1
|
Khan MA, Mousa AM, Alradhi AE, Allemailem K. Efficacy of lipid nanoparticles-based vaccine to protect against vulvovaginal candidiasis (VVC): Implications for women's reproductive health. Life Sci 2024; 361:123312. [PMID: 39674269 DOI: 10.1016/j.lfs.2024.123312] [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: 09/30/2024] [Revised: 12/03/2024] [Accepted: 12/11/2024] [Indexed: 12/16/2024]
Abstract
AIMS Vulvovaginal candidiasis (VVC) is a common women's health issue, with rising antifungal resistance. This study was aimed to prepare and evaluate the efficacy of a lipid nanoparticle-based vaccine in a murine model of VVC. MATERIALS AND METHODS Dried and reconstituted vesicles containing C. albicans antigens (DRNPs-Ca-Ags) vaccine, formulated with phosphatidylcholine and cholesterol-based lipid nanoparticles via film hydration and freeze-drying. The safety evaluation of DRNPs-CaAgs was conducted by determining hepatic (AST, ALT) or renal (BUN, creatinine) biomarkers. Female mice were immunized with DRNPs-CaAgs or Alum-CaAgs, and immune responses were evaluated via antibody titers, IgG isotypes, and splenocyte proliferation. Protective efficacy of vaccine formulations was assessed through fungal burden, biofilm formation, cytokine levels, and histopathological analysis of vaginal tissues. KEY FINDINGS Mice vaccinated with DRNPs-CaAgs showed significantly enhanced immune responses, with higher antibody titers and IgG2a levels as compared to the Alum-CaAgs group. Vaginal fungal burden was dramatically reduced (665 ± 78 CFUs in DRNPs-CaAgs immunized group vs. 12,944 ± 3540 CFUs in Alum-CaAgs group, p < 0.01). Biofilm formation decreased by 45 % (p < 0.05), and inflammatory cytokines were significantly lowered. Histopathological analysis revealed minimal tissue damage in DRNPs-CaAgs vaccinated mice. SIGNIFICANCE The findings suggest DRNPs-CaAgs as a promising vaccine for VVC, eliciting strong immunity, reducing fungal load, and minimizing inflammation. While the reliance on a murine model is a limitation, future clinical trials are essential to evaluate its efficacy and safety in humans, offering a potential strategy to combat drug-resistant infections and improve women's reproductive health.
Collapse
Affiliation(s)
- Masood Alam Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraidah 51452, Saudi Arabia.
| | - Ayman M Mousa
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraidah 51452, Saudi Arabia
| | - Arwa Essa Alradhi
- General Administration for Infectious Disease Control, Ministry of Health, Riyadh 12382, Saudi Arabia
| | - Khaled Allemailem
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraidah 51452, Saudi Arabia
| |
Collapse
|
2
|
Liu Y, Yin R, Tian Y, Xu S, Meng X. Curcumin nanopreparations: recent advance in preparation and application. Biomed Mater 2024; 19:052009. [PMID: 39189065 DOI: 10.1088/1748-605x/ad6dc7] [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: 04/15/2024] [Accepted: 08/09/2024] [Indexed: 08/28/2024]
Abstract
Curcumin is a natural polyphenolic compound extracted from turmeric with antibacterial, antioxidant, antitumor, preventive and therapeutic neurological disorders and a variety of bioactivities, which is widely used in the field of food and medicine. However, the drawbacks of curcumin such as poor aqueous solubility and stability have limited the practical application of curcumin. To overcome these defects and enhance its functional properties, various nanoscale systems (liposomes, polymer nanoparticles, protein nanoparticles, solid lipid nanoparticles, metal nanoparticles, etc) have been extensively employed for curcumin encapsulation and delivery. Despite the rapid development of curcumin nanoformulations, there is a lack of comprehensive reviews on their preparation and properties. This review provides an overview of the construction of curcumin nano-delivery systems, mechanisms of action, nanocarrier preparation methods and the applications of curcumin nanocarriers in the food and pharmaceutical fields to provide a theoretical basis and technological support for the efficient bio-utilization, product development and early clinical application of curcumin.
Collapse
Affiliation(s)
- Yan Liu
- School of Pharmacy, Heilongjiang University of Chinese Medicine, NO.24 Heping Road, Harbin, 150040, People's Republic of China
| | - Rui Yin
- School of Pharmacy, Heilongjiang University of Chinese Medicine, NO.24 Heping Road, Harbin, 150040, People's Republic of China
| | - Yuan Tian
- School of Pharmacy, Heilongjiang University of Chinese Medicine, NO.24 Heping Road, Harbin, 150040, People's Republic of China
| | - Shujun Xu
- School of Pharmacy, Heilongjiang University of Chinese Medicine, NO.24 Heping Road, Harbin, 150040, People's Republic of China
| | - Xin Meng
- School of Pharmacy, Heilongjiang University of Chinese Medicine, NO.24 Heping Road, Harbin, 150040, People's Republic of China
| |
Collapse
|
3
|
Zhang L, Li Y, Liu X, He X, Zhang J, Zhou J, Qiao Y, Wu H, Sun F, Zhou Q. Optimal development of apoptotic cells-mimicking liposomes targeting macrophages. J Nanobiotechnology 2024; 22:501. [PMID: 39169328 PMCID: PMC11337832 DOI: 10.1186/s12951-024-02755-3] [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: 03/12/2024] [Accepted: 08/05/2024] [Indexed: 08/23/2024] Open
Abstract
Macrophages are multifunctional innate immune cells that play indispensable roles in homeostasis, tissue repair, and immune regulation. However, dysregulated activation of macrophages is implicated in the pathogenesis of various human disorders, making them a potential target for treatment. Through the expression of pattern recognition and scavenger receptors, macrophages exhibit selective uptake of pathogens and apoptotic cells. Consequently, the utilization of drug carriers that mimic pathogenic or apoptotic signals shows potential for targeted delivery to macrophages. In this study, a series of mannosylated or/and phosphatidylserine (PS) -presenting liposomes were developed to target macrophages via the design of experiment (DoE) strategy and the trial-and-error (TaE) approach. The optimal molar ratio for the liposome formulation was DOPC: DSPS: Chol: PEG-PE = 20:60:20:2 based on the results of cellular uptake and cytotoxicity evaluation on RAW 264.7 and THP-1 in vitro. Results from in vivo distribution showed that, in the DSS-induced colitis model and collagen II-induced rheumatoid arthritis model, PS-presenting liposomes (PS-Lipo) showed the highest accumulation in intestine and paws respectively, which holds promising potential for macrophage target therapy since macrophages are abundant at inflammatory sites and contribute to the progression of corresponding diseases. Organs such as the heart, liver, spleen, lung, and kidney did not exhibit histological alterations such as inflammation or necrosis when exposed to PC-presenting liposomes (PC-Lipo) or PS-Lipo. In addition, liposomes demonstrated hemobiocompatibility and no toxicity to liver or kidney for circulation and did not induce metabolic injury in the animals. Thus, the well-designed PS-Lipo demonstrated the most potential for macrophage target therapy.
Collapse
Affiliation(s)
- Li Zhang
- Department of Prosthodontics, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School,Institute of Stomatology,Nanjing University, Nanjing, 210002, China
| | - Yujiao Li
- Department of Clinical Pharmacy, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Xing Liu
- Department of Infectious Disease and Liver Disease, the Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, 210003, China
| | - Xiaolu He
- Department of Clinical Pharmacy, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Jieyu Zhang
- Department of Clinical Pharmacy, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Jun Zhou
- Department of Clinical Pharmacy, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Youbei Qiao
- Department of Medicinal Chemistry and Pharmaceutical Analysis, School of Pharmacy, Air Force Medical University, Xi'an, 710032, China
| | - Hong Wu
- Department of Medicinal Chemistry and Pharmaceutical Analysis, School of Pharmacy, Air Force Medical University, Xi'an, 710032, China.
| | - Fangfang Sun
- Department of Prosthodontics, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School,Institute of Stomatology,Nanjing University, Nanjing, 210002, China.
| | - Qing Zhou
- Department of Clinical Pharmacy, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China.
| |
Collapse
|
4
|
Xu K, Zhang Q, Zhu D, Jiang Z. Hydrogels in Gene Delivery Techniques for Regenerative Medicine and Tissue Engineering. Macromol Biosci 2024; 24:e2300577. [PMID: 38265144 DOI: 10.1002/mabi.202300577] [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: 12/16/2023] [Revised: 01/16/2024] [Indexed: 01/25/2024]
Abstract
Hydrogels are 3D networks swollen with water. They are biocompatible, strong, and moldable and are emerging as a promising biomedical material for regenerative medicine and tissue engineering to deliver therapeutic genes. The excellent natural extracellular matrix simulation properties of hydrogels enable them to be co-cultured with cells or enhance the expression of viral or non-viral vectors. Its biocompatibility, high strength, and degradation performance also make the action process of carriers in tissues more ideal, making it an ideal biomedical material. It has been shown that hydrogel-based gene delivery technologies have the potential to play therapy-relevant roles in organs such as bone, cartilage, nerve, skin, reproductive organs, and liver in animal experiments and preclinical trials. This paper reviews recent articles on hydrogels in gene delivery and explains the manufacture, applications, developmental timeline, limitations, and future directions of hydrogel-based gene delivery techniques.
Collapse
Affiliation(s)
- Kexing Xu
- Zhejiang University School of Medicine, Hangzhou, China
| | - Qinmeng Zhang
- Zhejiang University School of Medicine, Hangzhou, China
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Danji Zhu
- Zhejiang University School of Medicine, Hangzhou, China
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Zhiwei Jiang
- Zhejiang University School of Medicine, Hangzhou, China
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| |
Collapse
|
5
|
Zachová K, Bartheldyová E, Hubatka F, Křupka M, Odehnalová N, Turánek Knötigová P, Vaškovicová N, Sloupenská K, Hromádka R, Paulovičová E, Effenberg R, Ledvina M, Raška M, Turánek J. The immunogenicity of p24 protein from HIV-1 virus is strongly supported and modulated by coupling with liposomes and mannan. Carbohydr Polym 2024; 332:121844. [PMID: 38431385 DOI: 10.1016/j.carbpol.2024.121844] [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: 09/14/2023] [Revised: 12/31/2023] [Accepted: 01/18/2024] [Indexed: 03/05/2024]
Abstract
Anti-viral and anti-tumor vaccines aim to induce cytotoxic CD8+ T cells (CTL) and antibodies. Conserved protein antigens, such as p24 from human immunodeficiency virus, represent promising component for elicitation CTLs, nevertheless with suboptimal immunogenicity, if formulated as recombinant protein. To enhance immunogenicity and CTL response, recombinant proteins may be targeted to dendritic cells (DC) for cross presentation on MHCI, where mannose receptor and/or other lectin receptors could play an important role. Here, we constructed liposomal carrier-based vaccine composed of recombinant p24 antigen bound by metallochelating linkage onto surface of nanoliposomes with surface mannans coupled by aminooxy ligation. Generated mannosylated proteonanoliposomes were analyzed by dynamic light scattering, isothermal titration, and electron microscopy. Using murine DC line MutuDC and murine bone marrow derived DC (BMDC) we evaluated their immunogenicity and immunomodulatory activity. We show that p24 mannosylated proteonanoliposomes activate DC for enhanced MHCI, MHCII and CD40, CD80, and CD86 surface expression both on MutuDC and BMDC. p24 mannosylated liposomes were internalized by MutuDC with p24 intracellular localization within 1 to 3 h. The combination of metallochelating and aminooxy ligation could be used simultaneously to generate nanoliposomal adjuvanted recombinant protein-based vaccines versatile for combination of recombinant antigens relevant for antibody and CTL elicitation.
Collapse
Affiliation(s)
- K Zachová
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Hněvotínská 3, Olomouc, Czech Republic
| | - E Bartheldyová
- C2P NEXARS, The Campus Science Park, Palachovo náměstí 2, Brno, Czech Republic
| | - F Hubatka
- C2P NEXARS, The Campus Science Park, Palachovo náměstí 2, Brno, Czech Republic
| | - M Křupka
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Hněvotínská 3, Olomouc, Czech Republic
| | - N Odehnalová
- C2P NEXARS, The Campus Science Park, Palachovo náměstí 2, Brno, Czech Republic
| | - P Turánek Knötigová
- C2P NEXARS, The Campus Science Park, Palachovo náměstí 2, Brno, Czech Republic
| | - N Vaškovicová
- Faculty of Medicine, Masaryk University, Kamenice 5, Brno, Czech Republic
| | - K Sloupenská
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Hněvotínská 3, Olomouc, Czech Republic
| | - R Hromádka
- C2P NEXARS, The Campus Science Park, Palachovo náměstí 2, Brno, Czech Republic
| | - E Paulovičová
- Center for Glycomics, Institute of Chemistry, Slovak Academy of Sciences, Dúbravská cesta 9, Bratislava, Slovakia
| | - R Effenberg
- Department of Chemistry of Natural Compounds, University of Chemistry and Technology, Technická 5, Prague, Czech Republic
| | - M Ledvina
- Department of Chemistry of Natural Compounds, University of Chemistry and Technology, Technická 5, Prague, Czech Republic
| | - M Raška
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Hněvotínská 3, Olomouc, Czech Republic.
| | - J Turánek
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Hněvotínská 3, Olomouc, Czech Republic; C2P NEXARS, The Campus Science Park, Palachovo náměstí 2, Brno, Czech Republic; Institute of Clinical Immunology & Allergology, Charles University Prague and University Hospital, Hradec Kralove, Sokolská 581, Hradec Kralove, Czech Republic.
| |
Collapse
|
6
|
Cai B, Thomas R. Dendritic cells and antigen-specific immunotherapy in autoimmune rheumatic diseases. Best Pract Res Clin Rheumatol 2024; 38:101940. [PMID: 38485600 DOI: 10.1016/j.berh.2024.101940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 02/29/2024] [Indexed: 09/02/2024]
Abstract
Dendritic cells (DCs) are professional antigen-presenting cells and trigger downstream immune responses to antigen while integrating cellular pathogen and damage-associated molecular pattern (PAMP and DAMP) or immunomodulatory signals. In healthy individuals, resting and tolerogenic DCs draining skin and intestine facilitate expansion of regulatory T cells (Treg) to maintain peripheral antigen-specific immune tolerance. In patients with rheumatic diseases, however, DCs activated by PAMPs and DAMPs expand self-reactive effector T cells, including follicular helper T cells that promote the expansion of activated autoreactive B cells, chronic inflammation and end-organ damage. With the development of cellular and nanoparticle (NP)-based self-antigen-specific immunotherapies we here consider the new opportunities and the challenges for restoring immunoregulation in the treatment and prevention of autoimmune inflammatory rheumatic conditions through DCs.
Collapse
Affiliation(s)
- Benjamin Cai
- Frazer Institute, The University of Queensland, Brisbane, Queensland, Australia.
| | - Ranjeny Thomas
- Frazer Institute, The University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
7
|
Koornneef A, Vanshylla K, Hardenberg G, Rutten L, Strokappe NM, Tolboom J, Vreugdenhil J, Boer KFD, Perkasa A, Blokland S, Burger JA, Huang WC, Lovell JF, van Manen D, Sanders RW, Zahn RC, Schuitemaker H, Langedijk JPM, Wegmann F. CoPoP liposomes displaying stabilized clade C HIV-1 Env elicit tier 2 multiclade neutralization in rabbits. Nat Commun 2024; 15:3128. [PMID: 38605096 PMCID: PMC11009251 DOI: 10.1038/s41467-024-47492-1] [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] [Accepted: 03/27/2024] [Indexed: 04/13/2024] Open
Abstract
One of the strategies towards an effective HIV-1 vaccine is to elicit broadly neutralizing antibody responses that target the high HIV-1 Env diversity. Here, we present an HIV-1 vaccine candidate that consists of cobalt porphyrin-phospholipid (CoPoP) liposomes decorated with repaired and stabilized clade C HIV-1 Env trimers in a prefusion conformation. These particles exhibit high HIV-1 Env trimer decoration, serum stability and bind broadly neutralizing antibodies. Three sequential immunizations of female rabbits with CoPoP liposomes displaying a different clade C HIV-1 gp140 trimer at each dosing generate high HIV-1 Env-specific antibody responses. Additionally, serum neutralization is detectable against 18 of 20 multiclade tier 2 HIV-1 strains. Furthermore, the peak antibody titers induced by CoPoP liposomes can be recalled by subsequent heterologous immunization with Ad26-encoded membrane-bound stabilized Env antigens. Hence, a CoPoP liposome-based HIV-1 vaccine that can generate cross-clade neutralizing antibody immunity could potentially be a component of an efficacious HIV-1 vaccine.
Collapse
Affiliation(s)
| | | | | | - Lucy Rutten
- Janssen Vaccines & Prevention, Leiden, The Netherlands
| | | | | | | | | | | | - Sven Blokland
- Janssen Vaccines & Prevention, Leiden, The Netherlands
| | - Judith A Burger
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Wei-Chiao Huang
- Department of Biomedical Engineering, University at Buffalo, Buffalo, NY, USA
| | - Jonathan F Lovell
- Department of Biomedical Engineering, University at Buffalo, Buffalo, NY, USA
| | | | - Rogier W Sanders
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Roland C Zahn
- Janssen Vaccines & Prevention, Leiden, The Netherlands
| | | | - Johannes P M Langedijk
- Janssen Vaccines & Prevention, Leiden, The Netherlands.
- ForgeBio, Amsterdam, The Netherlands.
| | - Frank Wegmann
- Janssen Vaccines & Prevention, Leiden, The Netherlands.
| |
Collapse
|
8
|
Rattan A, Malemnganba T, Sagar, Prajapati VK. Exploring structural engineering approach to formulate and characterize next-generation adjuvants. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 140:59-90. [PMID: 38762280 DOI: 10.1016/bs.apcsb.2023.12.020] [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: 05/20/2024]
Abstract
It is critical to emphasize the importance of vaccination as it protects us against harmful pathogens. Despite significant progress in vaccine development, there is an ongoing need to develop vaccines that are not only safe but also highly effective in protecting against severe infections. Subunit vaccines are generally safe, but they frequently fail to elicit strong immune responses. As a result, there is a need to improve vaccine effectiveness by combining them with adjuvants, which have the potential to boost the immune system many folds. The process of developing these adjuvants requires searching for molecules capable of activating the immune system, combining these promising compounds with an antigen, and then testing this combination using animal models before approving it for clinical use. Liposomal adjuvants work as delivery adjuvants and its activity depends on certain parameters such as surface charge, vesicle size, surface modification and route of administration. Self-assembly property of peptide adjuvants and discovery of hybrid peptides have widened the scope of peptides in vaccine formulations. Since most pathogenic molecules are not peptide based, phage display technique allows for screening peptide mimics for such pathogens that have potential as adjuvants. This chapter discusses about peptide and liposome-based adjuvants focusing on their properties imparting adjuvanticity along with the methods of formulating them. Methods of adjuvant characterization important for an adjuvant to be approved for clinical trials are also discussed. These include assays for cytotoxicity, T-lymphocyte proliferation, dendritic cell maturation, cytokine and antibody production, toll-like receptor dependent signaling and adjuvant half-life.
Collapse
Affiliation(s)
- Aditi Rattan
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India
| | - Takhellambam Malemnganba
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India
| | - Sagar
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India.
| |
Collapse
|
9
|
Sun Z, Zhao H, Ma L, Shi Y, Ji M, Sun X, Ma D, Zhou W, Huang T, Zhang D. The quest for nanoparticle-powered vaccines in cancer immunotherapy. J Nanobiotechnology 2024; 22:61. [PMID: 38355548 PMCID: PMC10865557 DOI: 10.1186/s12951-024-02311-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 01/26/2024] [Indexed: 02/16/2024] Open
Abstract
Despite recent advancements in cancer treatment, this disease still poses a serious threat to public health. Vaccines play an important role in preventing illness by preparing the body's adaptive and innate immune responses to combat diseases. As our understanding of malignancies and their connection to the immune system improves, there has been a growing interest in priming the immune system to fight malignancies more effectively and comprehensively. One promising approach involves utilizing nanoparticle systems for antigen delivery, which has been shown to potentiate immune responses as vaccines and/or adjuvants. In this review, we comprehensively summarized the immunological mechanisms of cancer vaccines while focusing specifically on the recent applications of various types of nanoparticles in the field of cancer immunotherapy. By exploring these recent breakthroughs, we hope to identify significant challenges and obstacles in making nanoparticle-based vaccines and adjuvants feasible for clinical application. This review serves to assess recent breakthroughs in nanoparticle-based cancer vaccinations and shed light on their prospects and potential barriers. By doing so, we aim to inspire future immunotherapies for cancer that harness the potential of nanotechnology to deliver more effective and targeted treatments.
Collapse
Affiliation(s)
- Zhe Sun
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Hui Zhao
- Department of Endodontics, East Branch of Jinan Stomatological Hospital, Jinan, 250000, Shandong, China
| | - Li Ma
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Yanli Shi
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Mei Ji
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Xiaodong Sun
- Department of Endodontics, Gaoxin Branch of Jinan Stomatological Hospital, Jinan, 250000, Shandong, China
| | - Dan Ma
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Wei Zhou
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Tao Huang
- Department of Biomedical Engineering, Graeme Clark Institute, The University of Melbourne, Parkville, VIC, 3010, Australia.
| | - Dongsheng Zhang
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| |
Collapse
|
10
|
Wang H, Wang S, Fang R, Li X, Xing J, Li Z, Song N. Enhancing TB Vaccine Efficacy: Current Progress on Vaccines, Adjuvants and Immunization Strategies. Vaccines (Basel) 2023; 12:38. [PMID: 38250851 PMCID: PMC10820143 DOI: 10.3390/vaccines12010038] [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: 11/11/2023] [Revised: 12/27/2023] [Accepted: 12/27/2023] [Indexed: 01/23/2024] Open
Abstract
Tuberculosis (TB) remains a global infectious disease primarily transmitted via respiratory tract infection. Presently, vaccination stands as the primary method for TB prevention, predominantly reliant on the Bacillus Calmette-Guérin (BCG) vaccine. Although it is effective in preventing disseminated diseases in children, its impact on adults is limited. To broaden vaccine protection, efforts are underway to accelerate the development of new TB vaccines. However, challenges arise due to the limited immunogenicity and safety of these vaccines, necessitating adjuvants to bolster their ability to elicit a robust immune response for improved and safer immunization. These adjuvants function by augmenting cellular and humoral immunity against M. tuberculosis antigens via different delivery systems, ultimately enhancing vaccine efficacy. Therefore, this paper reviews and summarizes the current research progress on M. tuberculosis vaccines and their associated adjuvants, aiming to provide a valuable reference for the development of novel TB vaccines and the screening of adjuvants.
Collapse
Affiliation(s)
- Hui Wang
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261053, China; (H.W.); (S.W.); (R.F.); (X.L.); (J.X.)
| | - Shuxian Wang
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261053, China; (H.W.); (S.W.); (R.F.); (X.L.); (J.X.)
| | - Ren Fang
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261053, China; (H.W.); (S.W.); (R.F.); (X.L.); (J.X.)
| | - Xiaotian Li
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261053, China; (H.W.); (S.W.); (R.F.); (X.L.); (J.X.)
| | - Jiayin Xing
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261053, China; (H.W.); (S.W.); (R.F.); (X.L.); (J.X.)
| | - Zhaoli Li
- SAFE Pharmaceutical Technology Co., Ltd., Beijing 100000, China
| | - Ningning Song
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261053, China; (H.W.); (S.W.); (R.F.); (X.L.); (J.X.)
| |
Collapse
|
11
|
Li H, Zhu Y, Wang X, Feng Y, Qian Y, Ma Q, Li X, Chen Y, Chen K. Joining Forces: The Combined Application of Therapeutic Viruses and Nanomaterials in Cancer Therapy. Molecules 2023; 28:7679. [PMID: 38005401 PMCID: PMC10674375 DOI: 10.3390/molecules28227679] [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: 09/29/2023] [Revised: 11/10/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023] Open
Abstract
Cancer, on a global scale, presents a monumental challenge to our healthcare systems, posing a significant threat to human health. Despite the considerable progress we have made in the diagnosis and treatment of cancer, realizing precision cancer therapy, reducing side effects, and enhancing efficacy remain daunting tasks. Fortunately, the emergence of therapeutic viruses and nanomaterials provides new possibilities for tackling these issues. Therapeutic viruses possess the ability to accurately locate and attack tumor cells, while nanomaterials serve as efficient drug carriers, delivering medication precisely to tumor tissues. The synergy of these two elements has led to a novel approach to cancer treatment-the combination of therapeutic viruses and nanomaterials. This advantageous combination has overcome the limitations associated with the side effects of oncolytic viruses and the insufficient tumoricidal capacity of nanomedicines, enabling the oncolytic viruses to more effectively breach the tumor's immune barrier. It focuses on the lesion site and even allows for real-time monitoring of the distribution of therapeutic viruses and drug release, achieving a synergistic effect. This article comprehensively explores the application of therapeutic viruses and nanomaterials in tumor treatment, dissecting their working mechanisms, and integrating the latest scientific advancements to predict future development trends. This approach, which combines viral therapy with the application of nanomaterials, represents an innovative and more effective treatment strategy, offering new perspectives in the field of tumor therapy.
Collapse
Affiliation(s)
- Hongyu Li
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China; (Y.Z.); (Y.F.); (Y.Q.); (Q.M.); (X.L.); (Y.C.)
- Ocean College, Beibu Gulf University, Qinzhou 535011, China
| | - Yunhuan Zhu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China; (Y.Z.); (Y.F.); (Y.Q.); (Q.M.); (X.L.); (Y.C.)
| | - Xin Wang
- Center of Infectious Disease Research, School of Life Science, Westlake University, Hangzhou 310024, China;
| | - Yilu Feng
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China; (Y.Z.); (Y.F.); (Y.Q.); (Q.M.); (X.L.); (Y.C.)
| | - Yuncheng Qian
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China; (Y.Z.); (Y.F.); (Y.Q.); (Q.M.); (X.L.); (Y.C.)
| | - Qiman Ma
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China; (Y.Z.); (Y.F.); (Y.Q.); (Q.M.); (X.L.); (Y.C.)
| | - Xinyuan Li
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China; (Y.Z.); (Y.F.); (Y.Q.); (Q.M.); (X.L.); (Y.C.)
| | - Yihan Chen
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China; (Y.Z.); (Y.F.); (Y.Q.); (Q.M.); (X.L.); (Y.C.)
| | - Keda Chen
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China; (Y.Z.); (Y.F.); (Y.Q.); (Q.M.); (X.L.); (Y.C.)
| |
Collapse
|
12
|
Dotiwala F, Upadhyay AK. Next Generation Mucosal Vaccine Strategy for Respiratory Pathogens. Vaccines (Basel) 2023; 11:1585. [PMID: 37896988 PMCID: PMC10611113 DOI: 10.3390/vaccines11101585] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/06/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
Inducing humoral and cytotoxic mucosal immunity at the sites of pathogen entry has the potential to prevent the infection from getting established. This is different from systemic vaccination, which protects against the development of systemic symptoms. The field of mucosal vaccination has seen fewer technological advances compared to nucleic acid and subunit vaccine advances for injectable vaccine platforms. The advent of the next-generation adenoviral vectors has given a boost to mucosal vaccine research. Basic research into the mechanisms regulating innate and adaptive mucosal immunity and the discovery of effective and safe mucosal vaccine adjuvants will continue to improve mucosal vaccine design. The results from clinical trials of inhaled COVID-19 vaccines demonstrate their ability to induce the proliferation of cytotoxic T cells and the production of secreted IgA and IgG antibodies locally, unlike intramuscular vaccinations. However, these mucosal vaccines induce systemic immune responses at par with systemic vaccinations. This review summarizes the function of the respiratory mucosa-associated lymphoid tissue and the advantages that the adenoviral vectors provide as inhaled vaccine platforms.
Collapse
Affiliation(s)
- Farokh Dotiwala
- Ocugen Inc., 11 Great Valley Parkway, Malvern, PA 19355, USA
| | | |
Collapse
|
13
|
Mishra M, Gupta AD, Dadhich R, Ahmad MN, Dasgupta A, Chopra S, Kapoor S. Mycobacterial lipid-derived immunomodulatory drug- liposome conjugate eradicates endosome-localized mycobacteria. J Control Release 2023; 360:578-590. [PMID: 37442202 PMCID: PMC7617025 DOI: 10.1016/j.jconrel.2023.07.013] [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: 02/02/2023] [Revised: 06/25/2023] [Accepted: 07/07/2023] [Indexed: 07/15/2023]
Abstract
Tuberculosis is a challenging disease due to the intracellular residence of its pathogen, Mycobacterium tuberculosis, and modulation of the host bactericidal responses. Lipids from Mycobacterium tuberculosis regulate macrophage immune responses dependent on the infection stage and intracellular location. We show that liposomes constituted with immunostimulatory lipids from mycobacteria modulate the cellular immune response and synergize with sustained drug delivery for effective pathogen eradication. We evaluate the pH-dependent release of Rifampicin from the mycobacterial-lipid-derived liposomes intracellularly and in vitro, their cell viability, long-term stability, and antimicrobial efficacy. Intracellular drug levels were higher following liposome treatment compared with the free drug in a temporal fashion underlying a sustained release. The drug-encapsulated liposomes were taken up by clathrin-mediated endocytosis and elicited a robust pro-inflammatory immune response while localizing in the recycling and late endosomes. Notably, these were the same cellular compartments that contained the pathogen underlying localized intracellular targeting. Our results also imply a lipid-centric and species-specific selectivity of the liposomal drug formulations. This work provides a proof-of-concept for the dual-action of liposomes derived from the pathogen itself for their effective eradication, in conjunction with the attuned host immunomodulation.
Collapse
Affiliation(s)
- Manjari Mishra
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai 400076, India.
| | - Aishi Das Gupta
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai 400076, India; IIT-Bombay Monash Academy, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Ruchika Dadhich
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Mohammad Naiyaz Ahmad
- Division of Microbiology, CSIR-Central Drug Research Institute, Lucknow, UP 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Arunava Dasgupta
- Division of Microbiology, CSIR-Central Drug Research Institute, Lucknow, UP 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Sidharth Chopra
- Division of Microbiology, CSIR-Central Drug Research Institute, Lucknow, UP 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Shobhna Kapoor
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai 400076, India; IIT-Bombay Monash Academy, Indian Institute of Technology Bombay, Mumbai 400076, India; Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8528, Japan.
| |
Collapse
|
14
|
Thakur A, Wadhwa A, Lokras A, Müllertz OAO, Christensen D, Franzyk H, Foged C. Method of manufacturing CAF®09 liposomes affects immune responses induced by adjuvanted subunit proteins. Eur J Pharm Biopharm 2023; 189:84-97. [PMID: 37059402 DOI: 10.1016/j.ejpb.2023.04.005] [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: 12/22/2022] [Revised: 04/03/2023] [Accepted: 04/06/2023] [Indexed: 04/16/2023]
Abstract
The ability to induce antigen-specific CD4+ and CD8+T-cell responses is one of the fundamental requirements when developing new efficacious vaccines against challenging infectious diseases and cancer. However, no adjuvants are currently approved for human subunit vaccines that induce T-cell immunity. Here, we incorporated a Toll-like receptor 4 agonist, i.e., the ionizable lipidoid L5N12, in the liposomal cationic adjuvant formulation 09 (CAF®09), and found that modified CAF®09 liposomes possess preserved adjuvant function as compared to unmodified CAF®09. CAF®09 consists of the cationic lipid dimethyldioctadecylammonium (DDA), monomycoloyl glycerol analogue 1 (MMG-1), and polyinosinic:polycytidylic acid [poly(I:C)]. By using the microfluidic mixing technology for liposome preparation, we gradually replaced DDA with L5N12, while keeping the molar ratios of MMG-1 and poly(I:C) constant. We found that this type of modification resulted in colloidally stable liposomes, which were significantly smaller and displayed reduced surface charge as compared to unmodified CAF®09, prepared by using the conventional thin film method. We showed that incorporation of L5N12 decreases the membrane rigidity of CAF®09 liposomes. Furthermore, vaccination with antigen adjuvanted with L5N12-modified CAF®09 or antigen adjuvanted with unmodified CAF®09, respectively, induced comparable antigen-specific serum antibody titers. We found that antigen adjuvanted with L5N12-modified CAF®09 induced antigen-specific effector and memory CD4+ and CD8+T-cell responses in the spleen comparable to those induced when unmodified CAF®09 was used as adjuvant. However, incorporating L5N12 did not have a synergistic immunopotentiating effect on the antibody and T-cell responses induced by CAF®09. Moreover, vaccination with antigen adjuvanted with unmodified CAF®09, which was manufactured by using microfluidic mixing, induced significantly lower antigen-specific CD4+ and CD8+T-cell responses than vaccination with antigen adjuvanted with unmodified CAF®09, which was prepared by using the thin film method. These results show that the method of manufacturing affects CAF®09 liposome adjuvanted antigen-specific immune responses, which should be taken into consideration when evaluating immunogenicity of subunit protein vaccines.
Collapse
Affiliation(s)
- Aneesh Thakur
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark.
| | - Abishek Wadhwa
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Abhijeet Lokras
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Olivia Amanda Oest Müllertz
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Dennis Christensen
- Department of Infectious Disease Immunology, Statens Serum Institut, Artillerivej 5, 2300 Copenhagen S, Denmark
| | - Henrik Franzyk
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 162, 2100 Copenhagen Ø, Denmark
| | - Camilla Foged
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| |
Collapse
|
15
|
Moradi M, Vahedi F, Abbassioun A, Ramezanpour Shahi A, Sholeh M, Taheri‐Anganeh M, Dargahi Z, Ghanavati R, Khatami SH, Movahedpour A. Liposomal delivery system/adjuvant for tuberculosis vaccine. Immun Inflamm Dis 2023; 11:e867. [PMID: 37382263 PMCID: PMC10251763 DOI: 10.1002/iid3.867] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 04/23/2023] [Accepted: 04/26/2023] [Indexed: 06/30/2023] Open
Abstract
As reported by the World Health Organization, about 10 million individuals were infected with tuberculosis (TB) worldwide. Moreover, approximately 1.5 million people died of TB, of which 214,000 were infected with HIV simultaneously. Due to the high infection rate, the need for effective TB vaccination is highly felt. Until now, various methodologies have been proposed for the development of a protein subunit vaccine for TB. These vaccines have shown higher protection than other vaccines, particularly the Bacillus culture vaccine. The delivery system and safety regulator are common characteristics of effective adjuvants in TB vaccines and the clinical trial stage. The present study investigates the current state of TB adjuvant research focusing on the liposomal adjuvant system. Based on our findings, the liposomal system is a safe and efficient adjuvant from nanosize to microsize for vaccinations against TB, other intracellular infections, and malignancies. Clinical studies can provide valuable feedback for developing novel TB adjuvants, which ultimately enhance the impact of adjuvants on next-generation TB vaccines.
Collapse
Affiliation(s)
- Melika Moradi
- Department of Microbiology, School of MedicineAhvaz Jundishapur University of Medical SciencesAhvazIran
| | - Farzaneh Vahedi
- Department of Medical Biotechnology, School of Advanced Medical Sciences and TechnologiesShiraz University of Medical SciencesShirazIran
| | - Arian Abbassioun
- Department of Virology, Faculty of Veterinary MediceneUniversity of TehranTehranIran
| | - Arash Ramezanpour Shahi
- Department of Veterinary Clinical Sciences, Poultry diseases and hygiene Resident, Faculty of Veterinary MedicineShahrekord UniversityShahrekordIran
| | - Mohammad Sholeh
- Department of BacteriologyPasteur Institute of IranTehranIran
| | - Mortaza Taheri‐Anganeh
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research InstituteUrmia University of Medical SciencesUrmiaIran
| | - Zahra Dargahi
- Department of Microbiology, School of MedicineAhvaz Jundishapur University of Medical SciencesAhvazIran
| | | | - Seyyed Hossein Khatami
- Department of Clinical Biochemistry, School of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | | |
Collapse
|
16
|
Multiepitope Subunit Peptide-Based Nanovaccine against Porcine Circovirus Type 2 (PCV2) Elicited High Antibody Titers in Vaccinated Mice. Molecules 2023; 28:molecules28052248. [PMID: 36903494 PMCID: PMC10005372 DOI: 10.3390/molecules28052248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/22/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023] Open
Abstract
Porcine circovirus 2 (PCV2) infection is one of the most serious threats to the swine industry. While the disease can be prevented, to some extent, by commercial PCV2a vaccines, the evolving nature of PCV2 necessitates the development of a novel vaccine that can compete with the mutations of the virus. Thus, we have developed novel multiepitope vaccines based on the PCV2b variant. Three PCV2b capsid protein epitopes, together with a universal T helper epitope, were synthesized and formulated with five delivery systems/adjuvants: complete Freund's adjuvant, poly(methyl acrylate) (PMA), poly(hydrophobic amino acid), liposomes and rod-shaped polymeric nanoparticles built from polystyrene-poly(N-isopropylacrylamide)-poly(N-dimethylacrylamide). Mice were subcutaneously immunized with the vaccine candidates three times at three-week intervals. All vaccinated mice produced high antibody titters after three immunizations as analyzed by the enzyme-linked immunosorbent assay (ELISA), while mice vaccinated with PMA-adjuvanted vaccine elicited high antibody titers even after a single immunization. Thus, the multiepitope PCV2 vaccine candidates designed and examined here show strong potential for further development.
Collapse
|
17
|
Cell-free protein synthesis systems for vaccine design and production. Curr Opin Biotechnol 2023; 79:102888. [PMID: 36641905 DOI: 10.1016/j.copbio.2022.102888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 12/14/2022] [Indexed: 01/15/2023]
Abstract
Vaccines are vital for protection against existing and emergent diseases. Current vaccine production strategies are limited by long production times, risky viral material, weak immunogenicity, and poor stability, ultimately restricting the safe or rapid production of vaccines for widespread utilization. Cell-free protein synthesis (CFPS) systems, which use extracted transcriptional and translational machinery from cells, are promising tools for vaccine production because they can rapidly produce proteins without the constraints of living cells, have a highly optimizable open system, and can be used for on-demand biomanufacturing. Here, we review how CFPS systems have been explored for the production of subunit, conjugate, virus-like particle (VLP), and membrane-augmented vaccines and as a tool in vaccine design. We also discuss efforts to address potential limitations with CFPS such as the presence of endotoxins, poor protein folding, reaction stability, and glycosylation to enable promising future vaccine design and production.
Collapse
|
18
|
Licensed liposomal vaccines and adjuvants in the antigen delivery system. BIOTECHNOLOGIA 2022; 103:409-423. [PMID: 36685697 PMCID: PMC9837556 DOI: 10.5114/bta.2022.120709] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/10/2022] [Accepted: 08/02/2022] [Indexed: 12/28/2022] Open
Abstract
Liposomes (LSs) are promising nanoparticles with unique properties such as controlled nanosize, large surface area, increased reactivity, and ability to undergo modification. Worldwide, licensed liposomal forms of antibiotics, hormones, antioxidants, cytostatics, ophthalmic drugs, etc., are available on the pharmaceutical market. This review focuses on the adjuvant properties of LSs in the production of vaccines (VACs). LS-VACs have the following advantages: antigens with low immunogenicity can become highly immunogenic; LSs can include both hydrophilic and hydrophobic antigens; LSs allow to achieve a prolonged specific action of antibodies; and LSs reduce the toxicity and pyrogenicity of encapsulated antigens and adjuvants. The immune response is influenced by the composition of the liposomal membrane, physicochemical characteristics of lipids, antigen localization in LSs, interaction of LSs with complement, and a number of proteins, which leads to opsonization. The major requirements for adjuvants are their ability to enhance the immune response, biodegradability, and elimination from the organism, and LSs fully meet these requirements. The effectiveness and safety of LSs as carriers in the antigen delivery system have been proven by the long-term clinical use of licensed vaccines against hepatitis A, influenza, herpes zoster, malaria, and COVID-19.
Collapse
|
19
|
Bressy C, Zemani A, Goyal S, Jishkariani D, Lee CN, Chen YH. Inhibition of c-Rel expression in myeloid and lymphoid cells with distearoyl -phosphatidylserine (DSPS) liposomal nanoparticles encapsulating therapeutic siRNA. PLoS One 2022; 17:e0276905. [PMID: 36520934 PMCID: PMC9754606 DOI: 10.1371/journal.pone.0276905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 10/14/2022] [Indexed: 12/23/2022] Open
Abstract
c-Rel, a member of the nuclear factor kappa B (NF-κB) family, is preferentially expressed by immune cells and is known to regulate inflammation, autoimmune diseases and cancer. However, there is a lack of therapeutic intervention to specifically inhibit c-Rel in immune cells. Recent success with Pfizer and Moderna mRNA lipid-encapsulated vaccines as well as FDA approved medicines based on siRNA prompted us to test a lipid nanoparticle-based strategy to silence c-Rel in immune cells. Specifically, we encapsulated c-Rel-targeting siRNA into distearoyl-phosphatidylserine (DSPS)-containing nanoparticles. DSPS is a saturated phospholipid that serves as the "eat-me" signal for professional phagocytes such as macrophages and neutrophils of the immune system. We demonstrated here that incorporation of DSPS in liposome nanoparticles (LNP) improved their uptake by immune cells. LNP containing high concentrations of DSPS were highly effective to transfect not only macrophages and neutrophils, but also lymphocytes, with limited toxicity to cells. However, LNP containing low concentrations of DSPS were more effective to transfect myeloid cells than lymphoid cells. Importantly, DSPS-LNP loaded with a c-Rel siRNA were highly effective to inhibit c-Rel expression in several professional phagocytes tested, which lasted for several days. Taken together, our results suggest that DSPS-LNP armed with c-Rel siRNA could be exploited to target immune cells to limit the development of inflammatory diseases or cancer caused by c-Rel upregulation. In addition, this newly developed DSPS-LNP system may be further tested to encapsulate and deliver other small molecule drugs to immune cells, especially macrophages, neutrophils, and lymphocytes for the treatment of diseases.
Collapse
Affiliation(s)
- Christian Bressy
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Ali Zemani
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Shreya Goyal
- Department of Biological Sciences, University of North Carolina, Charlotte, North Carolina, United States of America
| | - Davit Jishkariani
- Chemical and Nanoparticle Synthesis Core (CNSC), The University of Pennsylvania, Philadelphia, PA, United States of America
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Chin Nien Lee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Youhai H. Chen
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Faculty of Pharmaceutical Sciences, CAS Shenzhen Institute of Advanced Technology, Shenzhen, China
| |
Collapse
|
20
|
Bolhassani A. Lipid-Based Delivery Systems in Development of Genetic and Subunit Vaccines. Mol Biotechnol 2022; 65:669-698. [PMID: 36462102 PMCID: PMC9734811 DOI: 10.1007/s12033-022-00624-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 11/26/2022] [Indexed: 12/07/2022]
Abstract
Lipidic carriers are composed of natural, synthetic, or physiological lipid/phospholipid materials. The flexibility of lipid-based delivery systems for transferring a variety of molecules such as immunomodulators, antigens, and drugs play a key role in design of effective vaccination and therapeutic strategies against infectious and non-infectious diseases. Genetic and subunit vaccines are two major groups of promising vaccines that have the potential for improving the protective potency against different diseases. These vaccine strategies rely greatly on delivery systems with various functions, including cargo protection, targeted delivery, high bioavailability, controlled release of antigens, selective induction of antigen-specific humoral or cellular immune responses, and low side effects. Lipidic carriers play a key role in local tissue distribution, retention, trafficking, uptake and processing by antigen-presenting cells. Moreover, lipid nanoparticles have successfully achieved to the clinic for the delivery of mRNA. Their broad potential was shown by the recent approval of COVID-19 mRNA vaccines. However, size, charge, architecture, and composition need to be characterized to develop a standard lipidic carrier. Regarding the major roles of lipid-based delivery systems in increasing the efficiency and safety of vaccine strategies against different diseases, this review concentrates on their recent advancements in preclinical and clinical trials.
Collapse
Affiliation(s)
- Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
21
|
Bezbaruah R, Chavda VP, Nongrang L, Alom S, Deka K, Kalita T, Ali F, Bhattacharjee B, Vora L. Nanoparticle-Based Delivery Systems for Vaccines. Vaccines (Basel) 2022; 10:1946. [PMID: 36423041 PMCID: PMC9694785 DOI: 10.3390/vaccines10111946] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 11/08/2022] [Accepted: 11/09/2022] [Indexed: 11/19/2022] Open
Abstract
Vaccination is still the most cost-effective way to combat infectious illnesses. Conventional vaccinations may have low immunogenicity and, in most situations, only provide partial protection. A new class of nanoparticle-based vaccinations has shown considerable promise in addressing the majority of the shortcomings of traditional and subunit vaccines. This is due to recent breakthroughs in chemical and biological engineering, which allow for the exact regulation of nanoparticle size, shape, functionality, and surface characteristics, resulting in improved antigen presentation and robust immunogenicity. A blend of physicochemical, immunological, and toxicological experiments can be used to accurately characterize nanovaccines. This narrative review will provide an overview of the current scenario of the nanovaccine.
Collapse
Affiliation(s)
- Rajashri Bezbaruah
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh 786004, Assam, India
| | - Vivek P. Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L. M. College of Pharmacy, Ahmedabad 380008, Gujarat, India
| | - Lawandashisha Nongrang
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh 786004, Assam, India
| | - Shahnaz Alom
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh 786004, Assam, India
- Department of Pharmacology, Girijananda Chowdhury Institute of Pharmaceutical Science-Tezpur, Sonitpur 784501, Assam, India
| | - Kangkan Deka
- Department of Pharmacognosy, NETES Institute of Pharmaceutical Science, Mirza, Guwahati 781125, Assam, India
| | - Tutumoni Kalita
- Department of Pharmaceutical Chemistry, Girijananda Chowdhury Institute of Pharmaceutical Sciences, Azara, Guwahati 781017, Assam, India
| | - Farak Ali
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh 786004, Assam, India
- Department of Pharmaceutical Chemistry, Girijananda Chowdhury Institute of Pharmaceutical Science-Tezpur, Sonitpur 784501, Assam, India
| | - Bedanta Bhattacharjee
- Department of Pharmacology, Girijananda Chowdhury Institute of Pharmaceutical Science-Tezpur, Sonitpur 784501, Assam, India
| | | |
Collapse
|
22
|
Kazemi M, Aghamaali MR, Madani R, Emami T, Golchinfar F. Evaluating the Immunogenicity of Recombinant VP1 Protein from the Foot-and-Mouth Disease Virus Encapsulated in Nanoliposome in Guinea Pig Animal Model. Vet Immunol Immunopathol 2022; 253:110497. [DOI: 10.1016/j.vetimm.2022.110497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 08/08/2022] [Accepted: 09/22/2022] [Indexed: 10/14/2022]
|
23
|
Azuar A, Madge HYR, Boer JC, Gonzalez Cruz JL, Wang J, Khalil ZG, Deceneux C, Goodchild G, Yang J, Koirala P, Hussein WM, Capon RJ, Plebanski M, Toth I, Skwarczynski M. Poly(hydrophobic Amino Acids) and Liposomes for Delivery of Vaccine against Group A Streptococcus. Vaccines (Basel) 2022; 10:vaccines10081212. [PMID: 36016100 PMCID: PMC9413763 DOI: 10.3390/vaccines10081212] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/22/2022] [Accepted: 07/27/2022] [Indexed: 12/04/2022] Open
Abstract
Adjuvants and delivery systems are essential components of vaccines to increase immunogenicity against target antigens, particularly for peptide epitopes (poor immunogens). Emulsions, nanoparticles, and liposomes are commonly used as a delivery system for peptide-based vaccines. A Poly(hydrophobic amino acids) delivery system was previously conjugated to Group A Streptococcus (GAS)-derived peptide epitopes, allowing the conjugates to self-assemble into nanoparticles with self adjuvanting ability. Their hydrophobic amino acid tail also serves as an anchoring moiety for the peptide epitope, enabling it to be integrated into the liposome bilayer, to further boost the immunological responses. Polyleucine-based conjugates were anchored to cationic liposomes using the film hydration method and administered to mice subcutaneously. The polyleucine-peptide conjugate, its liposomal formulation, and simple liposomal encapsulation of GAS peptide epitope induced mucosal (saliva IgG) and systemic (serum IgG, IgG1 and IgG2c) immunity in mice. Polyleucine acted as a potent liposome anchoring portion, which stimulated the production of highly opsonic antibodies. The absence of polyleucine in the liposomal formulation (encapsulated GAS peptide) induced high levels of antibody titers, but with poor opsonic ability against GAS bacteria. However, the liposomal formulation of the conjugated vaccine was no more effective than conjugates alone self-assembled into nanoparticles.
Collapse
Affiliation(s)
- Armira Azuar
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (A.A.); (H.Y.R.M.); (J.W.); (J.Y.); (P.K.); (W.M.H.); (I.T.)
| | - Harrison Y. R. Madge
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (A.A.); (H.Y.R.M.); (J.W.); (J.Y.); (P.K.); (W.M.H.); (I.T.)
| | - Jennifer C. Boer
- School of Health and Biomedical Sciences, RMIT University, Bundoora West, VIC 3083, Australia; (J.C.B.); (C.D.); (G.G.); (M.P.)
| | - Jazmina L. Gonzalez Cruz
- Diamantina Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, QLD 4102, Australia;
| | - Jingwen Wang
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (A.A.); (H.Y.R.M.); (J.W.); (J.Y.); (P.K.); (W.M.H.); (I.T.)
| | - Zeinab G. Khalil
- Institute of Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia; (Z.G.K.); (R.J.C.)
| | - Cyril Deceneux
- School of Health and Biomedical Sciences, RMIT University, Bundoora West, VIC 3083, Australia; (J.C.B.); (C.D.); (G.G.); (M.P.)
| | - Georgia Goodchild
- School of Health and Biomedical Sciences, RMIT University, Bundoora West, VIC 3083, Australia; (J.C.B.); (C.D.); (G.G.); (M.P.)
| | - Jieru Yang
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (A.A.); (H.Y.R.M.); (J.W.); (J.Y.); (P.K.); (W.M.H.); (I.T.)
| | - Prashamsa Koirala
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (A.A.); (H.Y.R.M.); (J.W.); (J.Y.); (P.K.); (W.M.H.); (I.T.)
| | - Waleed M. Hussein
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (A.A.); (H.Y.R.M.); (J.W.); (J.Y.); (P.K.); (W.M.H.); (I.T.)
| | - Robert J. Capon
- Institute of Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia; (Z.G.K.); (R.J.C.)
| | - Magdalena Plebanski
- School of Health and Biomedical Sciences, RMIT University, Bundoora West, VIC 3083, Australia; (J.C.B.); (C.D.); (G.G.); (M.P.)
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (A.A.); (H.Y.R.M.); (J.W.); (J.Y.); (P.K.); (W.M.H.); (I.T.)
- Institute of Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia; (Z.G.K.); (R.J.C.)
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Mariusz Skwarczynski
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (A.A.); (H.Y.R.M.); (J.W.); (J.Y.); (P.K.); (W.M.H.); (I.T.)
- Correspondence: ; Tel.: +617-3346-9894
| |
Collapse
|
24
|
The Big Potential of Small Particles: Lipid-Based Nanoparticles and Exosomes in Vaccination. Vaccines (Basel) 2022; 10:vaccines10071119. [PMID: 35891282 PMCID: PMC9320421 DOI: 10.3390/vaccines10071119] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/07/2022] [Accepted: 07/11/2022] [Indexed: 12/13/2022] Open
Abstract
Some of the most significant medical achievements in recent history are the development of distinct and effective vaccines, and the improvement of the efficacy of previously existing ones, which have contributed to the eradication of many dangerous and life-threatening diseases. Immunization depends on the generation of a physiological memory response and protection against infection. It is therefore crucial that antigens are delivered in an efficient manner, to elicit a robust immune response. The recent approval of COVID-19 vaccines containing lipid nanoparticles encapsulating mRNA demonstrates the broad potential of lipid-based delivery systems. In light of this, the present review article summarizes currently synthesized lipid-based nanoparticles such as liposomes, lipid-nano particles, or cell-derived exosomes.
Collapse
|
25
|
Comparison of adjuvant properties of chitosan during oral and subcutaneous immunization of mice with BSA. UKRAINIAN BIOCHEMICAL JOURNAL 2022. [DOI: 10.15407/ubj94.02.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Vaccination is the best method to prevent the spread of infectious diseases, its disadvantages are side effects. Potentially safe DNA, RNA or protein molecules possess antigenic properties, but are low-immunogenic and therefore require conjugation with an adjuvant. The aim of the research was to evaluate Chitosan (CS) potency as an adjuvant and compare its effectiveness depending on the route of drug administration. The experiments were carried out on 3 groups of BALB/c mice. Mice of the first group were injected subcutaneously with 20 µl of a mixture of CS (3.3 mg/kg) and BSA (1.7 mg/kg). The mixture of CS and BSA at the same doses and volume was administered orally to mice of the second experimental group. The third group – control – unvaccinated mice. Anti-BSA antibody levels were measured by ELISA. Aspartate aminotransferase, alanine aminotransferase activity and cholesterol, creatinine and urea levels were determined in the serum. It was found that both subcutaneous and mucosal immunizations provided a 2-fold increase in anti-BSA antibody titers against the background of maintaining all biochemical blood parameters at the level of the physiological norm. However, AST activity in the serum of oral-immunized mice was elevated as compared to subcutaneous-immunized mice. Serum cholesterol level in the group of subcutaneously immunized mice and creatinine and urea levels in both experimental groups were reduced compared to the control. It is concluded that oral immunization with CS is the optimal route for antigen-specific IgG antibody response induction.
Collapse
|
26
|
AboulFotouh K, Xu H, Moon C, Williams RO, Cui Z. Development of (Inhalable) Dry Powder Formulations of AS01 B-Containing Vaccines Using Thin-Film Freeze-Drying. Int J Pharm 2022; 622:121825. [PMID: 35577037 DOI: 10.1016/j.ijpharm.2022.121825] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/07/2022] [Accepted: 05/09/2022] [Indexed: 01/08/2023]
Abstract
AS01B is a liposomal formulation of two immunostimulants namely 3-O-desacyl-4́-monophosphoryl lipid A (MPL) and QS-21. The liposomal formulation of AS01B reduces the endotoxicity of MPL and the lytic activity of QS-21. The AS01B-adjuvanted Shingrix vaccine is marketed in a two-vial presentation, with the liquid AS01B liposomes in one vial and the antigen as a dry powder in another vial. In the present study, we tested the feasibility of applying thin-film freeze-drying (TFFD) to engineer dry powders of the AS01B liposomal adjuvant alone or vaccines containing AS01B as an adjuvant. Initially, we showed that after the AS01B liposomal adjuvant was subjected to TFFD using sucrose as a stabilizer at 4% w/v, the particle size distribution of AS01B liposomes reconstituted from the dry powder was identical to the liquid adjuvant before drying. We then showed using ovalbumin (OVA) as a model antigen adjuvanted with AS01B (AS01B/OVA) that subjecting the AS01B/OVA vaccine to TFFD and subsequent reconstitution did not negatively affect the AS01B liposome particle size, nor the immunogenicity of the vaccine. Importantly, the thin-film freeze-dried AS01B/OVA vaccine, unlike its liquid counterpart, was not sensitive to repeated freezing-and-thawing. The developed AS01B/OVA dry powder also showed the desirable aerosol properties (i.e., fine particle fraction of 66.3 ± 4.9% and mass median aerodynamic diameter of 2.4 ± 0.1 µm) for potential pulmonary administration. Finally, the feasibility of using TFFD to prepare dry powders of AS01B-adjuvanted vaccines was further confirmed using AS01B-adjuvanted Fluzone Quadrivalent and Shingrix, which contains AS01B. It is concluded that the TFFD technology can enable the formulation of AS01B-adjuvanted vaccines as freezing-insensitive, inhalable dry powders in a single-vial presentation.
Collapse
Affiliation(s)
- Khaled AboulFotouh
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; Department of Pharmaceutics, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Haiyue Xu
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Chaeho Moon
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Robert O Williams
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA.
| | - Zhengrong Cui
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
27
|
Ou BS, Saouaf OM, Baillet J, Appel EA. Sustained delivery approaches to improving adaptive immune responses. Adv Drug Deliv Rev 2022; 187:114401. [PMID: 35750115 DOI: 10.1016/j.addr.2022.114401] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 06/14/2022] [Accepted: 06/17/2022] [Indexed: 11/17/2022]
Abstract
The immune system is one of the most important, complex biological networks regulating and protecting human health. Its precise modulation can prevent deadly infections and fight cancer. Accordingly, prophylactic vaccines and cancer immunotherapies are some of the most powerful technologies to protect against potential dangers through training of the immune system. Upon immunization, activation and maturation of B and T cells of the adaptive immune system are necessary for development of proper humoral and cellular protection. Yet, the exquisite organization of the immune system requires spatiotemporal control over the exposure of immunomodulatory signals. For example, while the human immune system has evolved to develop immunity to natural pathogenic infections that often last for weeks, current prophylactic vaccination technologies only expose the immune system to immunomodulatory signals for hours to days. It has become clear that leveraging sustained release technologies to prolong immunogen and adjuvant exposure can increase the potency, durability, and quality of adaptive immune responses. Over the past several years, tremendous breakthroughs have been made in the design of novel biomaterials such as nanoparticles, microparticles, hydrogels, and microneedles that can precisely control and the presentation of immunomodulatory signals to the immune system. In this review, we discuss relevant sustained release strategies and their corresponding benefits to cellular and humoral responses.
Collapse
Affiliation(s)
- Ben S Ou
- Department of Bioengineering, Stanford University, Stanford 94305, USA
| | - Olivia M Saouaf
- Department of Materials Science & Engineering, Stanford University, Stanford 94305, USA
| | - Julie Baillet
- Department of Materials Science & Engineering, Stanford University, Stanford 94305, USA; University of Bordeaux, CNRS, Bordeaux INP, LCPO, UMR 5629, Pessac 33600, France
| | - Eric A Appel
- Department of Bioengineering, Stanford University, Stanford 94305, USA; Department of Materials Science & Engineering, Stanford University, Stanford 94305, USA; Department of Pediatrics (Endocrinology), Stanford University, Stanford 94305, USA; ChEM-H Institute, Stanford University, Stanford CA 94305, USA; Woods Institute for the Environment, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
28
|
Osterloh A. Vaccination against Bacterial Infections: Challenges, Progress, and New Approaches with a Focus on Intracellular Bacteria. Vaccines (Basel) 2022; 10:751. [PMID: 35632507 PMCID: PMC9144739 DOI: 10.3390/vaccines10050751] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/08/2022] [Accepted: 05/09/2022] [Indexed: 12/13/2022] Open
Abstract
Many bacterial infections are major health problems worldwide, and treatment of many of these infectious diseases is becoming increasingly difficult due to the development of antibiotic resistance, which is a major threat. Prophylactic vaccines against these bacterial pathogens are urgently needed. This is also true for bacterial infections that are still neglected, even though they affect a large part of the world's population, especially under poor hygienic conditions. One example is typhus, a life-threatening disease also known as "war plague" caused by Rickettsia prowazekii, which could potentially come back in a war situation such as the one in Ukraine. However, vaccination against bacterial infections is a challenge. In general, bacteria are much more complex organisms than viruses and as such are more difficult targets. Unlike comparatively simple viruses, bacteria possess a variety of antigens whose immunogenic potential is often unknown, and it is unclear which antigen can elicit a protective and long-lasting immune response. Several vaccines against extracellular bacteria have been developed in the past and are still used successfully today, e.g., vaccines against tetanus, pertussis, and diphtheria. However, while induction of antibody production is usually sufficient for protection against extracellular bacteria, vaccination against intracellular bacteria is much more difficult because effective defense against these pathogens requires T cell-mediated responses, particularly the activation of cytotoxic CD8+ T cells. These responses are usually not efficiently elicited by immunization with non-living whole cell antigens or subunit vaccines, so that other antigen delivery strategies are required. This review provides an overview of existing antibacterial vaccines and novel approaches to vaccination with a focus on immunization against intracellular bacteria.
Collapse
Affiliation(s)
- Anke Osterloh
- Department of Infection Immunology, Research Center Borstel, Parkallee 22, 23845 Borstel, Germany
| |
Collapse
|
29
|
Costa B, Boueri B, Oliveira C, Silveira I, Ribeiro AJ. Lipoplexes and polyplexes as nucleic acids delivery nanosystems: The current state and future considerations. Expert Opin Drug Deliv 2022; 19:577-594. [DOI: 10.1080/17425247.2022.2075846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Bruno Costa
- University of Coimbra, Faculty of Pharmacy, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Beatriz Boueri
- University of Coimbra, Faculty of Pharmacy, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Claudia Oliveira
- Group Genetics of Cognitive Dysfunction, IBMC - Instituto de Biologia Molecular e Celular, I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
| | - Isabel Silveira
- University of Coimbra, Faculty of Pharmacy, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Group Genetics of Cognitive Dysfunction, IBMC - Instituto de Biologia Molecular e Celular, I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
| | - Antonio J. Ribeiro
- University of Coimbra, Faculty of Pharmacy, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Group Genetics of Cognitive Dysfunction, IBMC - Instituto de Biologia Molecular e Celular, I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
| |
Collapse
|
30
|
Development of Peptide-Based Vaccines for Cancer. JOURNAL OF ONCOLOGY 2022; 2022:9749363. [PMID: 35342400 PMCID: PMC8941562 DOI: 10.1155/2022/9749363] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 02/23/2022] [Indexed: 12/14/2022]
Abstract
Peptides cancer vaccines are designed based on the epitope peptides that can elicit humoral and cellular immune responses targeting tumor-associated antigens (TAAs) or tumor-specific antigens (TSAs). In order to develop a clinically safe and more effective vaccine for the future, several issues need to be addressed, and these include the selection of optimal antigen targets, adjuvants, and immunization regimens. Another emerging approach involves the use of personalized peptide-based vaccines based on neoantigens to enhance antitumor response. Rationally designed combinatorial therapy is currently being investigated with chemotherapeutic drugs or immune checkpoint inhibitor therapies to improve the efficacy. This review discusses an overview of the development of peptide-based vaccines, the role of adjuvants, and the delivery systems for peptide vaccines as well as combinatorial therapy as potential anticancer strategies.
Collapse
|
31
|
Tretiakova DS, Vodovozova EL. Liposomes as Adjuvants and Vaccine Delivery Systems. BIOCHEMISTRY (MOSCOW) SUPPLEMENT. SERIES A, MEMBRANE AND CELL BIOLOGY 2022; 16:1-20. [PMID: 35194485 PMCID: PMC8853224 DOI: 10.1134/s1990747822020076] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 09/22/2021] [Accepted: 09/24/2021] [Indexed: 12/12/2022]
Abstract
The review considers liposomes as systems of substantial interest as adjuvant carriers in vaccinology due to their versatility and maximal biocompatibility. Research and development on the use of liposomes and lipid nanoparticles to create subunit vaccines for the prevention and treatment of infectious diseases has been going on for several decades. In recent years, the area has seen serious progress due to the improvement of the technology of industrial production of various high-grade lipids suitable for parenteral administration and the emergence of new technologies and equipment for the production of liposomal preparations. When developing vaccines, it is necessary to take into account how the body’s immune system (innate and adaptive immunity) functions. The review briefly describes some of the fundamental mechanisms underlying the mobilization of immunity when encountering an antigen, as well as the influence of liposome carriers on the processes of internalization of antigens by immunocompetent cells and ways of immune response induction. The results of the studies on the interactions of liposomes with antigen-presenting cells in function of the liposome size, charge, and phase state of the bilayer, which depends on the lipid composition, are often contradictory and should be verified in each specific case. The introduction of immunostimulant components into the composition of liposomal vaccine complexes—ligands of the pathogen-associated molecular pattern receptors—permits modulation of the strength and type of the immune response. The review briefly discusses liposome-based vaccines approved for use in the clinic for the treatment and prevention of infectious diseases, including mRNA-loaded lipid nanoparticles. Examples of liposomal vaccines that undergo various stages of clinical trials are presented.
Collapse
Affiliation(s)
- D S Tretiakova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - E L Vodovozova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| |
Collapse
|
32
|
Zhou SH, Zhang RY, Zhang HW, Liu YL, Wen Y, Wang J, Li YT, You ZW, Yin XG, Qiu H, Gong R, Yang GF, Guo J. RBD conjugate vaccine with a built-in TLR1/2 agonist is highly immunogenic against SARS-CoV-2 and variants of concern. Chem Commun (Camb) 2022; 58:2120-2123. [PMID: 35040862 DOI: 10.1039/d1cc06520c] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The coronavirus 2019 (COVID-19) pandemic is causing serious impacts in the world, and safe and effective vaccines and medicines are the best methods to combat the disease. The receptor-binding domain (RBD) of the SARS-CoV-2 spike protein plays a key role in interacting with the angiotensin-converting enzyme 2 (ACE2) receptor, and is regarded as an important target of vaccines. Herein, we constructed the adjuvant-protein conjugate Pam3CSK4-RBD as a vaccine candidate, in which the N-terminal of the RBD was site-selectively oxidized by transamination and conjugated with the TLR1/2 agonist Pam3CSK4. This demonstrated that the conjugation of Pam3CSK4 significantly enhanced the anti-RBD antibody response and cellular response. In addition, sera from the Pam3CSK4-RBD immunized group efficiently inhibited the binding of the RBD to ACE2 and protected cells from SARS-CoV-2 and four variants of concern (alpha, beta, gamma and delta), indicating that this adjuvant strategy could be one of the effective means for protein vaccine development.
Collapse
Affiliation(s)
- Shi-Hao Zhou
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensing Technology and Health, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, College of Chemistry, Central China Normal University, Wuhan, 430079, China.
| | - Ru-Yan Zhang
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensing Technology and Health, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, College of Chemistry, Central China Normal University, Wuhan, 430079, China.
| | - Hai-Wei Zhang
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China.
| | - Yan-Ling Liu
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensing Technology and Health, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, College of Chemistry, Central China Normal University, Wuhan, 430079, China.
| | - Yu Wen
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensing Technology and Health, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, College of Chemistry, Central China Normal University, Wuhan, 430079, China.
| | - Jian Wang
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensing Technology and Health, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, College of Chemistry, Central China Normal University, Wuhan, 430079, China.
| | - Yu-Ting Li
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensing Technology and Health, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, College of Chemistry, Central China Normal University, Wuhan, 430079, China.
| | - Zi-Wei You
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensing Technology and Health, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, College of Chemistry, Central China Normal University, Wuhan, 430079, China.
| | - Xu-Guang Yin
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensing Technology and Health, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, College of Chemistry, Central China Normal University, Wuhan, 430079, China.
| | - Hong Qiu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Rui Gong
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China.
| | - Guang-Fu Yang
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensing Technology and Health, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, College of Chemistry, Central China Normal University, Wuhan, 430079, China.
| | - Jun Guo
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensing Technology and Health, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, College of Chemistry, Central China Normal University, Wuhan, 430079, China.
| |
Collapse
|
33
|
Kazemi M, Madani R, Aghamaali MR, Emami T, Golchinfar F, Heshmati L. Preparation and Characterization of Nanoliposome Containing Isolated VP1 Protein of Foot and Mouth Disease Virus as a Model of Vaccine. ARCHIVES OF RAZI INSTITUTE 2022; 77:37-44. [PMID: 35891774 PMCID: PMC9288613 DOI: 10.22092/ari.2021.353322.1596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 04/10/2021] [Indexed: 06/15/2023]
Abstract
Foot-and-mouth disease (FMD) is an acute and highly contagious disease in livestock, such as cattle, sheep, and pigs, leading to a lot of economic losses. The current FMD vaccines formulated by inactivated whole-virus and adjuvant successfully reduce disease outbreaks in many regions of the world. Immunological studies on FMD viruses revealed that the dominant epitope in arising neutral antibody response is amino acid residues constructing the G-H loop, constituting a surface loop of the structural protein, termed VP1. Liposomes as one of the most well-known vehicles are considered an important carrier in vaccine development, and their function is used to encapsulate purified VP1 protein based on their size, charge, and lipid content. Accordingly, the VP1 protein was isolated from the FMD virus. This study aimed to compare four methods of VP1 protein encapsulation in the liposome and the extruding effect, as follows: 1) VP1 protein was dissolved in dimethyl sulfoxide and added to the lipid film hydrated by ethanol, 2) the lipid film was hydrated by VP1 protein with 7M urea, 3) the lipid film was hydrated by VP1 protein and freeze-thawed, and 4) the lipid film was hydrated by VP1 protein. The highest encapsulation efficiency was 91% in the second method which purified protein-containing urea. The VP1 protein in the prepared liposome (1, 2-dimyristoyl-sn-glycero-3-phosphocholine: 1, 2-dimyristoyl-sn-glycero-3-phosphocholine: cholesterol) released more than 90% of protein content after 240 h.
Collapse
Affiliation(s)
- M Kazemi
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht, Iran
| | - R Madani
- Department of Pathobiology, Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
- Department of Proteomics and Biochemistry, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - M R Aghamaali
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht, Iran
| | - T Emami
- Department of Proteomics and Biochemistry, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - F Golchinfar
- Department of Proteomics and Biochemistry, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - L Heshmati
- Department of Medical Nanotechnology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
34
|
Thapa Magar K, Boafo GF, Li X, Chen Z, He W. Liposome-based delivery of biological drugs. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2021.08.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
35
|
Karczmarzyk K, Kęsik-Brodacka M. Attacking the Intruder at the Gate: Prospects of Mucosal Anti SARS-CoV-2 Vaccines. Pathogens 2022; 11:pathogens11020117. [PMID: 35215061 PMCID: PMC8876505 DOI: 10.3390/pathogens11020117] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/04/2022] [Accepted: 01/14/2022] [Indexed: 12/13/2022] Open
Abstract
The sudden outbreak of the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) pandemic in December 2019 caused crises and health emergencies worldwide. The rapid spread of the virus created an urgent need for the development of an effective vaccine and mass immunization to achieve herd immunity. Efforts of scientific teams at universities and pharmaceutical companies around the world allowed for the development of various types of preparations and made it possible to start the vaccination process. However, it appears that the developed vaccines are not effective enough and do not guarantee long-lasting immunity, especially for new variants of SARS-CoV-2. Considering this problem, it is promising to focus on developing a Coronavirus Disease 2019 (COVID-19) mucosal vaccine. Such a preparation applied directly to the mucous membranes of the upper respiratory tract might provide an immune barrier at the primary point of virus entry into the human body while inducing systemic immunity. A number of such preparations against SARS-CoV-2 are already in various phases of preclinical and clinical trials, and several of them are very close to being accepted for general use, constituting a milestone toward pandemic containment.
Collapse
Affiliation(s)
- Kacper Karczmarzyk
- Department of Bacterial Genetics, Institute of Microbiology, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland
- Correspondence:
| | | |
Collapse
|
36
|
Attia MA, Essa EA, Elebyary TT, Faheem AM, Elkordy AA. Brief on Recent Application of Liposomal Vaccines for Lower Respiratory Tract Viral Infections: From Influenza to COVID-19 Vaccines. Pharmaceuticals (Basel) 2021; 14:1173. [PMID: 34832955 PMCID: PMC8619292 DOI: 10.3390/ph14111173] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/12/2021] [Accepted: 11/14/2021] [Indexed: 12/11/2022] Open
Abstract
Vaccination is the most effective means of preventing infectious diseases and saving lives. Modern biotechnology largely enabled vaccine development. In the meantime, recent advances in pharmaceutical technology have resulted in the emergence of nanoparticles that are extensively investigated as promising miniaturized drug delivery systems. Scientists are particularly interested in liposomes as an important carrier for vaccine development. Wide acceptability of liposomes lies in their flexibility and versatility. Due to their unique vesicular structure with alternating aqueous and lipid compartments, liposomes can enclose both hydrophilic and lipophilic compounds, including antigens. Liposome composition can be tailored to obtain the desired immune response and adjuvant characteristics. During the current pandemic of COVID-19, many liposome-based vaccines have been developed with great success. This review covers a liposome-based vaccine designed particularly to combat viral infection of the lower respiratory tract (LRT), i.e., infection of the lung, specifically in the lower airways. Viruses such as influenza, respiratory syncytial virus (RSV), severe acute respiratory syndrome (SARS-CoV-1 and SARS-CoV-2) are common causes of LRT infections, hence this review mainly focuses on this category of viruses.
Collapse
Affiliation(s)
- Mohamed Ahmed Attia
- School of Pharmacy and Pharmaceutical Sciences, Faculty of Health Sciences and Wellbeing, University of Sunderland, Sunderland SR1 3SD, UK; (M.A.A.); (A.M.F.)
| | - Ebtessam Ahmed Essa
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Tanta University, Tanta 31511, Egypt; (E.A.E.); (T.T.E.)
| | - Toka Tarek Elebyary
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Tanta University, Tanta 31511, Egypt; (E.A.E.); (T.T.E.)
| | - Ahmed Mostafa Faheem
- School of Pharmacy and Pharmaceutical Sciences, Faculty of Health Sciences and Wellbeing, University of Sunderland, Sunderland SR1 3SD, UK; (M.A.A.); (A.M.F.)
| | - Amal Ali Elkordy
- School of Pharmacy and Pharmaceutical Sciences, Faculty of Health Sciences and Wellbeing, University of Sunderland, Sunderland SR1 3SD, UK; (M.A.A.); (A.M.F.)
| |
Collapse
|
37
|
Volynsky PE, Smirnova AI, Akimov SA, Sokolov SS, Severin FF. The Membrane-Water Partition Coefficients of Antifungal, but Not Antibacterial, Membrane-Active Compounds Are Similar. Front Microbiol 2021; 12:756408. [PMID: 34803981 PMCID: PMC8602886 DOI: 10.3389/fmicb.2021.756408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 10/12/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Pavel E. Volynsky
- Laboratory of Biomolecular Modeling, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences (RAS), Moscow, Russia
| | - Alexandra I. Smirnova
- Department of Molecular Energetics of Microorganisms, Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
| | - Sergey A. Akimov
- Laboratory of Bioelectrochemistry, A.N. Frumkin Institute of Physical Chemistry and Electrochemistry, Russian Academy of Sciences (RAS), Moscow, Russia
| | - Svyatoslav S. Sokolov
- Department of Molecular Energetics of Microorganisms, Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Fedor F. Severin
- Department of Molecular Energetics of Microorganisms, Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
38
|
Koo BI, Jin S, Kim H, Lee DJ, Lee E, Nam YS. Conjugation-Free Multilamellar Protein-Lipid Hybrid Vesicles for Multifaceted Immune Responses. Adv Healthc Mater 2021; 10:e2101239. [PMID: 34467659 DOI: 10.1002/adhm.202101239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/04/2021] [Indexed: 12/11/2022]
Abstract
Various lipid-based nanocarriers have been developed for the co-delivery of protein antigens with immunological adjuvants. However, their in vivo potency in vaccine delivery is limited by structural instability, which causes off-target delivery and low cross-presentation efficacies. Recent works employ covalent cross-linking to stabilize the lipid nanostructures, though the immunogenicity and side effects of chemically modified protein antigens and lipids can cause a long-lasting safety issue. Here robust "conjugation-free" multilamellar protein antigen-lipid hybrid nanovesicles (MPLVs) are introduced through the antigen-mediated self-assembly of unilamellar lipid vesicles for the co-delivery of protein antigens and immunologic adjuvants. The nanocarriers coated with monophosphoryl lipid A and hyaluronic acids elicit highly increase antigen-specific immune responses in vitro and in vivo. The MPLVs increase the generation of immunological surface markers and cytokines in mouse-derived bone-marrow dendritic cells compared to soluble antigens with adjuvants. Besides, the vaccination of mice with the MPLVs significantly increase the production of anti-antigen antibody and interferon-gamma via the activation of CD4+ and CD8+ T cells, respectively. These findings suggest that MPLVs can serve as a promising nanovaccine delivery platform for efficient antigen cross-presentation through the efficient co-delivery of protein antigens with adjuvants.
Collapse
Affiliation(s)
- Bon Il Koo
- Department of Materials Science and Engineering Korea Advanced Institute of Science and Technology 291 Daehak‐ro, Yuseong‐gu Daejeon 34141 Republic of Korea
| | - Seon‐Mi Jin
- School of Materials Science and Engineering Gwangju Institute of Science and Technology 123 Cheomdan‐gwagiro Gwangju 61005 Republic of Korea
| | - Hayeon Kim
- School of Materials Science and Engineering Gwangju Institute of Science and Technology 123 Cheomdan‐gwagiro Gwangju 61005 Republic of Korea
| | - Dong Jae Lee
- Department of Materials Science and Engineering Korea Advanced Institute of Science and Technology 291 Daehak‐ro, Yuseong‐gu Daejeon 34141 Republic of Korea
| | - Eunji Lee
- School of Materials Science and Engineering Gwangju Institute of Science and Technology 123 Cheomdan‐gwagiro Gwangju 61005 Republic of Korea
| | - Yoon Sung Nam
- Department of Materials Science and Engineering Korea Advanced Institute of Science and Technology 291 Daehak‐ro, Yuseong‐gu Daejeon 34141 Republic of Korea
- KAIST Institute for NanoCentury Korea Advanced Institute of Science and Technology 291 Daehak‐ro, Yuseong‐gu Daejeon 34141 Republic of Korea
- KAIST Institute for Health Science and Technology Korea Advanced Institute of Science and Technology 291 Daehak‐ro, Yuseong‐gu Daejeon 34141 Republic of Korea
| |
Collapse
|
39
|
Heshmati L, Rezayat SM, Madani R, Emami T, Jafari MR, Golchinfar F, Kazemi M, Azimi Dezfouli SM. Immunity Evaluation of an Experimental Designed Nanoliposomal Vaccine Containing FMDV Immunodominant Peptides. ARCHIVES OF RAZI INSTITUTE 2021; 76:1183-1190. [PMID: 35355777 PMCID: PMC8934104 DOI: 10.22092/ari.2021.352498.1566] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/06/2021] [Indexed: 06/14/2023]
Abstract
Foot-and-mouth disease (FMD) is a highly contagious viral disease affecting cloven-hoofed animals. The particular virus causing FMD disease is called FMD virus and is a member of the Aphthovirus genus in the Picornaviridae family. The FMD virus has an 8500 nt long single strain positive RNA genome with one open reading frame (ORF) trapped in an icosahedral capsid protein. This virus genome doesn't have proofreading property which leads to high mutagenesis. It has seven serotypes, including O, A, ASIA, SAT1, SAT2, and C serotypes, as well as many subtypes. Iran is an endemic region for foot-and-mouth disease. Vaccination of susceptible animals with an inactivated whole-virus vaccine is the only way to control the epidemic in many developing countries. Today, conventionally attenuated and killed virus vaccines are being used worldwide. In Iran, animals have been vaccinated every 105 days with an inactivated FMD vaccine. Although commercially available FMD vaccines are effective, they provide short-term immunity requiring regular boosters. A new FMD vaccine is needed to improve immunization, safety, and long-term immune responses. A synthetic peptide vaccine is one of the safe and important vaccines. Peptide vaccine has low immunogenicity, requiring strong adjuvants. Nanoliposomes can be used as new adjuvants to improve immune response. In the current study, nanoliposomal carriers were selected using Dimyristoylphosphatidylcholine (DMPC), dimyristoyl phosphoglycerol (DMPG), and Cholesterol (Chol) as an adjuvant containing two immunodominant synthetic FMDV peptides. The liposomal formulations were characterized by various physicochemical properties. The size, zeta potential, and encapsulation efficiency were optimized, and the obtained nanoliposome was suitable as a vaccine. The efficacy of vaccines has been evaluated in guinea pigs as animal models. Indirect ELISA was used to detect FMDV-specific IgG. The obtained results indicated that although antibody titer was observed, the amount was lower compared to the groups that received inactivated virus-containing liposomes. In addition, the results showed that liposome was an appropriate adjuvant, compared to other adjuvants, such as Alum and Freund, and can act as a depot and induce an immune response.
Collapse
Affiliation(s)
- L Heshmati
- Department of Medical Nanotechnology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - S M Rezayat
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Pharmaceutical Sciences Branch, Islamic Azad University of Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Nanotechnology in Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - R Madani
- Department of Pathobiology, Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
- Department of Proteomics and Biochemistry, Razi Vaccine and Serum Research Institute, Agricultural Research Education and Extension Organization (AREEO), Karaj, Iran
| | - T Emami
- Department of Proteomics and Biochemistry, Razi Vaccine and Serum Research Institute, Agricultural Research Education and Extension Organization (AREEO), Karaj, Iran
| | - M R Jafari
- 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
| | - F Golchinfar
- Department of Proteomics and Biochemistry, Razi Vaccine and Serum Research Institute, Agricultural Research Education and Extension Organization (AREEO), Karaj, Iran
| | - M Kazemi
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht, Iran
| | - S M Azimi Dezfouli
- Department of Foot and Mouth Vaccine Production, Razi Vaccine and Serum Research Institute, Karaj, Iran
| |
Collapse
|
40
|
Abstract
INTRODUCTION Vaccination is so far the most effective way of eradicating infections. Rapidly emerging drug resistance against infectious diseases and chemotherapy-related toxicities in cancer warrant immediate vaccine development to save mankind. Subunit vaccines alone, however, fail to elicit sufficiently strong and long-lasting protective immunity against deadly pathogens. Nanoparticle (NP)-based delivery vehicles like microemulsions, liposomes, virosomes, nanogels, micelles and dendrimers offer promising strategies to overcome limitations of traditional vaccine adjuvants. Nanovaccines can improve targeted delivery, antigen presentation, stimulation of body's innate immunity, strong T cell response combined with safety to combat infectious diseases and cancers. Further, nanovaccines can be highly beneficial to generate effective immutherapeutic formulations against cancer. AREAS COVERED This review summarizes the emerging nanoparticle strategies highlighting their success and challenges in preclinical and clinical trials in infectious diseases and cancer. It provides a concise overview of current nanoparticle-based vaccines, their adjuvant potential and their cellular delivery mechanisms. EXPERT OPINION The nanovaccines (50-250 nm in size) are most efficient in terms of tissue targeting, prolonged circulation and preferential uptake by the professional APCs chiefly due to their small size. More rational designing, improved antigen loading, extensive functionalization and targeted delivery are some of the future goals of ideal nanovaccines.
Collapse
Affiliation(s)
- Amrita Das
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Nahid Ali
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
41
|
Christodoulides M, Humbert MV, Heckels JE. The potential utility of liposomes for Neisseria vaccines. Expert Rev Vaccines 2021; 20:1235-1256. [PMID: 34524062 DOI: 10.1080/14760584.2021.1981865] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Species of the genus Neisseria are important global pathogens. Neisseria gonorrhoeae (gonococcus) causes the sexually transmitted disease gonorrhea and Neisseria meningitidis (meningococcus) causes meningitis and sepsis. Liposomes are self-assembled spheres of phospholipid bilayers enclosing a central aqueous space, and they have attracted much interest and use as a delivery vehicle for Neisseria vaccine antigens. AREAS COVERED A brief background on Neisseria infections and the success of licensed meningococcal vaccines are provided. The absence of a gonococcal vaccine is highlighted. The use of liposomes for delivering Neisseria antigens and adjuvants, for the purposes of generating specific immune responses, is reviewed. The use of other lipid-based systems for antigen and adjuvant delivery is examined briefly. EXPERT OPINION With renewed interest in developing a gonococcal vaccine, liposomes remain an attractive option for delivering antigens. The discipline of nanotechnology provides additional nanoparticle-based options for gonococcal vaccine development. Future work would be needed to tailor the composition of liposomes and other nanoparticles to the specific vaccine antigen(s), in order to generate optimal anti-gonococcal immune responses. The potential use of liposomes and other nanoparticles to deliver anti-gonococcal compounds to treat infections also should be explored further.
Collapse
Affiliation(s)
- Myron Christodoulides
- Neisseria Research Group, Molecular Microbiology, School of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, UK
| | - Maria Victoria Humbert
- Neisseria Research Group, Molecular Microbiology, School of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, UK
| | - John E Heckels
- Neisseria Research Group, Molecular Microbiology, School of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, UK
| |
Collapse
|
42
|
The importance of nanoparticle physicochemical characterization for immunology research: What we learned and what we still need to understand. Adv Drug Deliv Rev 2021; 176:113897. [PMID: 34314786 DOI: 10.1016/j.addr.2021.113897] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/02/2021] [Accepted: 07/23/2021] [Indexed: 02/08/2023]
Abstract
Physicochemical characterization of nanoparticles intended for immunology research is important as it helps explain the observed immunological effects. More importantly, it relates the physicochemical properties with the immunological properties to draw meaningful conclusions. There are many physicochemical parameters, with each having numerous analytical techniques and instrumentation to measure them. Thus, where to begin can be challenging even for the experienced scientist. This paper aims to provide guidance to the immunology scientist on how best to characterize their nanoparticles. A step-by-step guide for the physicochemical characterization of liposomal formulations, based on the FDA's guidance for industry for Liposome Drug Products, is provided. Eight critical quality attributes have been identified and for each, the methodology and the physicochemical questions one should consider are discussed. This chapter also addresses common physicochemical characterization mistakes and concludes with a perspective on the type of measurements needed to address current physicochemical characterization gaps and challenges.
Collapse
|
43
|
Chatzikleanthous D, O'Hagan DT, Adamo R. Lipid-Based Nanoparticles for Delivery of Vaccine Adjuvants and Antigens: Toward Multicomponent Vaccines. Mol Pharm 2021; 18:2867-2888. [PMID: 34264684 DOI: 10.1021/acs.molpharmaceut.1c00447] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Despite the many advances that have occurred in the field of vaccine adjuvants, there are still unmet needs that may enable the development of vaccines suitable for more challenging pathogens (e.g., HIV and tuberculosis) and for cancer vaccines. Liposomes have already been shown to be highly effective as adjuvant/delivery systems due to their versatility and likely will find further uses in this space. The broad potential of lipid-based delivery systems is highlighted by the recent approval of COVID-19 vaccines comprising lipid nanoparticles with encapsulated mRNA. This review provides an overview of the different approaches that can be evaluated for the design of lipid-based vaccine adjuvant/delivery systems for protein, carbohydrate, and nucleic acid-based antigens and how these strategies might be combined to develop multicomponent vaccines.
Collapse
Affiliation(s)
- Despo Chatzikleanthous
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, G4 0RE Glasgow, U.K.,GSK, Via Fiorentina 1, 53100 Siena, Italy
| | | | | |
Collapse
|
44
|
Stephens AJ, Burgess-Brown NA, Jiang S. Beyond Just Peptide Antigens: The Complex World of Peptide-Based Cancer Vaccines. Front Immunol 2021; 12:696791. [PMID: 34276688 PMCID: PMC8279810 DOI: 10.3389/fimmu.2021.696791] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 06/17/2021] [Indexed: 12/14/2022] Open
Abstract
Peptide-based cancer vaccines rely upon the strong activation of the adaptive immune response to elicit its effector function. They have shown to be highly specific and safe, but have yet to prove themselves as an efficacious treatment for cancer in the clinic. This is for a variety of reasons, including tumour heterogeneity, self-tolerance, and immune suppression. Importance has been placed on the overall design of peptide-based cancer vaccines, which have evolved from simple peptide derivatives of a cancer antigen, to complex drugs; incorporating overlapping regions, conjugates, and delivery systems to target and stimulate different components of antigen presenting cells, and to bolster antigen cross-presentation. Peptide-based cancer vaccines are increasingly becoming more personalised to an individual's tumour antigen repertoire and are often combined with existing cancer treatments. This strategy ultimately aids in combating the shortcomings of a more generalised vaccine strategy and provides a comprehensive treatment, taking into consideration cancer cell variability and its ability to avoid immune interrogation.
Collapse
Affiliation(s)
- Alexander J Stephens
- Department of Oncology, Medical Sciences Division, University of Oxford, Oxford, United Kingdom.,Centre for Medicines Discovery, Nuffield Department of Medicine, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Nicola A Burgess-Brown
- Centre for Medicines Discovery, Nuffield Department of Medicine, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Shisong Jiang
- Department of Oncology, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
45
|
Alghuthaymi MA, Hassan AA, Kalia A, Sayed El Ahl RMH, El Hamaky AAM, Oleksak P, Kuca K, Abd-Elsalam KA. Antifungal Nano-Therapy in Veterinary Medicine: Current Status and Future Prospects. J Fungi (Basel) 2021; 7:494. [PMID: 34206304 PMCID: PMC8303737 DOI: 10.3390/jof7070494] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/15/2021] [Accepted: 06/16/2021] [Indexed: 12/15/2022] Open
Abstract
The global recognition for the potential of nanoproducts and processes in human biomedicine has given impetus for the development of novel strategies for rapid, reliable, and proficient diagnosis, prevention, and control of animal diseases. Nanomaterials exhibit significant antifungal and antimycotoxin activities against mycosis and mycotoxicosis disorders in animals, as evidenced through reports published over the recent decade and more. These nanoantifungals can be potentially utilized for the development of a variety of products of pharmaceutical and biomedical significance including the nano-scale vaccines, adjuvants, anticancer and gene therapy systems, farm disinfectants, animal husbandry, and nutritional products. This review will provide details on the therapeutic and preventative aspects of nanoantifungals against diverse fungal and mycotoxin-related diseases in animals. The predominant mechanisms of action of these nanoantifungals and their potential as antifungal and cytotoxicity-causing agents will also be illustrated. Also, the other theragnostic applications of nanoantifungals in veterinary medicine will be identified.
Collapse
Affiliation(s)
- Mousa A. Alghuthaymi
- Biology Department, Science and Humanities College, Shaqra University, Alquwayiyah 19245, Saudi Arabia;
| | - Atef A. Hassan
- Department of Mycology, Animal Health Research Institute (AHRI), Agriculture Research Center (ARC), 12611 Giza, Egypt; (A.A.H.); (R.M.H.S.E.A.); (A.A.M.E.H.)
| | - Anu Kalia
- Electron Microscopy and Nanoscience Laboratory, Department of Soil Science, College of Agriculture, Punjab Agricultural University, Ludhiana 141004, India
| | - Rasha M. H. Sayed El Ahl
- Department of Mycology, Animal Health Research Institute (AHRI), Agriculture Research Center (ARC), 12611 Giza, Egypt; (A.A.H.); (R.M.H.S.E.A.); (A.A.M.E.H.)
| | - Ahmed A. M. El Hamaky
- Department of Mycology, Animal Health Research Institute (AHRI), Agriculture Research Center (ARC), 12611 Giza, Egypt; (A.A.H.); (R.M.H.S.E.A.); (A.A.M.E.H.)
| | - Patrik Oleksak
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, 50003 Hradec Kralove, Czech Republic;
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, 50003 Hradec Kralove, Czech Republic;
| | - Kamel A. Abd-Elsalam
- Plant Pathology Research Institute, Agricultural Research Center (ARC), 9-Gamaa St., 12619 Giza, Egypt
| |
Collapse
|
46
|
Ho HM, Huang CY, Cheng YJ, Chen IH, Liu SJ, Huang CH, Huang MH. Squalene nanoemulsion reinforces mucosal and immunological fingerprints following intravaginal delivery. Biomed Pharmacother 2021; 141:111799. [PMID: 34098215 DOI: 10.1016/j.biopha.2021.111799] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 05/25/2021] [Accepted: 06/01/2021] [Indexed: 01/03/2023] Open
Abstract
This study describes the assessment of mucosal adjuvant activity of a squalene-based nanoemulsion (SQ@NE) following intravaginal delivery in mice. After immunization, a high level of recruitment of CD11b/c+ granulocytes and F4/80+ macrophages was observed in the vaginal mucosal tissues of the mice immunized with a model protein ovalbumin (OVA) formulated with SQ@NE, and then downstream regulated the expression of MHC II and costimulatory molecules CD40 and CD86 on CD11c+ cells harvested from the associated draining lymph node. With respect to cytotoxic T lymphocyte immunity, the mice immunized with SQ@NE-formulated OVA elicited a high population of OVA-specific CD8+ cells in the spleen and increased the secretion of IFN-γ, IL-2 and IL-17 from OVA-restimulated splenocytes compared with those immunized with OVA alone. By studying in vivo fluorescence imaging and B-cell immunoassays, we discovered how SQ@NE prolongs the retention of antigen depots at the mucosal membrane of the immune inductive site and allows them to properly drive the production of antibodies. The data demonstrated that SQ@NE prolonged fluorescence-labeled OVA retention at the genital tract and augmented the production of OVA-specific IgG in sera and IgA in vaginal washes. These results indicate that SQ@NE is a promising vaginal adjuvant for the induction of both mucosal and systemic immune responses, a feature that provides implications for the development of a mucosal vaccine against genital infections and sexually transmitted diseases.
Collapse
Affiliation(s)
- Hui-Min Ho
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Chiung-Yi Huang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Yu-Jhen Cheng
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
| | - I-Hua Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Shih-Jen Liu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan; Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan; Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Chung-Hsiung Huang
- Department of Food Science, National Taiwan Ocean University, Keelung 20224, Taiwan.
| | - Ming-Hsi Huang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan; Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan; Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| |
Collapse
|
47
|
Reactive Deep Eutectic Solvents (RDESs): A New Tool for Phospholipase D-Catalyzed Preparation of Phospholipids. Catalysts 2021. [DOI: 10.3390/catal11060655] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The use of Reactive Deep Eutectic Solvents (RDESs) in the preparation of polar head modified phospholipids (PLs) with phospholipase D (PLD)-catalyzed biotransformations has been investigated. Natural phosphatidylcholine (PC) has been submitted to PLD-catalyzed transphosphatidylations using a new reaction medium composed by a mixture of RDES/buffer. Instead of exploiting deep eutectic solvents conventionally, just as the reaction media, these solvents have been designed here in order to contribute actively to the synthetic processes by participating as reagents. RDESs were prepared using choline chloride or trimethyl glycine as hydrogen-bond acceptors and glycerol or ethylene glycol, as hydrogen-bond donors as well as nucleophiles for choline substitution. Specifically designed RDES/buffer reaction media allowed the obtainment of PLs with optimized yields in the perspective of a sustainable process implementation.
Collapse
|
48
|
Serag E, El-Zeftawy M. Environmental aspect and applications of nanotechnology to eliminate COVID-19 epidemiology risk. NANOTECHNOLOGY FOR ENVIRONMENTAL ENGINEERING 2021. [PMCID: PMC7917956 DOI: 10.1007/s41204-021-00108-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Herein, we discuss fast development of the new coronavirus disease COVID-19, emerged in late 2019 in Wuhan, Hubei Province, China, the ground zero of the coronavirus pandemic, and associated with relatively high mortality rate. COVID-19 risk originates from its ability to transmit easily from person to person through the respiratory droplets released during sneezing, breathing, talking, singing, or coughing within a range of nearly 1.5–2 m. The review begins with an overview of COVID-19 origin and symptoms that range from common cold to severe respiratory illnesses and death. Then, it sheds light on the role of nanotechnology as an effective tool for fighting COVID-19 via contributions in diagnosis, treatment, and manufacture of protective equipment for people and healthcare workers. Emergency-approved therapeutics for clinical trial and prospective vaccines are discussed. Additionally, the present work addresses the risk of severe acute respiratory syndrome coronavirus transmission via wastewater and means of wastewater treatment and disinfection via nanoscale materials. The review concludes with a brief assessment of the government's efforts and contemporary propositions to minimize COVID-19 hazard and spreading.
Collapse
Affiliation(s)
- Eman Serag
- Marine Pollution Department, Environmental Division, National Institute of Oceanography and Fisheries, Kayet Bey, Elanfoushy, Alexandria, Egypt
| | - Marwa El-Zeftawy
- Biochemistry Department, Faculty of Veterinary Medicine, New Valley University, El-Kharga, New Valley Egypt
- Biological Screening and Preclinical Trial Lab, Biochemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| |
Collapse
|
49
|
Cationic Nanoparticle-Based Cancer Vaccines. Pharmaceutics 2021; 13:pharmaceutics13050596. [PMID: 33919378 PMCID: PMC8143365 DOI: 10.3390/pharmaceutics13050596] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/14/2021] [Accepted: 04/17/2021] [Indexed: 12/15/2022] Open
Abstract
Cationic nanoparticles have been shown to be surprisingly effective as cancer vaccine vehicles in preclinical and clinical studies. Cationic nanoparticles deliver tumor-associated antigens to dendritic cells and induce immune activation, resulting in strong antigen-specific cellular immune responses, as shown for a wide variety of vaccine candidates. In this review, we discuss the relation between the cationic nature of nanoparticles and the efficacy of cancer immunotherapy. Multiple types of lipid- and polymer-based cationic nanoparticulate cancer vaccines with various antigen types (e.g., mRNA, DNA, peptides and proteins) and adjuvants are described. Furthermore, we focus on the types of cationic nanoparticles used for T-cell induction, especially in the context of therapeutic cancer vaccination. We discuss different cationic nanoparticulate vaccines, molecular mechanisms of adjuvanticity and biodistribution profiles upon administration via different routes. Finally, we discuss the perspectives of cationic nanoparticulate vaccines for improving immunotherapy of cancer.
Collapse
|
50
|
Kumari S, Chatterjee K. Biomaterials-based formulations and surfaces to combat viral infectious diseases. APL Bioeng 2021; 5:011503. [PMID: 33598595 PMCID: PMC7881627 DOI: 10.1063/5.0029486] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 12/28/2020] [Indexed: 12/13/2022] Open
Abstract
Rapidly growing viral infections are potent risks to public health worldwide. Accessible virus-specific antiviral vaccines and drugs are therapeutically inert to emerging viruses, such as Zika, Ebola, and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Therefore, discovering ways to prevent and control viral infections is among the foremost medical challenge of our time. Recently, innovative technologies are emerging that involve the development of new biomaterial-based formulations and surfaces endowed with broad-spectrum antiviral properties. Here, we review emerging biomaterials technologies for controlling viral infections. Relevant advances in biomaterials employed with nanotechnology to inactivate viruses or to inhibit virus replication and further their translation in safe and effective antiviral formulations in clinical trials are discussed. We have included antiviral approaches based on both organic and inorganic nanoparticles (NPs), which offer many advantages over molecular medicine. An insight into the development of immunomodulatory scaffolds in designing new platforms for personalized vaccines is also considered. Substantial research on natural products and herbal medicines and their potential in novel antiviral drugs are discussed. Furthermore, to control contagious viral infections, i.e., to reduce the viral load on surfaces, current strategies focusing on biomimetic anti-adhesive surfaces through nanostructured topography and hydrophobic surface modification techniques are introduced. Biomaterial surfaces functionalized with antimicrobial polymers and nanoparticles against viral infections are also discussed. We recognize the importance of research on antiviral biomaterials and present potential strategies for future directions in applying these biomaterial-based approaches to control viral infections and SARS-CoV-2.
Collapse
Affiliation(s)
- Sushma Kumari
- Department of Materials Engineering, Indian Institute of Science, Bangalore 560012, India
| | - Kaushik Chatterjee
- Department of Materials Engineering, Indian Institute of Science, Bangalore 560012, India
| |
Collapse
|