lei
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Kexun Zhou
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ye Lei
- College of Liberal Arts, Neijiang Normal University, Neijiang, 641100, Sichuan, China
| | - Qiu Li
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Hong Zhu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
9
|
de Almeida CMF, Rios WM, Duarte MPF, Brandão IT, Paiva NF, Vicentini FTMDC. Self-assembled nanovaccine based on apoferritin: Development and vaccine regimen evaluation. Eur J Pharm Biopharm 2025; 206:114589. [PMID: 39613269 DOI: 10.1016/j.ejpb.2024.114589] [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: 07/16/2024] [Revised: 09/18/2024] [Accepted: 11/26/2024] [Indexed: 12/01/2024]
Abstract
Apoferritin-based systems have been explored last decade for their potential as vaccine delivery for viral diseases. The nanosized properties of an apoferritin-based system could increase immunogenicity, contribute to antigen stability, and reduce the vaccines' adverse effects. The mutated extracellular portion of the epidermal growth factor receptor (EGFRvIII peptide, PEPvIII) can be applied as a specific tumoral antigen due to rare expression in normal cells. In this context, the present study proposed the development and the immunogenicity evaluation of an apoferritin-based system (AFt) to deliver a peptide vaccine for an antitumoral purpose. We developed a formulation with different PEPvIII:AFt ratios and during the association efficiency analysis, identified the dependence between the AFt concentration and the PEPvIII association percentage in the formulation. Besides, differences in the immune responses against EGFRvIII were observed depending on the PEPvIII concentration due to the different association efficiencies. Finally, the humoral immune response results showed a high antibody production against AFt, which might affect the immunological tolerance. Collectively, this study establishes the PEPvIII:AFt formulation process and highlights the determinant factors for guaranteeing vaccine safety and efficacy.
Collapse
Affiliation(s)
| | - Wendy Martin Rios
- Department of Biochemistry and Immunology, School of Medicine of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, SP, Brazil.
| | | | - Izaíra Tincani Brandão
- Department of Biochemistry and Immunology, School of Medicine of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, SP, Brazil.
| | - Natalia Floriano Paiva
- School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, SP, Brazil.
| | | |
Collapse
|
10
|
Zhang R, Li G, Wu Y, Wang X, Luan Q. Pathogenic mechanisms and potential applications of extracellular vesicles from periodontal pathogens in periodontitis. Front Immunol 2024; 15:1513983. [PMID: 39759521 PMCID: PMC11695242 DOI: 10.3389/fimmu.2024.1513983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 12/03/2024] [Indexed: 01/07/2025] Open
Abstract
Periodontitis is a multifactorial disease characterized by chronic destruction of the periodontal supporting tissues and is closely associated with the dysbiosis of the plaque biofilm. It is the leading cause of tooth loss in adults. Bacterial extracellular vesicles (BEVs) are released from bacteria, which range in size from 20 to 400 nm. These vesicles contain various components derived from their parent bacteria, including nucleic acids, proteins, lipids, and other molecules, which facilitate functions such as molecular transfer, metabolic regulation, bacterial interactions, biofilm formation, and immune modulation. BEVs participated in the pathophysiological process of periodontitis. Recently emerging evidence also showed that the contents of EVs in saliva and gingival crevicular fluid (miRNAs, mRNAs, and proteins) could be used as potential biomarkers for periodontitis. While most current research focuses on human-derived components, much less is known about BEVs. Therefore, this review introduces the formation mechanisms and components of BEVs related to periodontitis. Then, this review summarizes the current information about the mechanism, the diagnostic and theraputic value of periodontal pathogen-derived extracellular vesicles in the development of periodontitis. Furthermore, the future challenges of exploring the role of BEVs in periodontitis are also discussed.
Collapse
Affiliation(s)
- Ruiqing Zhang
- Department of Periodontology, Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao, Shandong, China
- Department of Periodontology, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Guoliang Li
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yingtao Wu
- Department of Periodontology, Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao, Shandong, China
| | - Xiaoxuan Wang
- Department of Periodontology, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Qingxian Luan
- Department of Periodontology, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, China
| |
Collapse
|
11
|
Gu P, Zhu Y, Xu P, Zhao Q, Zhao X, Zhao K, Wang X, Zhang W, Bao Y, Shi W. Poria cocos polysaccharide-loaded Alum Pickering emulsion as vaccine adjuvant to enhance immune responses. Colloids Surf B Biointerfaces 2024; 244:114144. [PMID: 39116600 DOI: 10.1016/j.colsurfb.2024.114144] [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: 06/15/2024] [Revised: 08/02/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024]
Abstract
Traditional Alum adjuvants mainly elicit a Th2 humoral immune response, but fail to generate a robust Th1 cellular immune response. However, the cellular immune response is essential for vaccination against cancer and a number of chronic infectious diseases, including human immunodeficiency virus infection and tuberculosis. In our previous study, we demonstrated that the polysaccharide from Poria cocos (PCP) has the potential to serve as an immunologic stimulant, enhancing both humoral and cellular immune responses. However, this effect was only observed at high concentrations. In this study, to enhance the immune-stimulation effect of PCP and modify the type of immune response elicited by Alum adjuvant, we successfully developed a Pickering emulsion delivery system (PCP-Al-Pickering) using PCP-loaded Alhydrogel particles as the stabilizer. After optimization, the Pickering emulsion exhibited excellent storage capacity and effectively adsorbed the PCP and antigen. As an adjuvant delivery system, the PCP-Al-Pickering emulsion facilitated the antigen uptake by macrophages, increased the recruitment of cells at injection sites, improved the activation of dendritic cells in draining lymph nodes, elicited a potent and durable antibody response, and promoted the activation of CD4+ and CD8+ T cells. Importantly, the PCP-Al-Pickering emulsion adjuvant elicited a balanced Th1 and Th2 immune response, in comparison to Alum adjuvant. The PCP-Al-Pickering emulsion may serve as a safe and promising adjuvant delivery system to enhance immune responses.
Collapse
Affiliation(s)
- Pengfei Gu
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Yixuan Zhu
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Panpan Xu
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Qi Zhao
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Xinghua Zhao
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Kuan Zhao
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Xiao Wang
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Wuchao Zhang
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Yongzhan Bao
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Wanyu Shi
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China.
| |
Collapse
|
12
|
Yang M, Zhou J, Lu L, Deng D, Huang J, Tang Z, Shi X, Lo P, Lovell JF, Zheng Y, Jin H. Tumor cell membrane-based vaccines: A potential boost for cancer immunotherapy. EXPLORATION (BEIJING, CHINA) 2024; 4:20230171. [PMID: 39713208 PMCID: PMC11655317 DOI: 10.1002/exp.20230171] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 03/08/2024] [Indexed: 12/24/2024]
Abstract
Because therapeutic cancer vaccines can, in theory, eliminate tumor cells specifically with relatively low toxicity, they have long been considered for application in repressing cancer progression. Traditional cancer vaccines containing a single or a few discrete tumor epitopes have failed in the clinic, possibly due to challenges in epitope selection, target downregulation, cancer cell heterogeneity, tumor microenvironment immunosuppression, or a lack of vaccine immunogenicity. Whole cancer cell or cancer membrane vaccines, which provide a rich source of antigens, are emerging as viable alternatives. Autologous and allogenic cellular cancer vaccines have been evaluated as clinical treatments. Tumor cell membranes (TCMs) are an intriguing antigen source, as they provide membrane-accessible targets and, at the same time, serve as integrated carriers of vaccine adjuvants and other therapeutic agents. This review provides a summary of the properties and technologies for TCM cancer vaccines. Characteristics, categories, mechanisms, and preparation methods are discussed, as are the demonstrable additional benefits derived from combining TCM vaccines with chemotherapy, sonodynamic therapy, phototherapy, and oncolytic viruses. Further research in chemistry, biomedicine, cancer immunology, and bioinformatics to address current drawbacks could facilitate the clinical adoption of TCM vaccines.
Collapse
Affiliation(s)
- Muyang Yang
- College of Biomedicine and Health and College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
| | - Jie Zhou
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory HealthThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Liseng Lu
- College of Biomedicine and Health and College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
| | - Deqiang Deng
- College of Biomedicine and Health and College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
| | - Jing Huang
- College of Biomedicine and Health and College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
| | - Zijian Tang
- College of Biomedicine and Health and College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
| | - Xiujuan Shi
- College of Biomedicine and Health and College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
| | - Pui‐Chi Lo
- Department of Biomedical SciencesCity University of Hong KongKowloonHong KongChina
| | - Jonathan F. Lovell
- Department of Biomedical EngineeringUniversity at BuffaloState University of New YorkBuffaloNew YorkUSA
| | - Yongfa Zheng
- Department of OncologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Honglin Jin
- College of Biomedicine and Health and College of Life Science and TechnologyHuazhong Agricultural UniversityWuhanChina
| |
Collapse
|
13
|
He J, Ding X, Zhao J, Zeng J, Zhou Y, Xiao W, Hua D, Liu M, Guo H, Zhang Y, Qiu M, Li J. A novel pan-epitope based nanovaccine self-assembled with CpG enhances immune responses against flavivirus. J Nanobiotechnology 2024; 22:738. [PMID: 39609873 PMCID: PMC11603839 DOI: 10.1186/s12951-024-03031-0] [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: 08/14/2024] [Accepted: 11/21/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND Flavivirus is a highly prevalent and outbreak-prone disease, affecting billions of individuals annually and posing substantial public health challenges. Vaccination is critical to reducing the global impact of flavivirus infections, making the development of a safe and effective vaccine a top priority. The self-assembled pan-epitope vaccine presents key advantages for improving immunogenicity and safety without relying on external vectors or adding immunomodulatory elements, both of which are essential for successful vaccine development. RESULTS In this study, the pan-epitope peptide TBT was combined with adjuvant CpG to form the TBT-CpG nanovaccine (TBT-CpG NaVs), which was found to be spherical, uniform in shape, and demonstrated strong serum stability. In vitro studies showed that the TBT-CpG NaVs were efficiently taken up and internalized by bone marrow-derived dendritic cells (BMDCs). Flow cytometry and transcriptomic analysis indicated that the antigens were effectively presented to antigen-presenting cells (APCs) via the MHC II pathway, which facilitated BMDCs maturation and promoted the release of pro-inflammatory cytokines IL-1β, TNF-α, and IL-6. In vivo studies confirmed that TBT-CpG NaVs enhanced antigen-specific IgG levels, significantly increased IFN-γ and IL-4 expression in spleen cells, and offered protective effects against Dengue virus (DENV) and Zika virus (ZIKV) infections. Safety evaluations revealed no hepatotoxicity and no significant organ damage in immunized mice. CONCLUSION The self-assembled candidate nanovaccine TBT-CpG NaVs effectively activates BMDCs and triggers a targeted immune response, providing antiviral effects against DENV and ZIKV. This vaccine demonstrates good immunogenicity and safety, establishing a promising foundation and a new strategy for the development of safe and effective vaccines.
Collapse
Affiliation(s)
- Jiuxiang He
- College of Basic Medicine, Army Medical University, Gaotanyan str. 30, Chongqing, 400038, China
| | - Xiaoyan Ding
- College of Basic Medicine, Army Medical University, Gaotanyan str. 30, Chongqing, 400038, China
- Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig Maximilians University, Munich, Germany
| | - Jing Zhao
- College of Basic Medicine, Army Medical University, Gaotanyan str. 30, Chongqing, 400038, China
| | - Jie Zeng
- College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Yuxin Zhou
- College of Basic Medicine, Army Medical University, Gaotanyan str. 30, Chongqing, 400038, China
| | - Wen Xiao
- College of Basic Medicine, Army Medical University, Gaotanyan str. 30, Chongqing, 400038, China
| | - Dong Hua
- College of Basic Medicine, Army Medical University, Gaotanyan str. 30, Chongqing, 400038, China
| | - Minchi Liu
- College of Basic Medicine, Army Medical University, Gaotanyan str. 30, Chongqing, 400038, China
| | - Hongxia Guo
- College of Basic Medicine, Army Medical University, Gaotanyan str. 30, Chongqing, 400038, China
| | - Yu Zhang
- College of Basic Medicine, Army Medical University, Gaotanyan str. 30, Chongqing, 400038, China
| | - Minyue Qiu
- College of Basic Medicine, Army Medical University, Gaotanyan str. 30, Chongqing, 400038, China
| | - Jintao Li
- College of Basic Medicine, Army Medical University, Gaotanyan str. 30, Chongqing, 400038, China.
| |
Collapse
|
14
|
Chulanetra M, Punnakitikashem P, Mahasongkram K, Chaicumpa W, Glab-Ampai K. Immunogenicity of intraperitoneal and intranasal liposome adjuvanted VLP vaccines against SARS-CoV-2 infection. Sci Rep 2024; 14:27311. [PMID: 39516286 PMCID: PMC11549293 DOI: 10.1038/s41598-024-79122-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024] Open
Abstract
Humans get SARS-CoV-2 infection mainly through inhalation; thus, vaccine that induces protective immunity at the virus entry site is important for early control of the infection. In this study, two anionic liposome (L)-adjuvanted VLP vaccines against SARS-CoV-2 were formulated. Baculovirus-Sf21 insect cell system was used for production of VLPs made of full-length S, M and E proteins. S protein of one vaccine (L-SME-VLPs) contained furin cleavage site at the S1/S2 junction, while that of another vaccine (L-S'ME-VLPs) did not. Both vaccines were innocuous and immunogenic when administered IP and IN to mice. Mice immunized IP with L-SME-VLPs/L-S'ME-VLPs (three doses, two-weeks intervals) had serum virus neutralizing (VN) antibodies (in falling order of isotype frequency): IgG3, IgA and IgG2a/IgG3, IgA and IgM, respectively. The L-S'ME VLPs vaccine induced significantly higher serum VN antibody titers than the L-SME-VLPs vaccine. All mice immunized IN with both vaccines had significant rise of VN antibodies in their bronchoalveolar lavage fluids (BALF). The VN antibodies in 67% of immunized mice were Th1- isotypes (IgG2a and/or IgG2b); the immunized mice had also other antibody isotypes in BALF. The intranasal L-S'ME-VLPs should be tested further step-by-step towards the clinical use as effective and safe vaccine against SARS-CoV-2.
Collapse
MESH Headings
- Animals
- Liposomes
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- SARS-CoV-2/immunology
- Administration, Intranasal
- Mice
- COVID-19/prevention & control
- COVID-19/immunology
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/blood
- Female
- Mice, Inbred BALB C
- Vaccines, Virus-Like Particle/immunology
- Vaccines, Virus-Like Particle/administration & dosage
- Immunogenicity, Vaccine
- Injections, Intraperitoneal
- Humans
- Adjuvants, Immunologic/administration & dosage
- Spike Glycoprotein, Coronavirus/immunology
- Adjuvants, Vaccine/administration & dosage
Collapse
Affiliation(s)
- Monrat Chulanetra
- Department of Parasitology, Center of Research Excellence in Therapeutic Proteins and Antibody Engineering, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Primana Punnakitikashem
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Kodchakorn Mahasongkram
- Department of Parasitology, Center of Research Excellence in Therapeutic Proteins and Antibody Engineering, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Wanpen Chaicumpa
- Department of Parasitology, Center of Research Excellence in Therapeutic Proteins and Antibody Engineering, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Kantaphon Glab-Ampai
- Department of Parasitology, Center of Research Excellence in Therapeutic Proteins and Antibody Engineering, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.
| |
Collapse
|
15
|
Berger S, Zeyn Y, Wagner E, Bros M. New insights for the development of efficient DNA vaccines. Microb Biotechnol 2024; 17:e70053. [PMID: 39545748 PMCID: PMC11565620 DOI: 10.1111/1751-7915.70053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 10/29/2024] [Indexed: 11/17/2024] Open
Abstract
Despite the great potential of DNA vaccines for a broad range of applications, ranging from prevention of infections, over treatment of autoimmune and allergic diseases to cancer immunotherapies, the implementation of such therapies for clinical treatment is far behind the expectations up to now. The main reason is the poor immunogenicity of DNA vaccines in humans. Consequently, the improvement of the performance of DNA vaccines in vivo is required. This mini-review provides an overview of the current state of DNA vaccines and the various strategies to enhance the immunogenic potential of DNA vaccines, including (i) the optimization of the DNA construct itself regarding size, nuclear transfer and transcriptional regulation; (ii) the use of appropriate adjuvants; and (iii) improved delivery, for example, by careful choice of the administration route, physical methods such as electroporation and nanomaterials that may allow cell type-specific targeting. Moreover, combining nanoformulated DNA vaccines with other immunotherapies and prime-boost strategies may help to enhance success of treatment.
Collapse
Affiliation(s)
- Simone Berger
- Pharmaceutical Biotechnology, Department of Pharmacy, Center for NanoScienceLudwig‐Maximilians‐Universität (LMU) MunichMunichGermany
| | - Yanira Zeyn
- Department of DermatologyUniversity Medical Center of the Johannes Gutenberg University (JGU) MainzMainzGermany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, Center for NanoScienceLudwig‐Maximilians‐Universität (LMU) MunichMunichGermany
| | - Matthias Bros
- Department of DermatologyUniversity Medical Center of the Johannes Gutenberg University (JGU) MainzMainzGermany
| |
Collapse
|
16
|
Rodrigues Rodrigues R, Alves MLF, Bilhalva MA, Kremer FS, Junior CM, Ferreira MRA, Galvão CC, Quatrin PHDN, Conceição FR. Large Clostridial Toxins: A Brief Review and Insights into Antigen Design for Veterinary Vaccine Development. Mol Biotechnol 2024:10.1007/s12033-024-01303-6. [PMID: 39472390 DOI: 10.1007/s12033-024-01303-6] [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: 08/12/2024] [Accepted: 10/08/2024] [Indexed: 12/18/2024]
Abstract
The group of large clostridial toxins (LCTs) includes toxins A (TcdA) and B (TcdB) from Clostridioides difficile, hemorrhagic and lethal toxins from Paeniclostridium sordellii, alpha toxin from Clostridium novyi (TcnA), and cytotoxin from Clostridium perfringens. These toxins are associated with severe pathologies in livestock, including gas gangrene (P. sordellii and C. novyi), infectious necrotic hepatitis (C. novyi), avian necrotic enteritis (C. perfringens), and enterocolitis (C. difficile). Immunoprophylaxis is crucial for controlling these diseases, but traditional vaccines face production challenges, such as labor-intensive processes, and often exhibit low immunogenicity. This has led to increased interest in recombinant vaccines. While TcdA and TcdB are well-studied for human immunization, other LCTs remain poorly characterized and require further investigation. Therefore, this study emphasizes the importance of understanding lesser-explored toxins and proposes using immunoinformatics to identify their immunodominant regions. By mapping these regions using silico tools and considering their homology with TcdA and TcdB, the study aims to guide future research in veterinary vaccinology. It also explores alternatives to overcome the limitations of conventional and recombinant vaccines, offering guidelines for developing more effective vaccination strategies against severe infections in animals.
Collapse
Affiliation(s)
- Rafael Rodrigues Rodrigues
- Centro de Desenvolvimento Tecnológico, Biotecnologia, Universidade Federal de Pelotas - Campus Universitário, Capão do Leão, Rio Grande Do Sul, CEP 96160-000, Brazil.
| | - Mariliana Luiza Ferreira Alves
- Centro de Desenvolvimento Tecnológico, Biotecnologia, Universidade Federal de Pelotas - Campus Universitário, Capão do Leão, Rio Grande Do Sul, CEP 96160-000, Brazil
- Instituto Federal Sul-Rio-Grandense, IFSul, Campus Pelotas, Pelotas, Rio Grande Do Sul, Brasil
| | - Miguel Andrade Bilhalva
- Centro de Desenvolvimento Tecnológico, Biotecnologia, Universidade Federal de Pelotas - Campus Universitário, Capão do Leão, Rio Grande Do Sul, CEP 96160-000, Brazil
| | - Frederico Schmitt Kremer
- Centro de Desenvolvimento Tecnológico, Biotecnologia, Universidade Federal de Pelotas - Campus Universitário, Capão do Leão, Rio Grande Do Sul, CEP 96160-000, Brazil
| | - Clóvis Moreira Junior
- Centro de Desenvolvimento Tecnológico, Biotecnologia, Universidade Federal de Pelotas - Campus Universitário, Capão do Leão, Rio Grande Do Sul, CEP 96160-000, Brazil
| | - Marcos Roberto Alves Ferreira
- Centro de Desenvolvimento Tecnológico, Biotecnologia, Universidade Federal de Pelotas - Campus Universitário, Capão do Leão, Rio Grande Do Sul, CEP 96160-000, Brazil
| | - Cleideanny Cancela Galvão
- Centro de Desenvolvimento Tecnológico, Biotecnologia, Universidade Federal de Pelotas - Campus Universitário, Capão do Leão, Rio Grande Do Sul, CEP 96160-000, Brazil
| | - Pedro Henrique Dala Nora Quatrin
- Centro de Desenvolvimento Tecnológico, Biotecnologia, Universidade Federal de Pelotas - Campus Universitário, Capão do Leão, Rio Grande Do Sul, CEP 96160-000, Brazil
| | - Fabricio Rochedo Conceição
- Centro de Desenvolvimento Tecnológico, Biotecnologia, Universidade Federal de Pelotas - Campus Universitário, Capão do Leão, Rio Grande Do Sul, CEP 96160-000, Brazil
| |
Collapse
|
17
|
Carmona-Ribeiro AM, Pérez-Betancourt Y. Emerging Cationic Nanovaccines. Pharmaceutics 2024; 16:1362. [PMID: 39598488 PMCID: PMC11597065 DOI: 10.3390/pharmaceutics16111362] [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: 09/26/2024] [Revised: 10/15/2024] [Accepted: 10/22/2024] [Indexed: 11/29/2024] Open
Abstract
Cationic vaccines of nanometric sizes can directly perform the delivery of antigen(s) and immunomodulator(s) to dendritic cells in the lymph nodes. The positively charged nanovaccines are taken up by antigen-presenting cells (APCs) of the lymphatic system often originating the cellular immunological defense required to fight intracellular microbial infections and the proliferation of cancers. Cationic molecules imparting the positive charges to nanovaccines exhibit a dose-dependent toxicity which needs to be systematically addressed. Against the coronavirus, mRNA cationic nanovaccines evolved rapidly. Nowadays cationic nanovaccines have been formulated against several infections with the advantage of cationic compounds granting protection of nucleic acids in vivo against biodegradation by nucleases. Up to the threshold concentration of cationic molecules for nanovaccine delivery, cationic nanovaccines perform well eliciting the desired Th 1 improved immune response in the absence of cytotoxicity. A second strategy in the literature involves dilution of cationic components in biocompatible polymeric matrixes. Polymeric nanoparticles incorporating cationic molecules at reduced concentrations for the cationic component often result in an absence of toxic effects. The progress in vaccinology against cancer involves in situ designs for cationic nanovaccines. The lysis of transformed cancer cells releases several tumoral antigens, which in the presence of cationic nanoadjuvants can be systemically presented for the prevention of metastatic cancer. In addition, these local cationic nanovaccines allow immunotherapeutic tumor treatment.
Collapse
Affiliation(s)
- Ana Maria Carmona-Ribeiro
- Biocolloids Laboratory, Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo 05508-000, Brazil
| | - Yunys Pérez-Betancourt
- Department of Microbiology, University of Chicago, Cummings Life Science Center 920 E 58th St., Chicago, IL 60637, USA;
| |
Collapse
|
18
|
Henderson E, Wilson K, Huynh G, Plebanski M, Corrie S. Bionano Interactions of Organosilica Nanoparticles with Myeloid Derived Immune Cells. ACS APPLIED MATERIALS & INTERFACES 2024; 16:43329-43340. [PMID: 39109853 DOI: 10.1021/acsami.4c08415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
Investigating the interactions between nanomaterials and the cells they are likely to encounter in vivo is a critical aspect of designing nanomedicines for imaging and therapeutic applications. Immune cells such as dendritic cells, macrophages, and myeloid derived suppressor cells have a frontline role in the identification and removal of foreign materials from the body, with interactions shown to be heavily dependent on variables such as nanoparticle size, charge, and surface chemistry. Interactions such as cellular association or uptake of nanoparticles can lead to diminished functionality or rapid clearance from the body, making it critical to consider these interactions when designing and synthesizing nanomaterials for biomedical applications ranging from drug delivery to imaging and biosensing. We investigated the interactions between PEGylated organosilica nanoparticles and naturally endocytic immune cells grown from stem cells in murine bone marrow. Specifically, we varied the particle size from 60 nm up to 1000 nm and investigated the effects of size on immune cell association, activation, and maturation with these critical gatekeeper cells. These results will help inform future design parameters for in vitro and in vivo biomedical applications utilizing organosilica nanoparticles.
Collapse
Affiliation(s)
- Edward Henderson
- Department of Chemical and Biological Engineering, Monash University, Clayton, Melbourne, Victoria 3800, Australia
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Melbourne, Victoria 3083, Australia
| | - Kirsty Wilson
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Melbourne, Victoria 3083, Australia
| | - Gabriel Huynh
- Department of Chemical and Biological Engineering, Monash University, Clayton, Melbourne, Victoria 3800, Australia
| | - Magdalena Plebanski
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Melbourne, Victoria 3083, Australia
| | - Simon Corrie
- Department of Chemical and Biological Engineering, Monash University, Clayton, Melbourne, Victoria 3800, Australia
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Melbourne, Victoria 3083, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, Melbourne, Victoria 3800, Australia
| |
Collapse
|
19
|
Vega E, Burgos JM, Souto EB, García ML, Pujol M, Sánchez-López E. Biodegradable nanoplatforms for antigen delivery: part I - state of the art review of polymeric nanoparticles for cancer immunotherapy. Expert Opin Drug Deliv 2024; 21:1251-1262. [PMID: 39245953 DOI: 10.1080/17425247.2024.2400293] [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: 06/20/2024] [Accepted: 08/30/2024] [Indexed: 09/10/2024]
Abstract
INTRODUCTION Polymeric nanoparticles used for antigen delivery against infections and for cancer immunotherapy are an emerging therapeutic strategy in promoting the development of innovative vaccines. Beyond their capability to create targeted delivery systems with controlled release of payloads, biodegradable polymers are utilized for their ability to enhance the immunogenicity and stability of antigens. AREAS COVERED This review extensively discusses the physicochemical parameters that affect the behavior of nanoparticles as antigen-delivery systems. Additionally, various types of natural and synthetic polymers and recent advancements in nanoparticle-based targeted vaccine production are reviewed. EXPERT OPINION Biodegradable polymeric nanoparticles have gained major interest in the vaccination filed and have been extensively used to encapsulate antigens against a wide variety of tumors. Moreover, their versatility in terms of tunning their physicochemical characteristics, and their surface, facilitates the targeting to antigen presenting cells and enhances immune response.
Collapse
Affiliation(s)
- Estefanía Vega
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| | - Jordi Madariaga Burgos
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
| | - Eliana B Souto
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- UCD School of Chemical and Bioprocess Engineering, University College Dublin, Belfield, Ireland
| | - María Luisa García
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| | - Montserrat Pujol
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| | - Elena Sánchez-López
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| |
Collapse
|
20
|
Prosper P, Rodríguez Puertas R, Guérin DMA, Branda MM. Computational method for designing vaccines applied to virus-like particles (VLPs) as epitope carriers. Vaccine 2024; 42:3916-3929. [PMID: 38782665 DOI: 10.1016/j.vaccine.2024.05.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 04/06/2024] [Accepted: 05/04/2024] [Indexed: 05/25/2024]
Abstract
Nonenveloped virus-like particles (VLPs) are self-assembled oligomeric structures composed of one or more proteins that originate from diverse viruses. Because these VLPs have similar antigenicity to the parental virus, they are successfully used as vaccines against cognate virus infection. Furthermore, after foreign antigenic sequences are inserted in their protein components (chimVLPs), some VLPs are also amenable to producing vaccines against pathogens other than the virus it originates from (these VLPs are named platform or epitope carrier). Designing chimVLP vaccines is challenging because the immunogenic response must be oriented against a given antigen without altering stimulant properties inherent to the VLP. An important step in this process is choosing the location of the sequence modifications because this must be performed without compromising the assembly and stability of the original VLP. Currently, many immunogenic data and computational tools can help guide the design of chimVLPs, thus reducing experimental costs and work. In this study, we analyze the structure of a novel VLP that originate from an insect virus and describe the putative regions of its three structural proteins amenable to insertion. For this purpose, we employed molecular dynamics (MD) simulations to assess chimVLP stability by comparing mutated and wild-type (WT) VLP protein trajectories. We applied this procedure to design a chimVLP that can serve as a prophylactic vaccine against the SARS-CoV-2 virus. The methodology described in this work is generally applicable for VLP-based vaccine development.
Collapse
Affiliation(s)
- Pascalita Prosper
- Instituto de Física Aplicada - INFAP, Universidad Nacional de San Luis/CONICET, Argentina, Av. Ejército de los Andes 950, 5700 San Luis, San Luis, Argentina
| | - Rafael Rodríguez Puertas
- Universidad del País Vasco (UPV/EHU), Dept. Farmacología, Facultad de Medicina, B° Sarriena S/N, 48940 Leioa, Vizcaya, Spain; Neurodegenerative Diseases, BioCruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Diego M A Guérin
- Universidad del País Vasco (UPV/EHU) and Instituto Biofisika (CSIC, UPV/EHU), B° Sarriena S/N, 48940 Leioa, Vizcaya, Spain
| | - María Marta Branda
- Instituto de Física Aplicada - INFAP, Universidad Nacional de San Luis/CONICET, Argentina, Av. Ejército de los Andes 950, 5700 San Luis, San Luis, Argentina.
| |
Collapse
|
21
|
Ma B, Tao M, Li Z, Zheng Q, Wu H, Chen P. Mucosal vaccines for viral diseases: Status and prospects. Virology 2024; 593:110026. [PMID: 38373360 DOI: 10.1016/j.virol.2024.110026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 02/08/2024] [Accepted: 02/12/2024] [Indexed: 02/21/2024]
Abstract
Virus-associated infectious diseases are highly detrimental to human health and animal husbandry. Among all countermeasures against infectious diseases, prophylactic vaccines, which developed through traditional or novel approaches, offer potential benefits. More recently, mucosal vaccines attract attention for their extraordinary characteristics compared to conventional parenteral vaccines, particularly for mucosal-related pathogens. Representatively, coronavirus disease 2019 (COVID-19), a respiratory disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), further accelerated the research and development efforts for mucosal vaccines by thoroughly investigating existing strategies or involving novel techniques. While several vaccine candidates achieved positive progresses, thus far, part of the current COVID-19 mucosal vaccines have shown poor performance, which underline the need for next-generation mucosal vaccines and corresponding platforms. In this review, we summarized the typical mucosal vaccines approved for humans or animals and sought to elucidate the underlying mechanisms of these successful cases. In addition, mucosal vaccines against COVID-19 that are in human clinical trials were reviewed in detail since this public health event mobilized all advanced technologies for possible solutions. Finally, the gaps in developing mucosal vaccines, potential solutions and prospects were discussed. Overall, rational application of mucosal vaccines would facilitate the establishing of mucosal immunity and block the transmission of viral diseases.
Collapse
Affiliation(s)
- Bingjie Ma
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China
| | - Mengxiao Tao
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China
| | - Zhili Li
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China
| | - Quanfang Zheng
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China
| | - Haigang Wu
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China
| | - Peirong Chen
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China.
| |
Collapse
|
22
|
Riccardi D, Baldino L, Reverchon E. Liposomes, transfersomes and niosomes: production methods and their applications in the vaccinal field. J Transl Med 2024; 22:339. [PMID: 38594760 PMCID: PMC11003085 DOI: 10.1186/s12967-024-05160-4] [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: 12/30/2023] [Accepted: 04/03/2024] [Indexed: 04/11/2024] Open
Abstract
One of the most effective strategies to fight viruses and handle health diseases is vaccination. Recent studies and current applications are moving on antigen, DNA and RNA-based vaccines to overcome the limitations related to the conventional vaccination strategies, such as low safety, necessity of multiple injection, and side effects. However, due to the instability of pristine antigen, RNA and DNA molecules, the use of nanocarriers is required. Among the different nanocarriers proposed for vaccinal applications, three types of nanovesicles were selected and analysed in this review: liposomes, transfersomes and niosomes. PubMed, Scopus and Google Scholar databases were used for searching recent papers on the most frequently used conventional and innovative methods of production of these nanovesicles. Weaknesses and limitations of conventional methods (i.e., multiple post-processing, solvent residue, batch-mode processes) can be overcome using innovative methods, in particular, the ones assisted by supercritical carbon dioxide. SuperSomes process emerged as a promising production technique of solvent-free nanovesicles, since it can be easily scaled-up, works in continuous-mode, and does not require further post-processing steps to obtain the desired products. As a result of the literature analysis, supercritical carbon dioxide assisted methods attracted a lot of interest for nanovesicles production in the vaccinal field. However, despite their numerous advantages, supercritical processes require further studies for the production of liposomes, transfersomes and niosomes with the aim of reaching well-defined technologies suitable for industrial applications and mass production of vaccines.
Collapse
Affiliation(s)
- Domenico Riccardi
- Department of Industrial Engineering, University of Salerno, Via Giovanni Paolo II, 132, 84084, Fisciano, SA, Italy
| | - Lucia Baldino
- Department of Industrial Engineering, University of Salerno, Via Giovanni Paolo II, 132, 84084, Fisciano, SA, Italy.
| | - Ernesto Reverchon
- Department of Industrial Engineering, University of Salerno, Via Giovanni Paolo II, 132, 84084, Fisciano, SA, Italy
| |
Collapse
|
23
|
Wang X, Sogo Y, Li X. Size Tuning of Mesoporous Silica Adjuvant for One-Shot Vaccination with Long-Term Anti-Tumor Effect. Pharmaceutics 2024; 16:516. [PMID: 38675177 PMCID: PMC11053635 DOI: 10.3390/pharmaceutics16040516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/05/2024] [Accepted: 04/05/2024] [Indexed: 04/28/2024] Open
Abstract
Despite recent clinical successes in cancer immunotherapy, it remains difficult to initiate a long-term anti-tumor effect. Therefore, repeated administrations of immune-activating agents are generally required in most cases. Herein, we propose an adjuvant particle size tuning strategy to initiate a long-term anti-tumor effect by one-shot vaccination. This strategy is based on the size-dependent immunostimulation mechanism of mesoporous silica particles. Hollow mesoporous silica (HMS) nanoparticles enhance the antigen uptake with dendritic cells around the immunization site in vivo. In contrast, hierarchically porous silica (HPS) microparticles prolong cancer antigen retention and release in vivo. The size tuning of the mesoporous silica adjuvant prepared by combining both nanoparticles and microparticles demonstrates the immunological properties of both components and has a long-term anti-tumor effect after one-shot vaccination. One-shot vaccination with HMS-HPS-ovalbumin (OVA)-Poly IC (PIC, a TLR3 agonist) increases CD4+ T cell, CD8+ T cell, and CD86+ cell populations in draining lymph nodes even 4 months after vaccination, as well as effector memory CD8+ T cell and tumor-specific tetramer+CD8+ T cell populations in splenocytes. The increases in the numbers of effector memory CD8+ T cells and tumor-specific tetramer+CD8+ T cells indicate that the one-shot vaccination with HMS-HPS-OVA-PIC achieved the longest survival time after a challenge with E.G7-OVA cells among all groups. The size tuning of the mesoporous silica adjuvant shows promise for one-shot vaccination that mimics multiple clinical vaccinations in future cancer immunoadjuvant development. This study may have important implications in the long-term vaccine design of one-shot vaccinations.
Collapse
Affiliation(s)
- Xiupeng Wang
- Health and Medical Research Institute, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba 305-8566, Ibaraki, Japan; (Y.S.); (X.L.)
| | | | | |
Collapse
|
24
|
Hyder A, Ali A, Buledi JA, Memon AA, Iqbal M, Bangalni TH, Solangi AR, Thebo KH, Akhtar J. Nanodiamonds: A Cutting-Edge Approach to Enhancing Biomedical Therapies and Diagnostics in Biosensing. CHEM REC 2024; 24:e202400006. [PMID: 38530037 DOI: 10.1002/tcr.202400006] [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: 01/11/2024] [Revised: 02/25/2024] [Indexed: 03/27/2024]
Abstract
Nanodiamonds (NDs) have garnered attention in the field of nanomedicine due to their unique properties. This review offers a comprehensive overview of NDs synthesis methods, properties, and their uses in biomedical applications. Various synthesis techniques, such as detonation, high-pressure, high-temperature, and chemical vapor deposition, offer distinct advantages in tailoring NDs' size, shape, and surface properties. Surface modification methods further enhance NDs' biocompatibility and enable the attachment of bioactive molecules, expanding their applicability in biological systems. NDs serve as promising nanocarriers for drug delivery, showcasing biocompatibility and the ability to encapsulate therapeutic agents for targeted delivery. Additionally, NDs demonstrate potential in cancer treatment through hyperthermic therapy and vaccine enhancement for improved immune responses. Functionalization of NDs facilitates their utilization in biosensors for sensitive biomolecule detection, aiding in precise diagnostics and rapid detection of infectious diseases. This review underscores the multifaceted role of NDs in advancing biomedical applications. By synthesizing NDs through various methods and modifying their surfaces, researchers can tailor their properties for specific biomedical needs. The ability of NDs to serve as efficient drug delivery vehicles holds promise for targeted therapy, while their applications in hyperthermic therapy and vaccine enhancement offer innovative approaches to cancer treatment and immunization. Furthermore, the integration of NDs into biosensors enhances diagnostic capabilities, enabling rapid and sensitive detection of biomolecules and infectious diseases. Overall, the diverse functionalities of NDs underscore their potential as valuable tools in nanomedicine, paving the way for advancements in healthcare and biotechnology.
Collapse
Affiliation(s)
- Ali Hyder
- National Centre of Excellence in Analytical Chemistry, University of Sindh, Jamshoro, 76080, Pakistan
| | - Akbar Ali
- State Key Laboratory of Multi-phase Complex Systems, Institute of Process Engineering (IPE), Chinese Academy of Sciences, Beijing, 100F190, China
- University of the Chinese Academy of Sciences, 19 A Yuquan Road, Beijing, 100049, China
| | - Jamil A Buledi
- National Centre of Excellence in Analytical Chemistry, University of Sindh, Jamshoro, 76080, Pakistan
| | - Ayaz Ali Memon
- National Centre of Excellence in Analytical Chemistry, University of Sindh, Jamshoro, 76080, Pakistan
| | - Muzaffar Iqbal
- Department of Chemistry, Faculty of Physical and Applied Sciences, The University of Haripur KPK, Haripur, 22620, Pakistan
| | - Talib Hussain Bangalni
- National Centre of Excellence in Analytical Chemistry, University of Sindh, Jamshoro, 76080, Pakistan
| | - Amber R Solangi
- National Centre of Excellence in Analytical Chemistry, University of Sindh, Jamshoro, 76080, Pakistan
| | - Khalid Hussain Thebo
- Institute of Metal Research (IMR), Chinese Academy of Science, 2 Wenhua Rood, Shenyang, China
- Department of Chemistry Mirpur, University of Science and Technology (MUST), 10250 (AJK), Mirpur, Pakistan
| | - Javeed Akhtar
- Department of Chemistry Mirpur, University of Science and Technology (MUST), 10250 (AJK), Mirpur, Pakistan
| |
Collapse
|
25
|
Steffens RC, Folda P, Fendler NL, Höhn M, Bücher-Schossau K, Kempter S, Snyder NL, Hartmann L, Wagner E, Berger S. GalNAc- or Mannose-PEG-Functionalized Polyplexes Enable Effective Lectin-Mediated DNA Delivery. Bioconjug Chem 2024; 35:351-370. [PMID: 38440876 DOI: 10.1021/acs.bioconjchem.3c00546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
A cationic, dendrimer-like oligo(aminoamide) carrier with four-arm topology based on succinoyl tetraethylene pentamine and histidines, cysteines, and N-terminal azido-lysines was screened for plasmid DNA delivery on various cell lines. The incorporated azides allow modification with various shielding agents of different polyethylene glycol (PEG) lengths and/or different ligands by copper-free click reaction, either before or after polyplex formation. Prefunctionalization was found to be advantageous over postfunctionalization in terms of nanoparticle formation, stability, and efficacy. A length of 24 ethylene oxide repetition units and prefunctionalization of ≥50% of azides per carrier promoted optimal polyplex shielding. PEG shielding resulted in drastically reduced DNA transfer, which could be successfully restored by active lectin targeting via novel GalNAc or mannose ligands, enabling enhanced receptor-mediated endocytosis of the carrier system. The involvement of the asialoglycoprotein receptor (ASGPR) in the uptake of GalNAc-functionalized polyplexes was confirmed in the ASGPR-positive hepatocarcinoma cell lines HepG2 and Huh7. Mannose-modified polyplexes showed superior cellular uptake and transfection efficacy compared to unmodified and shielded polyplexes in mannose-receptor-expressing dendritic cell-like DC2.4 cells.
Collapse
Affiliation(s)
- Ricarda C Steffens
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany
- Center for NanoScience (CeNS), LMU Munich, 80799 Munich, Germany
| | - Paul Folda
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany
| | - Nikole L Fendler
- Department of Chemistry, Davidson College, Davidson, North Carolina 28035, United States
| | - Miriam Höhn
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany
| | - Katharina Bücher-Schossau
- Institute of Organic Chemistry and Macromolecular Chemistry, Heinrich-Heine-University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Susanne Kempter
- Faculty of Physics, LMU Munich, 80539 Munich, Germany
- Center for NanoScience (CeNS), LMU Munich, 80799 Munich, Germany
| | - Nicole L Snyder
- Department of Chemistry, Davidson College, Davidson, North Carolina 28035, United States
| | - Laura Hartmann
- Institute of Organic Chemistry and Macromolecular Chemistry, Heinrich-Heine-University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
- Institute for Macromolecular Chemistry, University Freiburg, Stefan-Meier-Str. 31, 79104 Freiburg im Breisgau, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany
- Center for NanoScience (CeNS), LMU Munich, 80799 Munich, Germany
| | - Simone Berger
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany
- Center for NanoScience (CeNS), LMU Munich, 80799 Munich, Germany
| |
Collapse
|
26
|
Dehghankhold M, Sadat Abolmaali S, Nezafat N, Mohammad Tamaddon A. Peptide nanovaccine in melanoma immunotherapy. Int Immunopharmacol 2024; 129:111543. [PMID: 38301413 DOI: 10.1016/j.intimp.2024.111543] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 01/03/2024] [Accepted: 01/11/2024] [Indexed: 02/03/2024]
Abstract
Melanoma is an especially fatal neoplasm resistant to traditional treatment. The advancement of novel therapeutical approaches has gained attention in recent years by shedding light on the molecular mechanisms of melanoma tumorigenesis and their powerful interplay with the immune system. The presence of many mutations in melanoma cells results in the production of a varied array of antigens. These antigens can be recognized by the immune system, thereby enabling it to distinguish between tumors and healthy cells. In the context of peptide cancer vaccines, generally, they are designed based on tumor antigens that stimulate immunity through antigen-presenting cells (APCs). As naked peptides often have low potential in eliciting a desirable immune reaction, immunization with such compounds usually necessitates adjuvants and nanocarriers. Actually, nanoparticles (NPs) can provide a robust immune response to peptide-based melanoma vaccines. They improve the directing of peptide vaccines to APCs and induce the secretion of cytokines to get maximum immune response. This review provides an overview of the current knowledge of the utilization of nanotechnology in peptide vaccines emphasizing melanoma, as well as highlights the significance of physicochemical properties in determining the fate of these nanovaccines in vivo, including their drainage to lymph nodes, cellular uptake, and influence on immune responses.
Collapse
Affiliation(s)
- Mahvash Dehghankhold
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Samira Sadat Abolmaali
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Navid Nezafat
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Computational vaccine and Drug Design Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Ali Mohammad Tamaddon
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
27
|
Zhou Z, Shu T, Su L, Zhang X. Size-matching compositing nanoprobe of AIE-type gold nanocluster supramolecular nanogels wrapped by hypergravity-tailored MnO 2 nanosheets for cellular glutathione detection. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 308:123690. [PMID: 38043289 DOI: 10.1016/j.saa.2023.123690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 11/08/2023] [Accepted: 11/26/2023] [Indexed: 12/05/2023]
Abstract
Compositing has been the main approach for material creation via wisely combining material components with different properties. MnO2 nanosheets (MNSs) with thin 2 D morphology are usually applied to composite molecules or nanomaterials for biosensing and bioimaging applications. However, such composition is actually structurally unmatched, albeit performance matching. Here, a series of benefits merely on the basis of structural match have been unearthed via tailoring MNSs with four sizes by synthesis under controllable hypergravity field. The classical fluorophore-quencher couple was utilized as the subject model, where the soft supramolecular nanogels based on aggregation-induced emission (AIE)-active gold nanoclusters were wrapped by MNSs of strong absorption. By comparative study of one-on-one wrapping and one-to-many encapsulation with geometrical selection of different MNSs, we found that the one-on-one wrapping model protected weakly-bonded nanogels from combination-induced distortion and strengthened nanogel networks via endowing exoskeleton. Besides, wrapping pattern and size-match significantly enhanced the quenching efficiency of MNSs towards the emissive nanogels. More importantly, the well-wrapped nanocomposites had considerable enhanced biological compatibility with much lower cytotoxicity and higher transfection capacity than the untailored MNSs composite and could serve as cellular glutathione detection.
Collapse
Affiliation(s)
- Ziping Zhou
- Shenzhen Key Laboratory for Nano-Biosensing Technology, Research Center for Biosensor and Nanotheranostic, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518060, PR China; Beijing Key Laboratory for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, PR China; Aerospace Research Institute of Materials & Processing Technology, Science and Technology on Advanced Functional Composites Laboratory, Beijing 100076, PR China
| | - Tong Shu
- Shenzhen Key Laboratory for Nano-Biosensing Technology, Research Center for Biosensor and Nanotheranostic, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518060, PR China.
| | - Lei Su
- Shenzhen Key Laboratory for Nano-Biosensing Technology, Research Center for Biosensor and Nanotheranostic, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518060, PR China
| | - Xueji Zhang
- Shenzhen Key Laboratory for Nano-Biosensing Technology, Research Center for Biosensor and Nanotheranostic, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518060, PR China.
| |
Collapse
|
28
|
Puccetti M, Pariano M, Schoubben A, Giovagnoli S, Ricci M. Biologics, theranostics, and personalized medicine in drug delivery systems. Pharmacol Res 2024; 201:107086. [PMID: 38295917 DOI: 10.1016/j.phrs.2024.107086] [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: 12/26/2023] [Revised: 01/25/2024] [Accepted: 01/25/2024] [Indexed: 02/05/2024]
Abstract
The progress in human disease treatment can be greatly advanced through the implementation of nanomedicine. This approach involves targeted and cell-specific therapy, controlled drug release, personalized dosage forms, wearable drug delivery, and companion diagnostics. By integrating cutting-edge technologies with drug delivery systems, greater precision can be achieved at the tissue and cellular levels through the use of stimuli-responsive nanoparticles, and the development of electrochemical sensor systems. This precision targeting - by virtue of nanotechnology - allows for therapy to be directed specifically to affected tissues while greatly reducing side effects on healthy tissues. As such, nanomedicine has the potential to transform the treatment of conditions such as cancer, genetic diseases, and chronic illnesses by facilitating precise and cell-specific drug delivery. Additionally, personalized dosage forms and wearable devices offer the ability to tailor treatment to the unique needs of each patient, thereby increasing therapeutic effectiveness and compliance. Companion diagnostics further enable efficient monitoring of treatment response, enabling customized adjustments to the treatment plan. The question of whether all the potential therapeutic approaches outlined here are viable alternatives to current treatments is also discussed. In general, the application of nanotechnology in the field of biomedicine may provide a strong alternative to existing treatments for several reasons. In this review, we aim to present evidence that, although in early stages, fully merging advanced technology with innovative drug delivery shows promise for successful implementation across various disease areas, including cancer and genetic or chronic diseases.
Collapse
Affiliation(s)
- Matteo Puccetti
- Department of Pharmaceutical Sciences, University of Perugia, Italy,.
| | | | | | | | - Maurizio Ricci
- Department of Pharmaceutical Sciences, University of Perugia, Italy,.
| |
Collapse
|
29
|
Ma B, Li Q, Mi Y, Zhang J, Tan W, Guo Z. pH-responsive nanogels with enhanced antioxidant and antitumor activities on drug delivery and smart drug release. Int J Biol Macromol 2024; 257:128590. [PMID: 38056756 DOI: 10.1016/j.ijbiomac.2023.128590] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 11/12/2023] [Accepted: 12/01/2023] [Indexed: 12/08/2023]
Abstract
pH-responsive nanogels have played an increasingly momentous role in tumor treatment. The focus of this study is to design and develop pH-responsive benzimidazole-chitosan quaternary ammonium salt (BIMIXHAC) nanogels for the controlled release of doxorubicin hydrochloride (DOX) while enhancing its hydrophilicity. BIMIXHAC is crosslinked with carboxymethyl chitosan (CMC), hyaluronic acid sodium salt (HA), and sodium alginates (SA) using an ion crosslinking method. The chemical structure of chitosan derivatives was verified by 1H NMR and FT-IR techniques. Compared to hydroxypropyl trimethyl ammonium chloride chitosan (HACC)-based nanogels, BIMIXHAC-based nanogels exhibit better drug encapsulation efficiency and loading capacity (BIMIXHAC-D-HA 91.76 %, and 32.23 %), with pH-responsive release profiles and accelerated release in vitro. The series of nanogels formed by crosslinking with three different polyanionic crosslinkers have different particle size potentials and antioxidant properties. BIMIXHAC-HA, BIMIXHAC-SA and BIMIXHAC-CMC demonstrate favorable antioxidant capability. In addition, cytotoxicity tests showed that BIMIXHAC-based nanogels have high biocompatibility. BIMIXHAC-based nanogels exhibit preferable anticancer effects on MCF-7 and A549 cells. Furthermore, the BIMIXHAC-D-HA nanogel was 2.62 times less toxic than DOX to L929 cells. These results suggest that BIMIXHAC-based nanogels can serve as pH-responsive nanoplatforms for the delivery of anticancer drugs.
Collapse
Affiliation(s)
- Bing Ma
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; University of Chinese Academy of Sciences, Beijing 100049, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China
| | - Qing Li
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China
| | - Yingqi Mi
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China
| | - Jingjing Zhang
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China
| | - Wenqiang Tan
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China
| | - Zhanyong Guo
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; University of Chinese Academy of Sciences, Beijing 100049, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China.
| |
Collapse
|
30
|
Ma B, Li Q, Zhang J, Mi Y, Tan W, Guo Z. Improvement of the Antioxidant and Antitumor Activities of Benzimidazole-Chitosan Quaternary Ammonium Salt on Drug Delivery Nanogels. Mar Drugs 2024; 22:40. [PMID: 38248665 PMCID: PMC10817266 DOI: 10.3390/md22010040] [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: 12/04/2023] [Revised: 01/07/2024] [Accepted: 01/10/2024] [Indexed: 01/23/2024] Open
Abstract
The present study focused on the design and preparation of acid-responsive benzimidazole-chitosan quaternary ammonium salt (BIMIXHAC) nanogels for a controlled, slow-release of Doxorubicin HCl (DOX.HCl). The BIMIXHAC was crosslinked with sodium tripolyphosphate (TPP) using the ion crosslinking method. The method resulted in nanogels with low polydispersity index, small particle size, and positive zeta potential values, indicating the good stability of the nanogels. Compared to hydroxypropyl trimethyl ammonium chloride chitosan-Doxorubicin HCl-sodium tripolyphosphate (HACC-D-TPP) nanogel, the benzimidazole-chitosan quaternary ammonium salt-Doxorubicin HCl-sodium tripolyphosphate (BIMIXHAC-D-TPP) nanogel show higher drug encapsulation efficiency and loading capacity (BIMIXHAC-D-TPP 93.17 ± 0.27% and 31.17 ± 0.09%), with acid-responsive release profiles and accelerated release in vitro. The hydroxypropyl trimethyl ammonium chloride chitosan-sodium tripolyphosphate (HACC-TPP), and benzimidazole-chitosan quaternary ammonium salt-sodium tripolyphosphate (BIMIXHAC-TPP) nanogels demonstrated favorable antioxidant capability. The assay of cell viability, measured by the MTT assay, revealed that nanogels led to a significant reduction in the cell viability of two cancer cells: the human lung adenocarcinoma epithelial cell line (A549) and the human breast cancer cell line (MCF-7). Furthermore, the BIMIXHAC-D-TPP nanogel was 2.96 times less toxic than DOX.HCl to the mouse fibroblast cell line (L929). It was indicated that the BIMIXHAC-based nanogel with enhanced antioxidant and antitumor activities and acidic-responsive release could serve as a potential nanocarrier.
Collapse
Affiliation(s)
- Bing Ma
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; (B.M.); (Q.L.); (J.Z.); (Y.M.); (W.T.)
- University of Chinese Academy of Sciences, Beijing 100049, China
- Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China
| | - Qing Li
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; (B.M.); (Q.L.); (J.Z.); (Y.M.); (W.T.)
- Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China
| | - Jingjing Zhang
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; (B.M.); (Q.L.); (J.Z.); (Y.M.); (W.T.)
- Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China
| | - Yingqi Mi
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; (B.M.); (Q.L.); (J.Z.); (Y.M.); (W.T.)
- Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China
| | - Wenqiang Tan
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; (B.M.); (Q.L.); (J.Z.); (Y.M.); (W.T.)
- Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China
| | - Zhanyong Guo
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; (B.M.); (Q.L.); (J.Z.); (Y.M.); (W.T.)
- University of Chinese Academy of Sciences, Beijing 100049, China
- Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China
| |
Collapse
|
31
|
Margaroni M, Tsanaktsidou E, Agallou M, Kiparissides C, Kammona O, Karagouni E. Development of a novel squalene/α-tocopherol-based self-emulsified nanoemulsion incorporating Leishmania peptides for induction of antigen-specific immune responses. Int J Pharm 2024; 649:123621. [PMID: 38000650 DOI: 10.1016/j.ijpharm.2023.123621] [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: 07/25/2023] [Revised: 11/02/2023] [Accepted: 11/15/2023] [Indexed: 11/26/2023]
Abstract
Vaccination has emerged as the most effective strategy to confront infectious diseases, among which is leishmaniasis, that threat public health. Despite laborious efforts there is still no vaccine for humans to confront leishmaniasis. Multi-epitope protein/peptide vaccines present a number of advantages, however their use along with appropriate adjuvants that may also act as antigen carriers is considered essential to overcome subunit vaccines' low immunogenicity. In the present study, a stable self-emulsified nanoemulsion was developed and double-adjuvanted with squalene and α-tocopherol. The prepared nanoemulsion droplets exhibited low cytotoxicity in a certain range of concentrations, while they were efficiently taken up by macrophages and dendritic cells in vitro as well as in vivo in secondary lymphoid organs. To further characterize nanoformulation's potent antigen delivery capability, three multi-epitope Leishmania peptides were incorporated into the nanoemulsion. Peptide encapsulation resulted in dendritic cells' functional differentiation characterized by elevated levels of maturation markers and intracellular cytokine production. Intramuscular administration of the nanoemulsion incorporating Leishmania peptides induced antigen-specific spleen cell proliferation as well as elicitation of CD4+ central memory cells, supporting the potential of the developed nanoformulation to successfully act also as an antigen delivery vehicle and thus encouraging further preclinical studies on its vaccine candidate potency.
Collapse
Affiliation(s)
- Maritsa Margaroni
- Immunology of Infection Laboratory, Hellenic Pasteur Institute, 125 21 Athens, Greece.
| | - Evgenia Tsanaktsidou
- Chemical Process & Energy Resources Institute, Centre for Research and Technology Hellas, P.O. Box 60361, 57 001 Thessaloniki, Greece.
| | - Maria Agallou
- Immunology of Infection Laboratory, Hellenic Pasteur Institute, 125 21 Athens, Greece.
| | - Costas Kiparissides
- Chemical Process & Energy Resources Institute, Centre for Research and Technology Hellas, P.O. Box 60361, 57 001 Thessaloniki, Greece; Department of Chemical Engineering, Aristotle University of Thessaloniki, P.O. Box 472, 54 124 Thessaloniki, Greece.
| | - Olga Kammona
- Chemical Process & Energy Resources Institute, Centre for Research and Technology Hellas, P.O. Box 60361, 57 001 Thessaloniki, Greece.
| | - Evdokia Karagouni
- Immunology of Infection Laboratory, Hellenic Pasteur Institute, 125 21 Athens, Greece.
| |
Collapse
|
32
|
Gao Y, Yang J, Zhang Y, Zhao Y, Zhao X, Zhang X, Zhang J, Mao L, Wang H, Wang H, Wang L. In vitro and in vivo evaluation of immune response of poly(lactic acid) nanoparticles with different end groups. Int J Biol Macromol 2023; 253:126593. [PMID: 37659499 DOI: 10.1016/j.ijbiomac.2023.126593] [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: 05/12/2023] [Revised: 08/26/2023] [Accepted: 08/27/2023] [Indexed: 09/04/2023]
Abstract
Poly(lactic acid) (PLA) has excellent properties of biodegradability and biocompatibility, which is a US Food and Drug Administration (FDA) approved biopolymer for the preparation of safe and effective vaccines, drugs, and gene delivery systems. However, there still exists a great problem whether and how the end group affects the immune response of PLA vaccines. Therefore, the aim of this study was to evaluate the in vitro and in vivo of immune response of PLA nanoparticles (NPs) with carboxyl (COOH) and ester (COOR) end groups. In vitro experiments suggested COOH NPs could promote the higher phagocytosis and activation of bone marrow dendritic cells (BMDCs) with a lower cytotoxicity. In vivo experiments showed that COOR NPs and COOH NPs could strongly elicit IgG, IgG1, and IgG2a responses both in the short and long-terms. However, the highest T cell and B cell activation, and central memory T cells response was induced by COOH NPs. In addition, the COOH NPs could significantly enhance splenocytes proliferation and cytokines secretion. Thus, the PLA with the COOH end group shows greater potential as efficient carrier materials of NPs for enhancing cellular and humoral immune responses.
Collapse
Affiliation(s)
- Yuan Gao
- State Key Laboratory of Subtropical Silviculture, College of Forestry and Biotechnology, Zhejiang A&F University, Hangzhou 311300, Zhejiang, China; Key Laboratory of Green Process and Engineering, State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; Key Laboratory of Forest Plant Ecology, Ministry of Education, Heilongjiang Provincial Key Laboratory of Ecological Utilization of Forestry-Based Active Substances, College of Chemistry, Chemistry Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, China
| | - Jun Yang
- Beijing Tide Pharmaceutical Co., Ltd, No.8 East Rongjing Street, Beijing Econnomi Technological Development Area (BDA), Beijing 100176, China
| | - Yaru Zhang
- Key Laboratory of Green Process and Engineering, State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; Key Laboratory of Forest Plant Ecology, Ministry of Education, Heilongjiang Provincial Key Laboratory of Ecological Utilization of Forestry-Based Active Substances, College of Chemistry, Chemistry Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, China
| | - Ying Zhao
- Key Laboratory of Green Process and Engineering, State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; Key Laboratory of Forest Plant Ecology, Ministry of Education, Heilongjiang Provincial Key Laboratory of Ecological Utilization of Forestry-Based Active Substances, College of Chemistry, Chemistry Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, China
| | - Xin Zhao
- Key Laboratory of Green Process and Engineering, State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; Heilongjiang University of Chinese Medicine, Harbin 150036, China
| | - Xining Zhang
- Key Laboratory of Green Process and Engineering, State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jing Zhang
- Key Laboratory of Green Process and Engineering, State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Li Mao
- Beijing Tide Pharmaceutical Co., Ltd, No.8 East Rongjing Street, Beijing Econnomi Technological Development Area (BDA), Beijing 100176, China.
| | - Hongjun Wang
- Beijing Tide Pharmaceutical Co., Ltd, No.8 East Rongjing Street, Beijing Econnomi Technological Development Area (BDA), Beijing 100176, China.
| | - Huimei Wang
- State Key Laboratory of Subtropical Silviculture, College of Forestry and Biotechnology, Zhejiang A&F University, Hangzhou 311300, Zhejiang, China.
| | - Lianyan Wang
- Key Laboratory of Green Process and Engineering, State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
33
|
Parmaksız S, Pekcan M, Özkul A, Türkmen E, Rivero-Arredondo V, Ontiveros-Padilla L, Forbes N, Perrie Y, López-Macías C, Şenel S. In vivo evaluation of new adjuvant systems based on combination of Salmonella Typhi porins with particulate systems: Liposomes versus polymeric particles. Int J Pharm 2023; 648:123568. [PMID: 37925042 DOI: 10.1016/j.ijpharm.2023.123568] [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: 08/20/2023] [Revised: 10/29/2023] [Accepted: 10/30/2023] [Indexed: 11/06/2023]
Abstract
Subunit vaccines that have weak immunogenic activity require adjuvant systems for enhancedcellular and long-acting humoral immune responses. Both lipid-based and polymeric-based particulate adjuvants have been widely investigated to induce the desired immune responses against the subunit vaccines. The adjuvant efficacy of these particulate adjuvants depends upon their physicochemical properties such as particle size, surface charge, shape and their composition. Previously, we showed in vitro effect of adjuvant systems based on combination of chitosan and Salmonella Typhi porins in microparticle or nanoparticle form, which were spherical with positive surface charge. In the present study, we have further developed an adjuvant system based on combination of porins with liposomes (cationic and neutral) and investigated the adjuvant effect of both the liposomal and polymeric systems in BALB/c mice using a model antigen, ovalbumin. Humoral immune responses were determined following priming and booster dose at 15-day intervals. In overall, IgM and IgG levels were induced in the presence of both the liposomal and polymeric adjuvant systems indicating the positive impact of combination with porins. The highest IgM levels were obtained on Day 8, and liposomal adjuvant systems were found to elicit significantly higher IgM levels compared to polymeric systems. IgG levels were increased significantly after booster, particularly more profound with the micro-sized polymeric system when compared to cationic liposomal system with nano-size. Our results demonstrated that the developed particulate systems are promising both as an adjuvant and delivery system, providing enhanced immune responses against subunit antigens, and have the potential for long-term protection.
Collapse
Affiliation(s)
- Selin Parmaksız
- Hacettepe University, Faculty of Pharmacy, Department of Pharmaceutical Technology, 06100 Ankara, Turkey
| | - Mert Pekcan
- Ankara University, Faculty of Veterinary Medicine, Department of Biochemistry, 06110 Ankara, Turkey
| | - Aykut Özkul
- Ankara University, Faculty of Veterinary Medicine, Department of Virology, Ankara University, 06110 Ankara, Turkey
| | - Ece Türkmen
- Hacettepe University, Faculty of Pharmacy, Department of Pharmaceutical Technology, 06100 Ankara, Turkey
| | - Vanessa Rivero-Arredondo
- Medical Research Unit on Immunochemistry, Specialties Hospital, National Medical Centre "Siglo XXI", Mexican Social Security Institute (IMSS), Mexico City, Mexico
| | - Luis Ontiveros-Padilla
- Medical Research Unit on Immunochemistry, Specialties Hospital, National Medical Centre "Siglo XXI", Mexican Social Security Institute (IMSS), Mexico City, Mexico
| | - Neil Forbes
- University of Strathclyde, Strathclyde Institute of Pharmacy and Biomedical Sciences, Glasgow, United Kingdom
| | - Yvonne Perrie
- University of Strathclyde, Strathclyde Institute of Pharmacy and Biomedical Sciences, Glasgow, United Kingdom
| | - Constantino López-Macías
- Medical Research Unit on Immunochemistry, Specialties Hospital, National Medical Centre "Siglo XXI", Mexican Social Security Institute (IMSS), Mexico City, Mexico
| | - Sevda Şenel
- Hacettepe University, Faculty of Pharmacy, Department of Pharmaceutical Technology, 06100 Ankara, Turkey.
| |
Collapse
|
34
|
Rahiman N, Zamani P, Arabi L, Alavizadeh SH, Nikpoor A, Mashreghi M, Badiee A, Jaafari MR. Novel liposomal glatiramer acetate: Preparation and immunomodulatory evaluation in murine model of multiple sclerosis. Int J Pharm 2023; 648:123620. [PMID: 37981250 DOI: 10.1016/j.ijpharm.2023.123620] [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: 06/12/2023] [Revised: 10/23/2023] [Accepted: 11/15/2023] [Indexed: 11/21/2023]
Abstract
The frequent administration rate required for Glatiramer acetate (GA), a first-line therapy for Multiple sclerosis (MS), poses patient compliance issues. Only a small portion of the subcutaneously administered GA is available for phagocytosis by macrophages, as most of it is hydrolyzed at its administration site or excreted renally. To unravel these hurdles, we have prepared liposomal formulations of GA through thin film-hydration method plus extrusion. The clinical and histopathological efficacy of GA-loaded liposomes were assessed in prophylactic and therapeutic manners on murine model of MS (experimental autoimmune encephalomyelitis (EAE)). The selected GA liposomal formulation showed favorable size (275 nm on average), high loading efficiency, and high macrophage localization. Moreover, administration of GA-liposomes in mice robustly suppressed the inflammatory responses and decreased the inflammatory and demyelinated lesion regions in CNS compared to the free GA with subsequent reduction of the EAE clinical score. Our study indicated that liposomal GA could be served as a reliable nanomedicine-based platform to hopefully curb MS-related aberrant autoreactive immune responses with higher efficacy, longer duration of action, fewer administration frequencies, and higher delivery rate to macrophages. This platform has the potential to be introduced as a vaccine for MS after clinical translation and merits further investigations.
Collapse
Affiliation(s)
- Niloufar Rahiman
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parvin Zamani
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Arabi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyedeh Hoda Alavizadeh
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Aminreza Nikpoor
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Mohammad Mashreghi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Badiee
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
35
|
Joy R, Siddiqua H, Sharma S, Raveendran M, John F, Hassan P, Gawali SL, Raghavan SC, George J. Block Copolymer Encapsulation of Disarib, an Inhibitor of BCL2 for Improved Chemotherapeutic Potential. ACS OMEGA 2023; 8:40729-40740. [PMID: 37929147 PMCID: PMC10621013 DOI: 10.1021/acsomega.3c05802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/06/2023] [Accepted: 10/09/2023] [Indexed: 11/07/2023]
Abstract
A chemical inhibitor of antiapoptotic protein, BCL2, known as Disarib, suffers poor solubility in aqueous environments; thereby limiting its potential as a chemotherapeutic agent. To overcome this limitation and enhance the therapeutic efficacy of Disarib, we have employed the encapsulation of this small molecule inhibitor within P123 copolymer matrix. Micelles were synthesized using a thin-film hydration technique, and a comprehensive analysis was undertaken to evaluate the resulting micelle properties, including morphology, particle size, intermolecular interactions, encapsulation efficiency, and in vitro release characteristics. This assessment utilized various physicochemical techniques including UV spectroscopy, FTIR spectroscopy, dynamic light scattering (DLS), transmission electron microscopy (TEM), and small-angle X-ray scattering (SAXS). Disarib-loaded P123 micelle formulation denoted as P123D exhibited a well-defined particle size of approximately 29.2 nm spherical core-shell morphology. Our investigations revealed a notable encapsulation efficiency of 75%, and we observed a biphasic release pattern for the encapsulated Disarib. Furthermore, our cytotoxicity assessment of P123D micelles against mouse breast adenocarcinoma, mouse lymphoma, and human leukemic cell lines showed 40-45% increase in cytotoxicity compared with the administration of Disarib alone in the breast adenocarcinoma cell line. Enhancement in the cytotoxicity of P123D was found to be higher or limited; however, it is important to observe that the encapsulation method significantly enhanced the aqueous solubility of Disarib as it has the best solubility in dimethyl sulfoxide (DMSO) in the unencapsulated state.
Collapse
Affiliation(s)
- Reshma Joy
- Bio-organic
Laboratory, Department of Chemistry, Sacred
Heart College, Kochi 682013, India
| | - Humaira Siddiqua
- Department
of Biochemistry, Indian Institute of Science, Bangalore 560012, India
| | - Shivangi Sharma
- Department
of Biochemistry, Indian Institute of Science, Bangalore 560012, India
| | - Manthra Raveendran
- Department
of Biochemistry, Indian Institute of Science, Bangalore 560012, India
| | - Franklin John
- Bio-organic
Laboratory, Department of Chemistry, Sacred
Heart College, Kochi 682013, India
| | | | - Santosh L Gawali
- Chemistry
Division, Bhabha Atomic Research Centre, Mumbai 400085, India
| | - Sathees C. Raghavan
- Department
of Biochemistry, Indian Institute of Science, Bangalore 560012, India
| | - Jinu George
- Bio-organic
Laboratory, Department of Chemistry, Sacred
Heart College, Kochi 682013, India
| |
Collapse
|
36
|
Lazar KM, Shetty S, Chilkoti A, Collier JH. Immune-active polymeric materials for the treatment of inflammatory diseases. Curr Opin Colloid Interface Sci 2023; 67:101726. [DOI: 10.1016/j.cocis.2023.101726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
37
|
Shen M, Jiang H, Zhao Y, Wu L, Yang H, Yao Y, Meng H, Yang Q, Liu L, Li Y. Shear Stress and ROS Dual-Responsive RBC-Hitchhiking Nanoparticles for Atherosclerosis Therapy. ACS APPLIED MATERIALS & INTERFACES 2023; 15:43374-43386. [PMID: 37669139 DOI: 10.1021/acsami.3c07371] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/07/2023]
Abstract
Atherosclerosis (AS), a leading cause of death worldwide, is a chronic inflammatory disease rich in lipids and reactive oxygen species (ROS) within plaques. Therefore, lowering lipid and ROS levels is effective in treating AS and reducing AS-induced mortality. In this study, an intelligent biomimetic drug delivery system that specifically responded to both shear stress and ROS microenvironment was developed, consisting of red blood cells (RBCs) and cross-linked polyethyleneimine nanoparticles (SA PEI) loaded with a lipid-lowering drug simvastatin acid (SA), and RBCs were self-assembled with SA PEI to obtain biresponsive SA PEI@RBCs for the treatment of AS. SA PEI could achieve sustained release of SA in response to ROS and reduce ROS and lipid levels to achieve the purpose of treating AS. Shear stress model experiments showed that SA PEI@RBCs could respond to the high shear stress level (100 dynes/cm2) at plaques, realizing the desorption and enrichment of SA PEI and improving the therapeutic efficiency of SA PEI@RBCs. In vitro and in vivo experiments have confirmed that SA PEI@RBCs exhibits better in vivo safety and therapeutic efficacy than SA PEI and free SA. Therefore, shaping SA PEI@RBCs into a biomimetic drug delivery system with dual sensitivity to ROS and shear stress is an effective strategy and treatment to facilitate their delivery into plaques.
Collapse
Affiliation(s)
- Meili Shen
- Key Laboratory of Special Engineering Plastics Ministry of Education, College of Chemistry, Jilin University, Changchun 130012, Jilin, China
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun 130031, Jilin, China
| | - Hui Jiang
- Department of Blood Purification, Tong Liao City Hospital, Tong Liao 028000, Inner Mongolia, China
| | - Yan Zhao
- Department of Oncology and Hematology, China-Japan Union Hospital of Jilin University, Changchun 130031, Jilin, China
| | - Liangqiang Wu
- Key Laboratory of Special Engineering Plastics Ministry of Education, College of Chemistry, Jilin University, Changchun 130012, Jilin, China
| | - Haiqin Yang
- Key Laboratory of Special Engineering Plastics Ministry of Education, College of Chemistry, Jilin University, Changchun 130012, Jilin, China
| | - Yixuan Yao
- Key Laboratory of Special Engineering Plastics Ministry of Education, College of Chemistry, Jilin University, Changchun 130012, Jilin, China
| | - Hao Meng
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun 130031, Jilin, China
| | - Qingbiao Yang
- Key Laboratory of Special Engineering Plastics Ministry of Education, College of Chemistry, Jilin University, Changchun 130012, Jilin, China
| | - Linlin Liu
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun 130031, Jilin, China
| | - Yapeng Li
- Key Laboratory of Special Engineering Plastics Ministry of Education, College of Chemistry, Jilin University, Changchun 130012, Jilin, China
| |
Collapse
|
38
|
Liao X, Nie J, Yuan X, Feng Z, Cui E, Wu Y, Li Y, Scherman D, Liu Y. Carbopol dispersed PAA-modified UIO-66 with high colloidal stability as a combination nano-adjuvant boosts immune response and protection against pseudorabies virus in mice and pigs. Acta Biomater 2023; 168:540-550. [PMID: 37393970 DOI: 10.1016/j.actbio.2023.06.039] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 06/07/2023] [Accepted: 06/27/2023] [Indexed: 07/04/2023]
Abstract
Although inactivated vaccines have higher safety than live-attenuated vaccines in the control of pseudorabies virus (PRV), their protection efficacy is limited due to insufficient immunogenicity when used alone. High-performance adjuvants that can potentiate immune responses are highly desirable to improve the protection efficacy of inactivated vaccines. In this work, we have developed U@PAA-Car, a Carbopol dispersed zirconium-based metal-organic framework UIO-66 modified by polyacrylic acid (PAA), as a promising adjuvant for inactivated PRV vaccines. The U@PAA-Car has good biocompatibility, high colloidal stability, and antigen (vaccine) loading capacity. It significantly potentiates humoral and cellular immune responses over either U@PAA, Carbopol, or commercial adjuvants such as Alum and biphasic 201 by inducing a higher specific antibody titer, IgG2a/IgG1 ratio, cell cytokine secretion, and splenocyte proliferation. A protection rate of over 90% was observed in challenge tests in the model animal mice and the host animal pigs, which is much higher than that observed with commercial adjuvants. The high performance of the U@PAA-Car is attributed to antigen sustainable release at the injection site and highly efficient antigen internalization and presentation. In conclusion, this work not only demonstrates a great potential of the developed U@PAA-Car nano-adjuvant for the inactivated PRV vaccine but also gives a preliminary explanation of its action mechanism. STATEMENT OF SIGNIFICANCE: We have developed a Carbopol dispersed PAA-modified zirconium-based metal-organic framework UIO-66 (U@PAA-Car) as a promising combination nano-adjuvant for the inactivated PRV vaccine. The U@PAA-Car induced higher specific antibody titers and IgG2a/IgG1 ratio, increased cell cytokines secretion, and better splenocyte proliferation than U@PAA, Carbopol, and the commercial adjuvants Alum and biphasic 201, indicating that it induces a significant potentiation of humoral and cellular immune response. In addition, much higher protection rates were achieved with the U@PAA-Car-adjuvanted PRV vaccine in mice and pigs challenge than those observed from the commercial adjuvant groups. This work not only demonstrates the great potential of the U@PAA-Car nano-adjuvant in an inactivated PRV vaccine but also gives a preliminary explanation of its action mechanism.
Collapse
Affiliation(s)
- Xiaoling Liao
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Jingjing Nie
- Adjuvant Research Laboratory, Sinopharm Animal Health Co., Ltd., Wuhan 430073, China
| | - Xiangyang Yuan
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Zhao Feng
- Adjuvant Research Laboratory, Sinopharm Animal Health Co., Ltd., Wuhan 430073, China
| | - Endian Cui
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Yang Wu
- Adjuvant Research Laboratory, Sinopharm Animal Health Co., Ltd., Wuhan 430073, China.
| | - Yuan Li
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing 402160, China.
| | - Daniel Scherman
- Université Paris Cité, CNRS, INSERM, UTCBS, Unité de Technologies Chimiques et Biologiques pour la Santé, F-75006 Paris, France
| | - Yingshuai Liu
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy, Southwest University, Chongqing 400715, China.
| |
Collapse
|
39
|
Danaeifar M, Negahdari B, Eslam HM, Zare H, Ghanaat M, Koushali SS, Malekshahi ZV. Polymeric nanoparticles for DNA vaccine-based cancer immunotherapy: a review. Biotechnol Lett 2023; 45:1053-1072. [PMID: 37335426 DOI: 10.1007/s10529-023-03383-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 03/28/2023] [Accepted: 04/11/2023] [Indexed: 06/21/2023]
Abstract
Cancer is one of the leading causes of death and mortality in the world. There is an essential need to develop new drugs or therapeutic approaches to manage treatment-resistant cancers. Cancer immunotherapy is a type of cancer treatment that uses the power of the body's immune system to prevent, control, and eliminate cancer. One of the materials used as a vaccine in immunotherapy is DNA. The application of polymeric nanoparticles as carriers for DNA vaccines could be an effective therapeutic approach to activate immune responses and increase antigen presentation efficiency. Various materials have been used as polymeric nanoparticles, including: chitosan, poly (lactic-co-glycolic acid), Polyethylenimine, dendrimers, polypeptides, and polyesters. Application of these polymer nanoparticles has several advantages, including increased vaccine delivery, enhanced antigen presentation, adjuvant effects, and more sustainable induction of the immune system. Besides many clinical trials and commercial products that were developed based on polymer nanoparticles, there is still a need for more comprehensive studies to increase the DNA vaccine efficiency in cancer immunotherapy using this type of carrier.
Collapse
Affiliation(s)
- Mohsen Danaeifar
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Negahdari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Houra Mobaleghol Eslam
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamed Zare
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Momeneh Ghanaat
- Department of Microbiology, Ayatollah Amoli Branch, Islamic Azad University, Amol, Iran
| | - Sekinehe Shokouhi Koushali
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Ziba Veisi Malekshahi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
40
|
Sadr S, Poorjafari Jafroodi P, Haratizadeh MJ, Ghasemi Z, Borji H, Hajjafari A. Current status of nano-vaccinology in veterinary medicine science. Vet Med Sci 2023; 9:2294-2308. [PMID: 37487030 PMCID: PMC10508510 DOI: 10.1002/vms3.1221] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 04/11/2023] [Accepted: 07/14/2023] [Indexed: 07/26/2023] Open
Abstract
Vaccination programmes provide a safe, effective and cost-efficient strategy for maintaining population health. In veterinary medicine, vaccination not only reduces disease within animal populations but also serves to enhance public health by targeting zoonoses. Nevertheless, for many pathogens, an effective vaccine remains elusive. Recently, nanovaccines have proved to be successful for various infectious and non-infectious diseases of animals. These novel technologies, such as virus-like particles, self-assembling proteins, polymeric nanoparticles, liposomes and virosomes, offer great potential for solving many of the vaccine production challenges. Their benefits include low immunotoxicity, antigen stability, enhanced immunogenicity, flexibility sustained release and the ability to evoke both humoral and cellular immune responses. Nanovaccines are more efficient than traditional vaccines due to ease of control and plasticity in their physio-chemical properties. They use a highly targeted immunological approach which can provide strong and long-lasting immunity. This article reviews the currently available nanovaccine technology and considers its utility for both infectious diseases and non-infectious diseases such as auto-immunity and cancer. Future research opportunities and application challenges from bench to clinical usage are also discussed.
Collapse
Affiliation(s)
- Soheil Sadr
- Department of Clinical SciencesFaculty of Veterinary MedicineFerdowsi University of MashhadMashhadIran
| | | | | | - Zahra Ghasemi
- Department of Clinical SciencesFaculty of Veterinary MedicineFerdowsi University of MashhadMashhadIran
| | - Hassan Borji
- Department of PathobiologyFaculty of Veterinary MedicineFerdowsi University of MashhadMashhadIran
| | - Ashkan Hajjafari
- Department of PathobiologyFaculty of Veterinary MedicineIslamic Azad University, Science and Research BranchTehranIran
| |
Collapse
|
41
|
Shakib P, Zivdari M, Khalaf AK, Marzban A, Ganjalikhani-Hakemi M, Parvaneh J, Mahmoudvand H, Cheraghipour K. Nanoparticles as Potent Agents for Treatment of Schistosoma Infections: A Systematic Review. CURRENT THERAPEUTIC RESEARCH 2023; 99:100715. [PMID: 37743882 PMCID: PMC10511339 DOI: 10.1016/j.curtheres.2023.100715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 08/09/2023] [Indexed: 09/26/2023]
Abstract
Background Schistosomiasis is an acute and chronic parasitic disease caused by blood flukes of the genus Schistosoma. The current drugs for treating schistosomiasis are associated with some side effects. Objective The aim of this systematic study was an overview of the treatment of diseases caused by Schistosoma based on nanoparticles. Methods In the present systematic research with keywords "Schistosoma", "parasitism", "anti-Schistosoma activity", "nanoparticles", "metal nanoparticles", "silver nanoparticles", "gold nanoparticles", "polymer nanoparticles", "PLGA nanoparticles", "nanoemulsions", "in vitro", and "in vivo" from five English-language databases, including ScienceDirect, europePMC, PubMed, Scopus, Ovid, and Cochrane were searched from 2000 to 2022 by 2 researchers. Results In the initial search, 250 studies were selected. Based on the inclusion and exclusion criteria, 27 articles were finally selected after removing duplicate, unrelated, and articles containing full text. In present article, the most nanoparticles used against Schistosoma were gold nanoparticles (22%). Conclusions The results indicate the high potential of various nanoparticles, including metal nanoparticles, against Schistosoma. Also, the remarkable anti-schistosomal activity of nanoparticles suggests their use in different fields to eliminate this pathogenic microorganism so that it can be used as an effective candidate in the preparation of anti-schistosomal compounds because these compounds have fewer side effects than chemical drugs. Ther Res Clin Exp. 2023; XX:XXX-XXX).
Collapse
Affiliation(s)
- Pegah Shakib
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Masoomeh Zivdari
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | | | - Abdolrazagh Marzban
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Mazdak Ganjalikhani-Hakemi
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Immunology, Faculty of Medicine, Yedıtepe University, Istanbul, Turkey
| | | | - Hossein Mahmoudvand
- Hepatitis Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Kourosh Cheraghipour
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
- Lorestan Provincial Veterinary Service, Khorramabad, Iran
| |
Collapse
|
42
|
Desai DN, Mahal A, Varshney R, Obaidullah AJ, Gupta B, Mohanty P, Pattnaik P, Mohapatra NC, Mishra S, Kandi V, Rabaan AA, Mohapatra RK. Nanoadjuvants: Promising Bioinspired and Biomimetic Approaches in Vaccine Innovation. ACS OMEGA 2023; 8:27953-27968. [PMID: 37576639 PMCID: PMC10413842 DOI: 10.1021/acsomega.3c02030] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 07/13/2023] [Indexed: 08/15/2023]
Abstract
Adjuvants are the important part of vaccine manufacturing as they elicit the vaccination effect and enhance the durability of the immune response through controlled release. In light of this, nanoadjuvants have shown unique broad spectrum advantages. As nanoparticles (NPs) based vaccines are fast-acting and better in terms of safety and usability parameters as compared to traditional vaccines, they have attracted the attention of researchers. A vaccine nanocarrier is another interesting and promising area for the development of next-generation vaccines for prophylaxis. This review looks at the various nanoadjuvants and their structure-function relationships. It compiles the state-of-art literature on numerous nanoadjuvants to help domain researchers orient their understanding and extend their endeavors in vaccines research and development.
Collapse
Affiliation(s)
- Dhruv N. Desai
- Department
of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Ahmed Mahal
- Department
of Medical Biochemical Analysis, College of Health Technology, Cihan University−Erbil, Erbil, Kurdistan Region, Iraq
| | - Rajat Varshney
- Department
of Veterinary Microbiology, FVAS, Banaras
Hindu University, Mirzapur 231001, India
| | - Ahmad J. Obaidullah
- Department
of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Bhawna Gupta
- School
of Biotechnology, KIIT Deemed-to-be University, Bhubaneswar 751024, Odisha, India
| | - Pratikhya Mohanty
- Bioenergy
Lab, BDTC, School of Biotechnology, KIIT
Deemed-to-be University, Bhubaneswar 751024, Odisha, India
| | | | | | - Snehasish Mishra
- Bioenergy
Lab, BDTC, School of Biotechnology, KIIT
Deemed-to-be University, Bhubaneswar 751024, Odisha, India
| | - Venkataramana Kandi
- Department
of Microbiology, Prathima Institute of Medical
Sciences, Karimnagar 505 417, Telangana, India
| | - Ali A. Rabaan
- Molecular
Diagnostic Laboratory, Johns Hopkins Aramco
Healthcare, Dhahran 31311, Saudi Arabia
- College
of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
- Department
of Public Health and Nutrition, The University
of Haripur, Haripur 22610, Pakistan
| | - Ranjan K. Mohapatra
- Department
of Chemistry, Government College of Engineering, Keonjhar 758002, Odisha, India
| |
Collapse
|
43
|
Bhattacharjee B, Ikbal AMA, Farooqui A, Sahu RK, Ruhi S, Syed A, Miatmoko A, Khan D, Khan J. Superior possibilities and upcoming horizons for nanoscience in COVID-19: noteworthy approach for effective diagnostics and management of SARS-CoV-2 outbreak. CHEMICKE ZVESTI 2023; 77:1-24. [PMID: 37362791 PMCID: PMC10072050 DOI: 10.1007/s11696-023-02795-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 03/18/2023] [Indexed: 04/07/2023]
Abstract
The outbreak of COVID-19 has caused great havoc and affected many parts of the world. It has imposed a great challenge to the medical and health fraternity with its ability to continue mutating and increasing the transmission rate. Some challenges include the availability of current knowledge of active drugs against the virus, mode of delivery of the medicaments, its diagnosis, which are relatively limited and do not suffice for further prognosis. One recently developed drug delivery system called nanoparticles is currently being utilized in combating COVID-19. This article highlights the existing methods for diagnosis of COVID-19 such as computed tomography scan, reverse transcription-polymerase chain reaction, nucleic acid sequencing, immunoassay, point-of-care test, detection from breath, nanotechnology-based bio-sensors, viral antigen detection, microfluidic device, magnetic nanosensor, magnetic resonance platform and internet-of-things biosensors. The latest detection strategy based on nanotechnology, biosensor, is said to produce satisfactory results in recognizing SARS-CoV-2 virus. It also highlights the successes in the research and development of COVID-19 treatments and vaccines that are already in use. In addition, there are a number of nanovaccines and nanomedicines currently in clinical trials that have the potential to target COVID-19.
Collapse
Affiliation(s)
- Bedanta Bhattacharjee
- Girijananda Chowdhury Institute of Pharmaceutical Science, Tezpur, Assam 784501 India
| | - Abu Md Ashif Ikbal
- Department of Pharmaceutical Sciences, Assam University (A Central University), Silchar, 788011 India
| | - Atika Farooqui
- The Deccan College of Medical Sciences, Kanchan Bagh, Hyderabad, Telangana 500058 India
| | - Ram Kumar Sahu
- Department of Pharmaceutical Sciences, Hemvati Nandan Bahuguna Garhwal University (A Central University), Chauras Campus, Tehri Garhwal, Uttarakhand 249161 India
| | - Sakina Ruhi
- Department of Biochemistry, IMS, Management and Science University, University Drive, Off Persiaran Olahraga, 40100 Shah Alam, Selangor Malaysia
| | - Ayesha Syed
- International Medical School, Management and Science University, University Drive, Off Persiaran Olahraga, 40100 Shah Alam, Selangor Malaysia
| | - Andang Miatmoko
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Universitas Airlangga, Surabaya, East Java 60115 Indonesia
| | - Danish Khan
- Panineeya Institute of Dental Science and Research Centre, Kalonji Narayana Rao University of Health Sciences, Warangal, Telangana 506007 India
| | - Jiyauddin Khan
- School of Pharmacy, Management and Science University, 40100 Shah Alam, Selangor Malaysia
| |
Collapse
|
44
|
Abbo SR, Nguyen W, Abma-Henkens MHC, van de Kamer D, Savelkoul NHA, Geertsema C, Le TTT, Tang B, Yan K, Dumenil T, van Oers MM, Suhrbier A, Pijlman GP. Comparative Efficacy of Mayaro Virus-Like Particle Vaccines Produced in Insect or Mammalian Cells. J Virol 2023; 97:e0160122. [PMID: 36883812 PMCID: PMC10062127 DOI: 10.1128/jvi.01601-22] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 02/13/2023] [Indexed: 03/09/2023] Open
Abstract
Mayaro virus (MAYV) is a mosquito-transmitted alphavirus that causes often debilitating rheumatic disease in tropical Central and South America. There are currently no licensed vaccines or antiviral drugs available for MAYV disease. Here, we generated Mayaro virus-like particles (VLPs) using the scalable baculovirus-insect cell expression system. High-level secretion of MAYV VLPs in the culture fluid of Sf9 insect cells was achieved, and particles with a diameter of 64 to 70 nm were obtained after purification. We characterize a C57BL/6J adult wild-type mouse model of MAYV infection and disease and used this model to compare the immunogenicity of VLPs from insect cells with that of VLPs produced in mammalian cells. Mice received two intramuscular immunizations with 1 μg of nonadjuvanted MAYV VLPs. Potent neutralizing antibody responses were generated against the vaccine strain, BeH407, with comparable activity seen against a contemporary 2018 isolate from Brazil (BR-18), whereas neutralizing activity against chikungunya virus was marginal. Sequencing of BR-18 illustrated that this virus segregates with genotype D isolates, whereas MAYV BeH407 belongs to genotype L. The mammalian cell-derived VLPs induced higher mean neutralizing antibody titers than those produced in insect cells. Both VLP vaccines completely protected adult wild-type mice against viremia, myositis, tendonitis, and joint inflammation after MAYV challenge. IMPORTANCE Mayaro virus (MAYV) is associated with acute rheumatic disease that can be debilitating and can evolve into months of chronic arthralgia. MAYV is believed to have the potential to emerge as a tropical public health threat, especially if it develops the ability to be efficiently transmitted by urban mosquito vectors, such as Aedes aegypti and/or Aedes albopictus. Here, we describe a scalable virus-like particle vaccine against MAYV that induced neutralizing antibodies against a historical and a contemporary isolate of MAYV and protected mice against infection and disease, providing a potential new intervention for MAYV epidemic preparedness.
Collapse
Affiliation(s)
- Sandra R. Abbo
- Laboratory of Virology, Wageningen University & Research, Wageningen, the Netherlands
| | - Wilson Nguyen
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | | | - Denise van de Kamer
- Laboratory of Virology, Wageningen University & Research, Wageningen, the Netherlands
| | - Niek H. A. Savelkoul
- Laboratory of Virology, Wageningen University & Research, Wageningen, the Netherlands
| | - Corinne Geertsema
- Laboratory of Virology, Wageningen University & Research, Wageningen, the Netherlands
| | - Thuy T. T. Le
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Bing Tang
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Kexin Yan
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Troy Dumenil
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Monique M. van Oers
- Laboratory of Virology, Wageningen University & Research, Wageningen, the Netherlands
| | - Andreas Suhrbier
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- GVN Center of Excellence, Australian Infectious Disease Research Center, Brisbane, Queensland, Australia
| | - Gorben P. Pijlman
- Laboratory of Virology, Wageningen University & Research, Wageningen, the Netherlands
| |
Collapse
|
45
|
Karunakaran B, Gupta R, Patel P, Salave S, Sharma A, Desai D, Benival D, Kommineni N. Emerging Trends in Lipid-Based Vaccine Delivery: A Special Focus on Developmental Strategies, Fabrication Methods, and Applications. Vaccines (Basel) 2023; 11:661. [PMID: 36992244 PMCID: PMC10051624 DOI: 10.3390/vaccines11030661] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/09/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023] Open
Abstract
Lipid-based vaccine delivery systems such as the conventional liposomes, virosomes, bilosomes, vesosomes, pH-fusogenic liposomes, transferosomes, immuno-liposomes, ethosomes, and lipid nanoparticles have gained a remarkable interest in vaccine delivery due to their ability to render antigens in vesicular structures, that in turn prevents its enzymatic degradation in vivo. The particulate form of lipid-based nanocarriers confers immunostimulatory potential, making them ideal antigen carriers. Facilitation in the uptake of antigen-loaded nanocarriers, by the antigen-presenting cells and its subsequent presentation through the major histocompatibility complex molecules, leads to the activation of a cascade of immune responses. Further, such nanocarriers can be tailored to achieve the desired characteristics such as charge, size, size distribution, entrapment, and site-specificity through modifications in the composition of lipids and the selection of the appropriate method of preparation. This ultimately adds to its versatility as an effective vaccine delivery carrier. The current review focuses on the various lipid-based carriers that have been investigated to date as potential vaccine delivery systems, the factors that affect their efficacy, and their various methods of preparation. The emerging trends in lipid-based mRNA vaccines and lipid-based DNA vaccines have also been summarized.
Collapse
Affiliation(s)
- Bharathi Karunakaran
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad 382355, India
| | - Raghav Gupta
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad 382355, India
| | - Pranav Patel
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad 382355, India
| | - Sagar Salave
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad 382355, India
| | - Amit Sharma
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad 382355, India
| | - Dhruv Desai
- School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Derajram Benival
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad 382355, India
| | | |
Collapse
|
46
|
Qu Y, De Rose R, Kim C, Zhou J, Lin Z, Ju Y, Bhangu SK, Cortez‐Jugo C, Cavalieri F, Caruso F. Supramolecular Polyphenol-DNA Microparticles for In Vivo Adjuvant and Antigen Co-Delivery and Immune Stimulation. Angew Chem Int Ed Engl 2023; 62:e202214935. [PMID: 36700351 PMCID: PMC10946467 DOI: 10.1002/anie.202214935] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 01/25/2023] [Accepted: 01/25/2023] [Indexed: 01/27/2023]
Abstract
DNA-based materials have attracted interest due to the tunable structure and encoded biological functionality of nucleic acids. A simple and general approach to synthesize DNA-based materials with fine control over morphology and bioactivity is important to expand their applications. Here, we report the synthesis of DNA-based particles via the supramolecular assembly of tannic acid (TA) and DNA. Uniform particles with different morphologies are obtained using a variety of DNA building blocks. The particles enable the co-delivery of cytosine-guanine adjuvant sequences and the antigen ovalbumin in model cells. Intramuscular injection of the particles in mice induces antigen-specific antibody production and T cell responses with no apparent toxicity. Protein expression in cells is shown using capsules assembled from TA and plasmid DNA. This work highlights the potential of TA as a universal material for directing the supramolecular assembly of DNA into gene and vaccine delivery platforms.
Collapse
Affiliation(s)
- Yijiao Qu
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| | - Robert De Rose
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| | - Chan‐Jin Kim
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| | - Jiajing Zhou
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| | - Zhixing Lin
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| | - Yi Ju
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| | - Sukhvir Kaur Bhangu
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
- School of ScienceRMIT UniversityMelbourneVictoria3000Australia
| | - Christina Cortez‐Jugo
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| | - Francesca Cavalieri
- School of ScienceRMIT UniversityMelbourneVictoria3000Australia
- Dipartimento di Scienze e Tecnologie Chimiche Universita' di Roma “Tor Vergata”Via della Ricerca Scientifica 100133RomeItaly
| | - Frank Caruso
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| |
Collapse
|
47
|
Hashemi Goradel N, Nemati M, Bakhshandeh A, Arashkia A, Negahdari B. Nanovaccines for cancer immunotherapy: Focusing on complex formation between adjuvant and antigen. Int Immunopharmacol 2023; 117:109887. [PMID: 36841155 DOI: 10.1016/j.intimp.2023.109887] [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: 11/27/2022] [Revised: 01/29/2023] [Accepted: 02/10/2023] [Indexed: 02/27/2023]
Abstract
As an interesting cancer immunotherapy approach, cancer vaccines have been developed to deliver tumor antigens and adjuvants to antigen-presenting cells (APCs). Although the safety and easy production shifted the vaccine designing platforms toward the subunit vaccines, their efficacy is limited due to inefficient vaccine delivery. Nanotechnology-based vaccines, called nanovaccines, address the delivery limitations through co-delivery of antigens and adjuvants into lymphoid organs and APCs and their intracellular release, leading to cross-presentation of antigens and induction of potent anti-tumor immune responses. Although the nanovaccines, either as encapsulating agents or biomimetic nanoparticles, exert the desired anti-tumor activities, there is evidence that the mixing formulation to form nanocomplexes between antigens and adjuvants based on the electrostatic interactions provokes high levels of immune responses owing to Ags' availability and faster release. Here, we summarized the various platforms for developing cancer vaccines and the advantages of using delivery systems. The cancer nanovaccines, including nanoparticle-based and biomimetic-based nanovaccines, are discussed in detail. Finally, we focused on the nanocomplexes formation between antigens and adjuvants as promising cancer nanovaccine platforms.
Collapse
Affiliation(s)
- Nasser Hashemi Goradel
- Department of Medical Biotechnology, Maragheh University of Medical Sciences, Maragheh, Iran.
| | - Mahnaz Nemati
- Amir Oncology Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Azam Bakhshandeh
- Department of Industrial Engineering and Management Systems, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Arash Arashkia
- Department of Molecular Virology, Pasteur Institute of Iran, Tehran, Iran
| | - Babak Negahdari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
48
|
Yoshioka Y, Kobiyama K, Hayashi T, Onishi M, Yanagida Y, Nakagawa T, Hashimoto M, Nishinaka A, Hirose J, Asaoka Y, Tajiri M, Hayata A, Ishida S, Omoto S, Nagira M, Ishii KJ. A-910823, a squalene-based emulsion adjuvant, induces T follicular helper cells and humoral immune responses via α-tocopherol component. Front Immunol 2023; 14:1116238. [PMID: 36891311 PMCID: PMC9986537 DOI: 10.3389/fimmu.2023.1116238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 02/03/2023] [Indexed: 02/22/2023] Open
Abstract
Background Adjuvants are chemical or biological materials that enhance the efficacy of vaccines. A-910823 is a squalene-based emulsion adjuvant used for S-268019-b, a novel vaccine against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that is currently in clinical development. Published evidence has demonstrated that A-910823 can enhance the induction of neutralizing antibodies against SARS-CoV-2 in humans and animal models. However, the characteristics and mechanisms of the immune responses induced by A-910823 are not yet known. Methods and Results To characterize A-910823, we compared the adaptive immune response profile enhanced by A-910823 with that of other adjuvants (AddaVax, QS21, aluminum salt-based adjuvants, and empty lipid nanoparticle [eLNP]) in a murine model. Compared with other adjuvants, A-910823 enhanced humoral immune responses to an equal or greater extent following potent T follicular helper (Tfh) and germinal center B (GCB) cell induction, without inducing a strong systemic inflammatory cytokine response. Furthermore, S-268019-b containing A-910823 adjuvant produced similar results even when given as a booster dose following primary administration of a lipid nanoparticle-encapsulated messenger RNA (mRNA-LNP) vaccine. Preparation of modified A-910823 adjuvants to identify which components of A-910823 play a role in driving the adjuvant effect and detailed evaluation of the immunological characteristics induced by each adjuvant showed that the induction of humoral immunity and Tfh and GCB cell induction in A-910823 were dependent on α-tocopherol. Finally, we revealed that the recruitment of inflammatory cells to the draining lymph nodes and induction of serum cytokines and chemokines by A-910823 were also dependent on the α-tocopherol component. Conclusions This study demonstrates that the novel adjuvant A-910823 is capable of robust Tfh cell induction and humoral immune responses, even when given as a booster dose. The findings also emphasize that α-tocopherol drives the potent Tfh-inducing adjuvant function of A-910823. Overall, our data provide key information that may inform the future production of improved adjuvants.
Collapse
Affiliation(s)
- Yuya Yoshioka
- Laboratory for Bio-Drug Discovery, Shionogi & Co., Ltd., Osaka, Japan
| | - Kouji Kobiyama
- Division of Vaccine Science, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Tomoya Hayashi
- Division of Vaccine Science, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Motoyasu Onishi
- Laboratory for Bio-Drug Discovery, Shionogi & Co., Ltd., Osaka, Japan
| | - Yosuke Yanagida
- Formulation R&D Laboratory, Shionogi & Co., Ltd., Osaka, Japan
| | - Takayuki Nakagawa
- Laboratory for Bio-Drug Discovery, Shionogi & Co., Ltd., Osaka, Japan
| | | | - Anri Nishinaka
- Laboratory for Bio-Drug Discovery, Shionogi & Co., Ltd., Osaka, Japan
| | - Jun Hirose
- Formulation R&D Laboratory, Shionogi & Co., Ltd., Osaka, Japan
| | - Yoshiji Asaoka
- Laboratory for Drug Discovery and Development, Shionogi & Co., Ltd., Osaka, Japan
| | - Minako Tajiri
- Laboratory for Drug Discovery and Development, Shionogi & Co., Ltd., Osaka, Japan
| | - Atsushi Hayata
- Laboratory for Bio-Modality Research, Shionogi & Co., Osaka, Japan
| | - Satoru Ishida
- Laboratory for Bio-Drug Discovery, Shionogi & Co., Ltd., Osaka, Japan
| | - Shinya Omoto
- Laboratory for Bio-Drug Discovery, Shionogi & Co., Ltd., Osaka, Japan
| | - Morio Nagira
- Laboratory for Bio-Drug Discovery, Shionogi & Co., Ltd., Osaka, Japan
| | - Ken J. Ishii
- Division of Vaccine Science, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Vaccine and Adjuvant Research Center (CVAR), National Institute of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
- Laboratory of Vaccine Science, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| |
Collapse
|
49
|
Higuchi A, Morishita M, Nagata R, Maruoka K, Katsumi H, Yamamoto A. Functional Characterization of Extracellular Vesicles from Baker's Yeast Saccharomyces Cerevisiae as a Novel Vaccine Material for Immune Cell Maturation. J Pharm Sci 2023; 112:525-534. [PMID: 36057319 DOI: 10.1016/j.xphs.2022.08.032] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 08/26/2022] [Accepted: 08/26/2022] [Indexed: 01/18/2023]
Abstract
Extracellular vesicles (EVs) encapsulate various bioactive molecules, and much effort has been directed towards developing a novel EV-based therapy. Although recent studies reported the secretion of EVs from probiotics baker's yeast Saccharomyces cerevisiae (S. cerevisiae), their properties and functions remain obscure. The aim of this study was to clarify the usefulness of EVs from S. cerevisiae (S-EVs) as a novel vaccine material by defining their physicochemical properties and biological functions. The collected S-EVs contained β-D-glucan and showed particle sizes and zeta potentials approximately 128.8 nm and -7.39 mV, respectively. S-EVs were positive for heat shock protein 70 kDa (HSP70). These S-EVs considerably enhanced the production of proinflammatory tumor necrosis factor-α and interleukin 6 from RAW264.7 cells (mouse macrophage-like cells) and DC2.4 cells (mouse dendritic cells). The expression of maturation markers CD40, CD80 and CD86 on the surface of these immune cells incubated with S-EVs was remarkably upregulated. Immune cells endocytosed S-EVs, and toll like receptor 2 on immune cells was involved in immune activation by S-EVs. These results indicate that extracellular vesicles derived from baker's yeast Saccharomyces cerevisiae are an attractive source as a novel vaccine material for immune cells maturation.
Collapse
Affiliation(s)
- Ayaka Higuchi
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| | - Masaki Morishita
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan.
| | - Ryoga Nagata
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| | - Kento Maruoka
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| | - Hidemasa Katsumi
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| | - Akira Yamamoto
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| |
Collapse
|
50
|
Bjanes E, Zhou J, Qayum T, Krishnan N, Zurich RH, Menon ND, Hoffman A, Fang RH, Zhang L, Nizet V. Outer Membrane Vesicle-Coated Nanoparticle Vaccine Protects Against Acinetobacter baumannii Pneumonia and Sepsis. ADVANCED NANOBIOMED RESEARCH 2023; 3:2200130. [PMID: 37151210 PMCID: PMC10156090 DOI: 10.1002/anbr.202200130] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The highly multidrug-resistant (MDR) Gram-negative bacterial pathogen Acinetobacter baumannii is a top global health priority where an effective vaccine could protect susceptible populations and limit resistance acquisition. Outer membrane vesicles (OMVs) shed from Gram-negative bacteria are enriched with virulence factors and membrane lipids but heterogeneous in size and cargo. We report a vaccine platform combining precise and replicable nanoparticle technology with immunogenic A. baumannii OMVs (Ab-OMVs). Gold nanoparticle cores coated with Ab-OMVs (Ab-NPs) induced robust IgG titers in rabbits that enhanced human neutrophil opsonophagocytic killing and passively protected against lethal A. baumannii sepsis in mice. Active Ab-NP immunization in mice protected against sepsis and pneumonia, accompanied by B cell recruitment to draining lymph nodes, activation of dendritic cell markers, improved splenic neutrophil responses, and mitigation of proinflammatory cytokine storm. Nanoparticles are an efficient and efficacious platform for OMV vaccine delivery against A. baumannii and perhaps other high-priority MDR pathogens.
Collapse
Affiliation(s)
- Elisabet Bjanes
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, California 92093, USA
| | - Jiarong Zhou
- Department of Nanoengineering, University of California San Diego, La Jolla, California 92093, USA
| | - Tariq Qayum
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, California 92093, USA
| | - Nishta Krishnan
- Department of Nanoengineering, University of California San Diego, La Jolla, California 92093, USA
| | - Raymond H. Zurich
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, California 92093, USA
| | - Nitasha D. Menon
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, California 92093, USA
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Kerala, India
| | - Alexandria Hoffman
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, California 92093, USA
| | - Ronnie H. Fang
- Department of Nanoengineering, University of California San Diego, La Jolla, California 92093, USA
| | - Liangfang Zhang
- Department of Nanoengineering, University of California San Diego, La Jolla, California 92093, USA
| | - Victor Nizet
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, California 92093, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California 92093, USA
| |
Collapse
|