1
|
Li Y, Du B, Yu L, Luo H, Rong H, Gao X, Yin J. Strategies and challenges of cytosolic delivery of proteins. J Drug Target 2025; 33:837-852. [PMID: 39862226 DOI: 10.1080/1061186x.2025.2458616] [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: 12/09/2024] [Revised: 01/11/2025] [Accepted: 01/18/2025] [Indexed: 01/27/2025]
Abstract
The cytosolic delivery of therapeutic proteins represents a promising strategy for addressing diseases caused by protein dysfunction. Despite significant advances, efficient delivery remains challenging due to barriers such as cell membrane impermeability, endosomal sequestration and protein instability. This review summarises recent progress in protein delivery systems, including physical, chemical and biological approaches, with a particular focus on strategies that enhance endosomal escape and targeting specificity. We further discuss the clinical translatability of these approaches and propose future directions for improving delivery efficiency and safety, ultimately unlocking the therapeutic potential of intracellular proteins.
Collapse
Affiliation(s)
- Yuanyuan Li
- School of Life Science and Technology, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Baojie Du
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Lichao Yu
- School of Life Science and Technology, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Hong Luo
- School of Life Science and Technology, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Haibo Rong
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China
| | - Xiangdong Gao
- School of Life Science and Technology, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Jun Yin
- School of Life Science and Technology, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
2
|
Wang X, Tian N, He L, Yuan Z, Han L. Emerging Applications of Pickering Emulsions in Pharmaceutical Formulations: A Comprehensive Review. Int J Nanomedicine 2025; 20:5923-5947. [PMID: 40356863 PMCID: PMC12067652 DOI: 10.2147/ijn.s514928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
Over the past two decades, particle-stabilized Pickering emulsions (PEs) have emerged as a versatile platform in pharmaceutical formulations, demonstrating distinct advantages over surfactant-based systems through enhanced stability, reduced toxicity, and tunable interfacial properties. These systems exhibit unique drug delivery potential through their precisely controllable architecture, particularly in achieving spatiotemporal drug release patterns, tissue-specific targeting, and enhanced therapeutic payload encapsulation. In this review, the characteristics of PEs are first detailed, followed by an introduction to the main preparation methods and the key parameters for controlling the type, droplet size, and stability of PEs. The third section categorizes and discusses the advantages and disadvantages of various solid particles as emulsifiers. Lastly, emphasis is placed on the application of PEs in the pharmaceutical field, including functionalized designs and various administration routes to enlighten the rational design of PEs for effective drug delivery.
Collapse
Affiliation(s)
- Xingyue Wang
- College of Pharmacy, Southwest Minzu University, Chengdu, 610041, People’s Republic of China
| | - Na Tian
- College of Pharmacy, Southwest Minzu University, Chengdu, 610041, People’s Republic of China
| | - Lili He
- College of Pharmacy, Southwest Minzu University, Chengdu, 610041, People’s Republic of China
| | - Zhixiang Yuan
- College of Pharmacy, Southwest Minzu University, Chengdu, 610041, People’s Republic of China
| | - Lu Han
- College of Pharmacy, Southwest Minzu University, Chengdu, 610041, People’s Republic of China
| |
Collapse
|
3
|
Ye T, Chen Q, Yang S, Liu H, Zhou Y, Feng Y, Li J, Wang Y, Yu G, Dai J. Host-guest interfacial recognition of alginate-based β-cyclodextrin/dendrobine supra-amphiphiles reinforced the physicochemical stability and sustained-release properties of Pickering emulsions. Int J Biol Macromol 2025; 306:141746. [PMID: 40049470 DOI: 10.1016/j.ijbiomac.2025.141746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/26/2025] [Accepted: 03/03/2025] [Indexed: 03/10/2025]
Abstract
Achieving high bioavailability of dendrobine (DDB) necessitates the development of simplified available and efficient delivery systems. Pickering emulsions (PEs) derived from biomass represent a promising option. However, the physicochemical properties of PEs interfacial films were insufficient to prevent DDB leakage, thereby reducing bioavailability. Herein, a supramolecular host-guest interfacial recognition strategy was proposed in-situ between amphipathic sodium alginate-functionalized cyclodextrin (SAE-CD) and hydrophobic DDB at oil-water interface, further forming the SAE-based supra-amphiphiles to efficient stabilize the high internal phase Pickering emulsions (HIPPEs) with gel-like characteristics. A multiscale methodology was empolyed to investigate the interfacial assembly behavior and emulsification properties of supra-amphiphilic SAE-CD/DDB interfacial system, focusing on molecular interactions, interfacial adsorption, and overall stability. Notably, the SAE-CD/DDB-based supramolecular assembly/disassembly behaviors could be self-adjusted for regulating the aggregation particle size and thickness of interfacial self-assembled films. The SAE-CD/DDB co-stabilized HIPPEs exhibited favorable drug release capabilities, enabling sustained effects of DDB. Overall, the SAE-CD/DDB co-stabilized HIPPEs demonstrated excellent properties in terms of stability, drug loading capacity, and sustained release performance, highlighting their potential for in oral delivery and sustained-release systems.
Collapse
Affiliation(s)
- Tong Ye
- School of Pharmacy, Anhui University of Chinese Medicine, 350 Longzihu Road, Hefei 230012, Anhui Province, China
| | - Qile Chen
- School of Pharmacy, Anhui University of Chinese Medicine, 350 Longzihu Road, Hefei 230012, Anhui Province, China
| | - Shujuan Yang
- School of Pharmacy, Anhui University of Chinese Medicine, 350 Longzihu Road, Hefei 230012, Anhui Province, China
| | - Haifang Liu
- School of Pharmacy, Anhui University of Chinese Medicine, 350 Longzihu Road, Hefei 230012, Anhui Province, China
| | - Yan Zhou
- School of Pharmacy, Anhui University of Chinese Medicine, 350 Longzihu Road, Hefei 230012, Anhui Province, China
| | - Yuhong Feng
- School of Pharmacy, Anhui University of Chinese Medicine, 350 Longzihu Road, Hefei 230012, Anhui Province, China
| | - Jiacheng Li
- School of Pharmacy, Anhui University of Chinese Medicine, 350 Longzihu Road, Hefei 230012, Anhui Province, China
| | - Yujuan Wang
- School of Pharmacy, Anhui University of Chinese Medicine, 350 Longzihu Road, Hefei 230012, Anhui Province, China
| | - Gaobo Yu
- School of Pharmacy, Anhui University of Chinese Medicine, 350 Longzihu Road, Hefei 230012, Anhui Province, China.
| | - Jun Dai
- School of Pharmacy, Anhui University of Chinese Medicine, 350 Longzihu Road, Hefei 230012, Anhui Province, China.
| |
Collapse
|
4
|
Peng D, Cheng L, Tang J, Liu Z, Xue Y, Liu J. Engineered NK Exosomes Captured Antigens In Situ for Enhanced Tumor Immunotherapy. ACS APPLIED MATERIALS & INTERFACES 2025; 17:23740-23752. [PMID: 40202388 DOI: 10.1021/acsami.5c03195] [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: 04/10/2025]
Abstract
Natural killer (NK) cells are widely involved in the field of tumor immunotherapy due to their unique killing ability. However, the durability and efficacy of NK-cell monotherapy are facing great challenges owing to the limitation of immunosuppressive tumor microenvironment (TME). NK cell-derived exosomes (Neo) not only play an innate immunomodulatory role similar to NK cells but also emerge as promising antitumor nanocarriers. In this study, an engineered Neo (R@Neo-MN) was designed that encapsulates the multifunctional antitumor drug (Raddeanin a, RA) and modified with maleimide (Mal, M) and mannose (Man, N). The obtained R@Neo-MN could not only exert NK cell-like antitumor function but also induce the immunogenic cell death of tumors to release tumor-associated antigens (TAAs). Furthermore, R@Neo-MN activated the cyclic guanosine monophosphate-adenosine monophosphate synthase/interferon gene stimulator (cGAS/STING) to release type I interferons (IFN). Then, R@Neo-MN could capture TAAs through Mal and specifically target dendritic cells (DCs) through Man, thereby promoting the maturation of DCs and enhancing tumor-specific cytotoxic T-cell (CTL)-mediated adaptive immunity. The released IFN further promoted the infiltration and activition of NK cells and CTLs at the tumor site. Our study suggested a novel strategy that harnesses both innate and adaptive immunity for enhanced tumor immunotherapy.
Collapse
Affiliation(s)
- Dan Peng
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Lili Cheng
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Junjie Tang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Zhuoyin Liu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Yifan Xue
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Jie Liu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| |
Collapse
|
5
|
Rosalina R, Weerapreeyakul N, Sutthanut K, Kamwilaisak K, Sakonsinsiri C. Nanocellulose-based Pickering emulsion of sesamolin manifested increased anticancer activity and necrosis in human colon cancer (HCT116) cells. Int J Biol Macromol 2025; 292:139225. [PMID: 39732237 DOI: 10.1016/j.ijbiomac.2024.139225] [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: 09/29/2024] [Revised: 12/16/2024] [Accepted: 12/24/2024] [Indexed: 12/30/2024]
Abstract
Sesamolin possesses limited aqueous solubility, a drawback for biological activity study in cancer cell models. This study aimed to enhance sesamolin's ability to fight cancer, as it is a bioactive compound with low water solubility found in sesame. We developed different Pickering emulsion delivery systems and tested their anticancer effects on various cancer cell types. Sesamolin was incorporated into either sesame or olive oil and subsequently formulated as oil in water (o/w) Pickering emulsions stabilized by the carboxylated cellulose nanocrystal (cCNC). The anticancer activity was determined based on cell viability and the induction of cell death mechanisms. The results demonstrated a synergistic effect of the components in the emulsion, including sesamolin, sesame oil, and olive oil, and a decrease in HCT116 viability in a concentration-dependent manner and selectively on cancer cells compared to non-cancerous Vero cells. The primary mode of cell death was predominantly ROS-induced necrosis, with no change in caspase 3/7 activity, indicating the absence of apoptosis. This study first presents the necrotic cell death mechanism induced by sesamolin. The findings reveal that the cCNC emulsion delivery system is safe and appropriate for transporting lipophilic chemicals and can overcome solubility limitations.
Collapse
Affiliation(s)
- Reny Rosalina
- Graduate School (Biomedical Sciences Program), Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Natthida Weerapreeyakul
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand; Human High Performance and Health Promotion Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand.
| | - Khaetthareeya Sutthanut
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand; Human High Performance and Health Promotion Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Khanita Kamwilaisak
- Department of Chemical Engineering, Faculty of Engineering, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Chadamas Sakonsinsiri
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
6
|
Gu P, Zhu Y, Xu P, Zhao Q, Zhao X, Zhao K, Wang X, Zhang W, Bao Y, Shi W. Poria cocos polysaccharide-loaded Alum Pickering emulsion as vaccine adjuvant to enhance immune responses. Colloids Surf B Biointerfaces 2024; 244:114144. [PMID: 39116600 DOI: 10.1016/j.colsurfb.2024.114144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 08/02/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024]
Abstract
Traditional Alum adjuvants mainly elicit a Th2 humoral immune response, but fail to generate a robust Th1 cellular immune response. However, the cellular immune response is essential for vaccination against cancer and a number of chronic infectious diseases, including human immunodeficiency virus infection and tuberculosis. In our previous study, we demonstrated that the polysaccharide from Poria cocos (PCP) has the potential to serve as an immunologic stimulant, enhancing both humoral and cellular immune responses. However, this effect was only observed at high concentrations. In this study, to enhance the immune-stimulation effect of PCP and modify the type of immune response elicited by Alum adjuvant, we successfully developed a Pickering emulsion delivery system (PCP-Al-Pickering) using PCP-loaded Alhydrogel particles as the stabilizer. After optimization, the Pickering emulsion exhibited excellent storage capacity and effectively adsorbed the PCP and antigen. As an adjuvant delivery system, the PCP-Al-Pickering emulsion facilitated the antigen uptake by macrophages, increased the recruitment of cells at injection sites, improved the activation of dendritic cells in draining lymph nodes, elicited a potent and durable antibody response, and promoted the activation of CD4+ and CD8+ T cells. Importantly, the PCP-Al-Pickering emulsion adjuvant elicited a balanced Th1 and Th2 immune response, in comparison to Alum adjuvant. The PCP-Al-Pickering emulsion may serve as a safe and promising adjuvant delivery system to enhance immune responses.
Collapse
Affiliation(s)
- Pengfei Gu
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Yixuan Zhu
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Panpan Xu
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Qi Zhao
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Xinghua Zhao
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Kuan Zhao
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Xiao Wang
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Wuchao Zhang
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Yongzhan Bao
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Wanyu Shi
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China.
| |
Collapse
|
7
|
Yan Y, Huang X, Yuan L, Ngai T, Ma G, Xia Y. Dictating the spatial-temporal delivery of molecular adjuvant and antigen for the enhanced vaccination. Biomaterials 2024; 311:122697. [PMID: 38968687 DOI: 10.1016/j.biomaterials.2024.122697] [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: 04/01/2024] [Revised: 06/24/2024] [Accepted: 06/30/2024] [Indexed: 07/07/2024]
Abstract
The incorporation of molecular adjuvants has revolutionized vaccine by boosting overall immune efficacy. While traditional efforts have been concentrated on the quality and quantity of vaccine components, the impact of adjuvant and antigen delivery kinetics on immunity remains to be fully understood. Here, we employed poly (lactic-co-glycolic acid) nanoparticle (PLGA NP) -stabilized Pickering emulsion (PPE) to refine the delivery kinetics of molecular adjuvant CpG and antigen, aiming to optimize immune responses. The hierarchical structure of PPE enabled spatially differential loading of CpG and antigen. The component inserted on the oil-water interphase exhibited a rapid release profile, while the one encapsulated in the PLGA NPs demonstrated a sustained release. This led to distinct intracellular spatial-temporal release kinetics. Compared to the PPE with sustained CpG release and burst release of antigen, we found that the PPE with rapid CpG release and sustained antigen release triggered an early and robust activation of Toll-like receptor 9 (TLR9) in direct way. This fostered a more immunogenic microenvironment, significantly outperforming the inverted delivery profile in dendritic cells (DCs) activation, resulting in higher CD40 expression, elevated proinflammatory cytokine levels, sustained antigen cross-presentation, an enhanced Th1 response, and increased CD8+ T cells. Moreover, prior exposure of CpG led to suppressed tumor growth and enhanced efficacy in Varicella-zoster virus (VZV) vaccine. Our findings underscore the importance of tuning adjuvant and antigen delivery kinetics in vaccine design, proposing a novel path for enhancing vaccination outcomes.
Collapse
Affiliation(s)
- Yumeng Yan
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100081, PR China; School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China; Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, PR China
| | - Xiaonan Huang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100081, PR China; School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China; Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, PR China; Sinovac Biotech Ltd., Beijing, 100085, PR China
| | - Lili Yuan
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, PR China
| | - To Ngai
- Department of Chemistry, The Chinese University of Hong Kong, Hong Kong, Shatin, N.T., PR China
| | - Guanghui Ma
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100081, PR China; School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China; Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, PR China
| | - Yufei Xia
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100081, PR China; School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China; Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, PR China.
| |
Collapse
|
8
|
Du Y, Lv J, Hao Z, Li Z, Song T, Ge H, Wang H, Yu Z, Xie Z, Li D, Liu Y. Pickering emulsion-guided monomeric delivery of monophosphoryl lipid A for enhanced vaccination. J Control Release 2024; 374:39-49. [PMID: 39111597 DOI: 10.1016/j.jconrel.2024.08.005] [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: 04/08/2024] [Revised: 07/31/2024] [Accepted: 08/03/2024] [Indexed: 08/11/2024]
Abstract
Immunological adjuvants are vaccine components that enhance long-lasting adaptive immune responses to weakly immunogenic antigens. Monophosphoryl lipid A (MPLA) is a potent and safe vaccine adjuvant that initiates an early innate immune response by binding to the Toll-like receptor 4 (TLR4). Importantly, the binding and recognition process is highly dependent on the monomeric state of MPLA. However, current vaccine delivery systems often prioritize improving the loading efficiency of MPLA, while neglecting the need to maintain its monomeric form for optimal immune activation. Here, we introduce a Pickering emulsion-guided MPLA monomeric delivery system (PMMS), which embed MPLA into the oil-water interface to achieve the monomeric loading of MPLA. During interactions with antigen-presenting cells, PMMS functions as a chaperone for MPLA, facilitating efficient recognition by TLR4 regardless of the presence of lipopolysaccharide-binding proteins. At the injection site, PMMS efficiently elicited local immune responses, subsequently promoting the migration of antigen-internalized dendritic cells to the lymph nodes. Within the draining lymph nodes, PMMS enhanced antigen presentation and maturation of dendritic cells. In C57BL/6 mice models, PMMS vaccination provoked potent antigen-specific CD8+ T cell-based immune responses. Additionally, PMMS demonstrated strong anti-tumor effects against E.G7-OVA lymphoma. These data indicate that PMMS provides a straightforward and efficient strategy for delivering monomeric MPLA to achieve robust cellular immune responses and effective cancer immunotherapy.
Collapse
Affiliation(s)
- Yiqun Du
- Joint Research Center for Food Nutrition and Health of IHM, School of Tea and Food Science & Technology, State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei 230036, PR China; Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen University, Shenzhen 518035, PR China.
| | - Jiali Lv
- Joint Research Center for Food Nutrition and Health of IHM, School of Tea and Food Science & Technology, State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei 230036, PR China
| | - Zongwei Hao
- Joint Research Center for Food Nutrition and Health of IHM, School of Tea and Food Science & Technology, State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei 230036, PR China
| | - Zhaofeng Li
- Joint Research Center for Food Nutrition and Health of IHM, School of Tea and Food Science & Technology, State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei 230036, PR China
| | - Tiantian Song
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Huifang Ge
- Joint Research Center for Food Nutrition and Health of IHM, School of Tea and Food Science & Technology, State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei 230036, PR China
| | - Hongyan Wang
- Joint Research Center for Food Nutrition and Health of IHM, School of Tea and Food Science & Technology, State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei 230036, PR China
| | - Zhenyu Yu
- Joint Research Center for Food Nutrition and Health of IHM, School of Tea and Food Science & Technology, State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei 230036, PR China
| | - Zhongwen Xie
- Joint Research Center for Food Nutrition and Health of IHM, School of Tea and Food Science & Technology, State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei 230036, PR China
| | - Daxiang Li
- Joint Research Center for Food Nutrition and Health of IHM, School of Tea and Food Science & Technology, State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei 230036, PR China
| | - Yuchen Liu
- Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen University, Shenzhen 518035, PR China.
| |
Collapse
|
9
|
Hsu JC, Liu P, Song Y, Song W, Saladin RJ, Peng Y, Hu S, Lan X, Cai W. Lymphoid organ-targeted nanomaterials for immunomodulation of cancer, inflammation, and beyond. Chem Soc Rev 2024; 53:7657-7680. [PMID: 38958009 PMCID: PMC11334694 DOI: 10.1039/d4cs00421c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
Nanomaterials exhibit significant potential for stimulating immune responses, offering both local and systemic modulation across a variety of diseases. The lymphoid organs, such as the spleen and lymph nodes, are home to various immune cells, including monocytes and dendritic cells, which contribute to both the progression and prevention/treatment of diseases. Consequently, many nanomaterial formulations are being rationally designed to target these organs and engage with specific cell types, thereby inducing therapeutic and protective effects. In this review, we explore crucial cellular interactions and processes involved in immune regulation and highlight innovative nano-based immunomodulatory approaches. We outline essential considerations in nanomaterial design with an emphasis on their impact on biological interactions, targeting capabilities, and treatment efficacy. Through selected examples, we illustrate the strategic targeting of therapeutically active nanomaterials to lymphoid organs and the subsequent immunomodulation for infection resistance, inflammation suppression, self-antigen tolerance, and cancer immunotherapy. Additionally, we address current challenges, discuss emerging topics, and share our outlook on future developments in the field.
Collapse
Affiliation(s)
- Jessica C Hsu
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| | - Peng Liu
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA.
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha 410008, P. R. China
- Key Laboratory of Biological Nanotechnology of National Health Commission, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, P. R. China
| | - Yangmeihui Song
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430073, P. R. China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430073, P. R. China
- Key Laboratory of Biological Targeted Therapy of the Ministry of Education, Wuhan 430073, P. R. China
| | - Wenyu Song
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA.
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430073, P. R. China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430073, P. R. China
- Key Laboratory of Biological Targeted Therapy of the Ministry of Education, Wuhan 430073, P. R. China
| | - Rachel J Saladin
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| | - Ying Peng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Shuo Hu
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha 410008, P. R. China
- Key Laboratory of Biological Nanotechnology of National Health Commission, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, P. R. China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430073, P. R. China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430073, P. R. China
- Key Laboratory of Biological Targeted Therapy of the Ministry of Education, Wuhan 430073, P. R. China
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| |
Collapse
|
10
|
Liu T, Yao W, Sun W, Yuan Y, Liu C, Liu X, Wang X, Jiang H. Components, Formulations, Deliveries, and Combinations of Tumor Vaccines. ACS NANO 2024; 18:18801-18833. [PMID: 38979917 DOI: 10.1021/acsnano.4c05065] [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: 07/10/2024]
Abstract
Tumor vaccines, an important part of immunotherapy, prevent cancer or kill existing tumor cells by activating or restoring the body's own immune system. Currently, various formulations of tumor vaccines have been developed, including cell vaccines, tumor cell membrane vaccines, tumor DNA vaccines, tumor mRNA vaccines, tumor polypeptide vaccines, virus-vectored tumor vaccines, and tumor-in-situ vaccines. There are also multiple delivery systems for tumor vaccines, such as liposomes, cell membrane vesicles, viruses, exosomes, and emulsions. In addition, to decrease the risk of tumor immune escape and immune tolerance that may exist with a single tumor vaccine, combination therapy of tumor vaccines with radiotherapy, chemotherapy, immune checkpoint inhibitors, cytokines, CAR-T therapy, or photoimmunotherapy is an effective strategy. Given the critical role of tumor vaccines in immunotherapy, here, we look back to the history of tumor vaccines, and we discuss the antigens, adjuvants, formulations, delivery systems, mechanisms, combination therapy, and future directions of tumor vaccines.
Collapse
Affiliation(s)
- Tengfei Liu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Wenyan Yao
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Wenyu Sun
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Yihan Yuan
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Chen Liu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Xiaohui Liu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Xuemei Wang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Hui Jiang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| |
Collapse
|
11
|
Qian W, Ye J, Xia S. DNA sensing of dendritic cells in cancer immunotherapy. Front Mol Biosci 2024; 11:1391046. [PMID: 38841190 PMCID: PMC11150630 DOI: 10.3389/fmolb.2024.1391046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 05/02/2024] [Indexed: 06/07/2024] Open
Abstract
Dendritic cells (DCs) are involved in the initiation and maintenance of immune responses against malignant cells by recognizing conserved pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs) through pattern recognition receptors (PRRs). According to recent studies, tumor cell-derived DNA molecules act as DAMPs and are recognized by DNA sensors in DCs. Once identified by sensors in DCs, these DNA molecules trigger multiple signaling cascades to promote various cytokines secretion, including type I IFN, and then to induce DCs mediated antitumor immunity. As one of the potential attractive strategies for cancer therapy, various agonists targeting DNA sensors are extensively explored including the combination with other cancer immunotherapies or the direct usage as major components of cancer vaccines. Moreover, this review highlights different mechanisms through which tumor-derived DNA initiates DCs activation and the mechanisms through which the tumor microenvironment regulates DNA sensing of DCs to promote tumor immune escape. The contributions of chemotherapy, radiotherapy, and checkpoint inhibitors in tumor therapy to the DNA sensing of DCs are also discussed. Finally, recent clinical progress in tumor therapy utilizing agonist-targeted DNA sensors is summarized. Indeed, understanding more about DNA sensing in DCs will help to understand more about tumor immunotherapy and improve the efficacy of DC-targeted treatment in cancer.
Collapse
Affiliation(s)
- Wei Qian
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Jun Ye
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
- The Center for Translational Medicine, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu, China
| | - Sheng Xia
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
12
|
Zha Y, Fu L, Liu Z, Lin J, Huang L. Construction of lymph nodes-targeting tumor vaccines by using the principle of DNA base complementary pairing to enhance anti-tumor cellular immune response. J Nanobiotechnology 2024; 22:230. [PMID: 38720322 PMCID: PMC11077755 DOI: 10.1186/s12951-024-02498-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 04/27/2024] [Indexed: 05/12/2024] Open
Abstract
Tumor vaccines, a crucial immunotherapy, have gained growing interest because of their unique capability to initiate precise anti-tumor immune responses and establish enduring immune memory. Injected tumor vaccines passively diffuse to the adjacent draining lymph nodes, where the residing antigen-presenting cells capture and present tumor antigens to T cells. This process represents the initial phase of the immune response to the tumor vaccines and constitutes a pivotal determinant of their effectiveness. Nevertheless, the granularity paradox, arising from the different requirements between the passive targeting delivery of tumor vaccines to lymph nodes and the uptake by antigen-presenting cells, diminishes the efficacy of lymph node-targeting tumor vaccines. This study addressed this challenge by employing a vaccine formulation with a tunable, controlled particle size. Manganese dioxide (MnO2) nanoparticles were synthesized, loaded with ovalbumin (OVA), and modified with A50 or T20 DNA single strands to obtain MnO2/OVA/A50 and MnO2/OVA/T20, respectively. Administering the vaccines sequentially, upon reaching the lymph nodes, the two vaccines converge and simultaneously aggregate into MnO2/OVA/A50-T20 particles through base pairing. This process enhances both vaccine uptake and antigen delivery. In vitro and in vivo studies demonstrated that, the combined vaccine, comprising MnO2/OVA/A50 and MnO2/OVA/T20, exhibited robust immunization effects and remarkable anti-tumor efficacy in the melanoma animal models. The strategy of controlling tumor vaccine size and consequently improving tumor antigen presentation efficiency and vaccine efficacy via the DNA base-pairing principle, provides novel concepts for the development of efficient tumor vaccines.
Collapse
Affiliation(s)
- Yongchao Zha
- Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Li Fu
- Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Zonghua Liu
- Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China.
| | - Jiansheng Lin
- Department of Anatomy, Hunan University of Chinese Medicine, Changsha, China.
| | - Linghong Huang
- Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
13
|
Li M, Yao H, Yi K, Lao YH, Shao D, Tao Y. Emerging nanoparticle platforms for CpG oligonucleotide delivery. Biomater Sci 2024; 12:2203-2228. [PMID: 38293828 DOI: 10.1039/d3bm01970e] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Unmethylated cytosine-phosphate-guanine (CpG) oligodeoxynucleotides (ODNs), which were therapeutic DNA with high immunostimulatory activity, have been applied in widespread applications from basic research to clinics as therapeutic agents for cancer immunotherapy, viral infection, allergic diseases and asthma since their discovery in 1995. The major factors to consider for clinical translation using CpG motifs are the protection of CpG ODNs from DNase degradation and the delivery of CpG ODNs to the Toll-like receptor-9 expressed human B-cells and plasmacytoid dendritic cells. Therefore, great efforts have been devoted to the advances of efficient delivery systems for CpG ODNs. In this review, we outline new horizons and recent developments in this field, providing a comprehensive summary of the nanoparticle-based CpG delivery systems developed to improve the efficacy of CpG-mediated immune responses, including DNA nanostructures, inorganic nanoparticles, polymer nanoparticles, metal-organic-frameworks, lipid-based nanosystems, proteins and peptides, as well as exosomes and cell membrane nanoparticles. Moreover, future challenges in the establishment of CpG delivery systems for immunotherapeutic applications are discussed. We expect that the continuously growing interest in the development of CpG-based immunotherapy will certainly fuel the excitement and stimulation in medicine research.
Collapse
Affiliation(s)
- Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Haochen Yao
- Hepatobiliary and Pancreatic Surgery Department, General Surgery Center, First Hospital of Jilin University, No. 1 Xinmin Street, Changchun, 130021, Jilin, China
| | - Ke Yi
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Yeh-Hsing Lao
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| | - Dan Shao
- Institutes of Life Sciences, School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, China
| | - Yu Tao
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| |
Collapse
|
14
|
Zhang Y, Song Z, Zhang Z, Zhang T, Gu P, Feng Z, Xu S, Yang Y, Wang D, Liu Z. Preparation and characterization of pickering emulsion stabilized by lovastatin nanoparticles for vaccine adjuvants. Int J Pharm 2024; 653:123901. [PMID: 38368969 DOI: 10.1016/j.ijpharm.2024.123901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/11/2024] [Accepted: 02/08/2024] [Indexed: 02/20/2024]
Abstract
While research on mevalonate inhibitors as vaccine adjuvants has made great progress to enhance the effectiveness of the vaccine, co delivery of lovastatin and antigens (OVA) remains an enormous challenge. Here, we encapsulated lovastatin into PLGA nanoparticles. PLGA loading lovastatin was further emulsified with squalene to prepare Pickering emulsion. The emulsification conditions of Pickering emulsion were optimized, and the optimal preparation conditions were obtained. After loading lovastatin and OVA, the size and zeta potential of LS-PPAS/OVA was 1043.33 nm and -22.07 mv, the adsorption rate of OVA was 63.34 %. The adsorbing of LS-PLGA nanoparticles on the surface of squalene in Pickering emulsions was demonstrated by Fluorescent confocal microscopy. After immunization, LS-PPAS enhanced the activation of dendritic cells in lymph nodes, further study found LS-PPAS not only elicited elevated levels of OVA-specific IgG and its subtypes, but also promoted the secretion of TNF-α, IFN-γ, and IL-6 in serum as a marker of cellular immunity. Importantly, LS-PPAS showed sufficient security through monitoring levels of biochemical parameters in serum and pathological observation of organ following vaccinations. LS-PPAS may act as a promising vaccine carrier to produce strong humoral and cellular immunity with acceptable safety.
Collapse
Affiliation(s)
- Yue Zhang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Zuchen Song
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Zhimin Zhang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Tao Zhang
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, Beijing 102206, PR China
| | - Pengfei Gu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Zian Feng
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Shuwen Xu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yang Yang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Deyun Wang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Zhenguang Liu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China.
| |
Collapse
|
15
|
Qi L, Hang T, Jiang W, Li S, Zhang H, Liang X, Lei L, Bi Q, Jiang H, Li Y. Proteinaceous Microsphere-Based Water-in-Oil Pickering Emulsions for Preservation of Chlorella Cells. Polymers (Basel) 2024; 16:647. [PMID: 38475330 DOI: 10.3390/polym16050647] [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: 11/30/2023] [Revised: 02/23/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Microalgae are highly regarded as ideal materials for the creation of liquid biofuels and have substantial potential for growth and utilization. However, traditional storage and culture methods for microalgae are plagued by challenges such as uncontrolled growth, bacterial contamination, and self-shading among algae. These issues severely impede the photosynthetic process and the efficient extraction of biomass energy. This study tackles these problems by utilizing magnetic hydrophobic protein particles to stabilize water-in-oil Pickering emulsions. This allows for the micro-compartment storage and magnetic transfer of algae. Additionally, the successful encapsulation of Chlorella cells in high-internal-phase water-in-oil Pickering emulsions effectively mitigates the settling problem of Chlorella cells in the liquid phase, thereby enabling the potential use of Pickering emulsions for the confined cultivation of microalgae.
Collapse
Affiliation(s)
- Lin Qi
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education & School of Chemical and Material Engineering, Jiangnan University, Wuxi 214122, China
| | - Teng Hang
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education & School of Chemical and Material Engineering, Jiangnan University, Wuxi 214122, China
| | - Weijie Jiang
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education & School of Chemical and Material Engineering, Jiangnan University, Wuxi 214122, China
| | - Sinong Li
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education & School of Chemical and Material Engineering, Jiangnan University, Wuxi 214122, China
| | - Hui Zhang
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education & School of Chemical and Material Engineering, Jiangnan University, Wuxi 214122, China
| | - Xiang Liang
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education & School of Chemical and Material Engineering, Jiangnan University, Wuxi 214122, China
| | - Le Lei
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education & School of Chemical and Material Engineering, Jiangnan University, Wuxi 214122, China
| | - Qiangqiang Bi
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education & School of Chemical and Material Engineering, Jiangnan University, Wuxi 214122, China
| | - Hang Jiang
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education & School of Chemical and Material Engineering, Jiangnan University, Wuxi 214122, China
| | - Yunxing Li
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education & School of Chemical and Material Engineering, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
16
|
Zhang J, Dong F, Liu C, Nie J, Feng S, Yi T. Progress of Drug Nanocrystal Self-Stabilized Pickering Emulsions: Construction, Characteristics In Vitro, and Fate In Vivo. Pharmaceutics 2024; 16:293. [PMID: 38399347 PMCID: PMC10891687 DOI: 10.3390/pharmaceutics16020293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/02/2024] [Accepted: 02/04/2024] [Indexed: 02/25/2024] Open
Abstract
A drug nanocrystal self-stabilized Pickering emulsion (DNSPE) is a novel Pickering emulsion with drug nanocrystals as the stabilizer. As a promising drug delivery system, DNSPEs have attracted increasing attention in recent years due to their high drug loading capacity and ability to reduce potential safety hazards posed by surfactants or specific solid particles. This paper comprehensively reviews the progress of research on DNSPEs, with an emphasis on the main factors influencing their construction, characteristics and measurement methods in vitro, and fate in vivo, and puts forward issues that need to be studied further. The review contributes to the advancement of DNSPE research and the promotion of their application in the field of drug delivery.
Collapse
Affiliation(s)
- Jifen Zhang
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China; (J.Z.); (S.F.)
| | - Fangming Dong
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China; (J.Z.); (S.F.)
| | - Chuan Liu
- Chengdu Institute of Food Inspection, Chengdu 611130, China;
| | - Jinyu Nie
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China; (J.Z.); (S.F.)
| | - Shan Feng
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China; (J.Z.); (S.F.)
| | - Tao Yi
- Faculty of Health Sciences and Sports, Macao Polytechnic University, Macau 999078, China
| |
Collapse
|
17
|
Zhao Y, Song R, Zhang Z, Hu H, Ning W, Duan X, Jiao J, Fu X, Zhang G. Hollow metal-organic framework-based, stimulator of interferon genes pathway-activating nanovaccines for tumor immunotherapy. NANOSCALE ADVANCES 2023; 6:72-78. [PMID: 38125595 PMCID: PMC10729872 DOI: 10.1039/d3na00867c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023]
Abstract
Nanovaccines have emerged as promising agents for cancer therapy because of their ability to induce specific immune responses without off-target effects. However, inadequate cytotoxic T lymphocyte response and low antigen/adjuvant encapsulation remain major obstacles to vaccinating against cancer. Herein, we designed a stimulator of interferon genes (STING) pathway-activating nanovaccine based on hollow metal-organic frameworks (MOFs) for tumor treatment. The nanovaccine (OVA@HZIF-Mn) was constructed by encapsulating a model antigen ovalbumin (OVA) into zeolitic imidazolate framework-8, followed by etching with tannic acid and functionalizing with manganese ions. Studies have shown that the nanovaccine can effectively enhance antigen uptake, STING pathway activation and dendritic cell maturation, triggering a robust immune response to inhibit tumor growth. In addition, no infection or pathological signs were observed in mice organs after multiple administrations. This study combines a simple assembly approach and superior therapeutic effect, providing a promising strategy for engineering effective nanovaccines.
Collapse
Affiliation(s)
- Yilei Zhao
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences Jinan Shandong 250117 China
| | - Ruinan Song
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences Jinan Shandong 250117 China
| | - Zhen Zhang
- Jinan Vocational College of Nursing Jinan Shandong 250102 China
| | - Houyang Hu
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences Jinan Shandong 250117 China
| | - Wenli Ning
- School of Stomatology, Shandong First Medical University & Shandong Academy of Medical Sciences Jinan Shandong 250021 China
| | - Xiuying Duan
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences Jinan Shandong 250117 China
| | - Jianwei Jiao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Science Beijing 100101 China
| | - Xiao Fu
- School of Stomatology, Shandong First Medical University & Shandong Academy of Medical Sciences Jinan Shandong 250021 China
| | - Guiqiang Zhang
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences Jinan Shandong 250117 China
- Shandong Hongkui Medical Laboratory Co., Ltd Jinan 271100 P. R. China
| |
Collapse
|
18
|
Chen J, Pan J, Liu S, Zhang Y, Sha S, Guo H, Wang X, Hao X, Zhou H, Tao S, Wang Y, Fan JB. Fruit-Derived Extracellular-Vesicle-Engineered Structural Droplet Drugs for Enhanced Glioblastoma Chemotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2304187. [PMID: 37589312 DOI: 10.1002/adma.202304187] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 08/07/2023] [Indexed: 08/18/2023]
Abstract
Existing solid-nanoparticle-based drug delivery systems remain a great challenge for glioblastoma chemotherapy due to their poor capacities in crossing the blood-brain barrier/blood-brain tumor barrier (BBB/BBTB). Herein, fruit-derived extracellular-vesicle (EV)-engineered structural droplet drugs (ESDDs) are demonstrated by programming the self-assembly of fruit-derived EVs at the DOX@squalene-PBS interface, greatly enhancing the antitumor efficacy against glioblastoma. The ESDDs experience a flexible delivery via deformation-amplified macropinocytosis and membrane fusion, enabling them to highly efficiently cross the BBB/BBTB and deeply penetrate glioblastoma tissues. As expected, the ESDDs exhibit approximately 2.5-fold intracellular uptake, 2.2-fold transcytosis, and fivefold membrane fusion higher than cRGD-modified EVs (REs), allowing highly efficient accumulation, deep penetration, and cellular internalization into the glioblastoma tissues, and thereby significantly extending the survival time of glioblastoma mice.
Collapse
Affiliation(s)
- Jianping Chen
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
- Department of Radiotherapy, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, 511518, P. R. China
| | - Jiahao Pan
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Sijia Liu
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Yangning Zhang
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Suinan Sha
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Haoyan Guo
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Xuejiao Wang
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Xiangrong Hao
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Houwang Zhou
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Sijian Tao
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Ying Wang
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Jun-Bing Fan
- Cancer Research Institute, Experimental Education/Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| |
Collapse
|
19
|
Jin H, Liu X, Liu HX. Biological function, regulatory mechanism, and clinical application of mannose in cancer. Biochim Biophys Acta Rev Cancer 2023; 1878:188970. [PMID: 37657682 DOI: 10.1016/j.bbcan.2023.188970] [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: 04/19/2023] [Revised: 08/15/2023] [Accepted: 08/15/2023] [Indexed: 09/03/2023]
Abstract
Studies examining the regulatory roles and clinical applications of monosaccharides other than glucose in cancer have been neglected. Mannose, a common type of monosaccharide found in human body fluids and tissues, primarily functions in protein glycosylation rather than carbohydrate metabolism. Recent research has demonstrated direct anticancer effects of mannose in vitro and in vivo. Simply supplementing cell culture medium or drinking water with mannose achieved these effects. Moreover, mannose enhances the effectiveness of current cancer treatments including chemotherapy, radiotherapy, targeted therapy, and immune therapy. Besides the advancements in basic research on the anticancer effects of mannose, recent studies have reported its application as a biomarker for cancer or in the delivery of anticancer drugs using mannose-modified drug delivery systems. This review discusses the progress made in understanding the regulatory roles of mannose in cancer progression, the mechanisms underlying its anticancer effects, and its current application in cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Haoyi Jin
- Department of Thoracic Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, 110042, Liaoning, China
| | - Xi Liu
- Department of Urology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, 110042, Liaoning, China
| | - Hong-Xu Liu
- Department of Thoracic Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, 110042, Liaoning, China; Department of Urology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, 110042, Liaoning, China.
| |
Collapse
|
20
|
Peng S, Yan Y, Ngai T, Li J, Ogino K, Xia Y. Development and Optimal Immune Strategy of an Alum-Stabilized Pickering emulsion for Cancer Vaccines. Vaccines (Basel) 2023; 11:1169. [PMID: 37514985 PMCID: PMC10383433 DOI: 10.3390/vaccines11071169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/25/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Therapeutic cancer vaccines are considered as one of the most cost-effective ways to eliminate cancer cells. Although many efforts have been invested into improving their therapeutic effect, transient maturation and activations of dendritic cells (DCs) cause weak responses and hamper the subsequent T cell responses. Here, we report on an alum-stabilized Pickering emulsion (APE) that can load a high number of antigens and continue to release them for extensive maturation and activations of antigen-presenting cells (APCs). After two vaccinations, APE/OVA induced both IFN-γ-secreting T cells (Th1) and IL-4-secreting T cells (Th2), generating effector CD8+ T cells against tumor growth. Additionally, although they boosted the cellular immune responses in the spleen, we found that multiple administrations of cancer vaccines (three or four times in 3-day intervals) may increase the immunosuppression with more PD-1+ CD8+ and LAG-3+ CD8+ T cells within the tumor environment, leading to the diminished overall anti-tumor efficacy. Combining this with anti-PD-1 antibodies evidently hindered the suppressive effect of multiple vaccine administrations, leading to the amplified tumor regression in B16-OVA-bearing mice.
Collapse
Affiliation(s)
- Sha Peng
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- Graduate School of Bio-Applications and Systems Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Tokyo 184-8588, Japan
- Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Tokyo 184-8588, Japan
| | - Yumeng Yan
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - To Ngai
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | - Jianjun Li
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Innovation Academy for Green Manufacture, Chinese Academy of Sciences, Beijing 100190, China
| | - Kenji Ogino
- Graduate School of Bio-Applications and Systems Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Tokyo 184-8588, Japan
- Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Tokyo 184-8588, Japan
| | - Yufei Xia
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Innovation Academy for Green Manufacture, Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
21
|
Xia Y, Fu S, Ma Q, Liu Y, Zhang N. Application of Nano-Delivery Systems in Lymph Nodes for Tumor Immunotherapy. NANO-MICRO LETTERS 2023; 15:145. [PMID: 37269391 PMCID: PMC10239433 DOI: 10.1007/s40820-023-01125-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 05/07/2023] [Indexed: 06/05/2023]
Abstract
Immunotherapy has become a promising research "hotspot" in cancer treatment. "Soldier" immune cells are not uniform throughout the body; they accumulate mostly in the immune organs such as the spleen and lymph nodes (LNs), etc. The unique structure of LNs provides the microenvironment suitable for the survival, activation, and proliferation of multiple types of immune cells. LNs play an important role in both the initiation of adaptive immunity and the generation of durable anti-tumor responses. Antigens taken up by antigen-presenting cells in peripheral tissues need to migrate with lymphatic fluid to LNs to activate the lymphocytes therein. Meanwhile, the accumulation and retaining of many immune functional compounds in LNs enhance their efficacy significantly. Therefore, LNs have become a key target for tumor immunotherapy. Unfortunately, the nonspecific distribution of the immune drugs in vivo greatly limits the activation and proliferation of immune cells, which leads to unsatisfactory anti-tumor effects. The efficient nano-delivery system to LNs is an effective strategy to maximize the efficacy of immune drugs. Nano-delivery systems have shown beneficial in improving biodistribution and enhancing accumulation in lymphoid tissues, exhibiting powerful and promising prospects for achieving effective delivery to LNs. Herein, the physiological structure and the delivery barriers of LNs were summarized and the factors affecting LNs accumulation were discussed thoroughly. Moreover, developments in nano-delivery systems were reviewed and the transformation prospects of LNs targeting nanocarriers were summarized and discussed.
Collapse
Affiliation(s)
- Yiming Xia
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Shunli Fu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Qingping Ma
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Yongjun Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China.
| | - Na Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China.
| |
Collapse
|
22
|
Zhang Y, Zhu T, Xu S, Gu P, Cai G, Peng S, Liu Z, Yang Y, Hu Y, Liu J, Wang D. Cationic Nanoparticle-Stabilized Vaccine Delivery System for the H9N2 Vaccine to Promote Immune Response in Chickens. Mol Pharm 2023; 20:1613-1623. [PMID: 36795759 DOI: 10.1021/acs.molpharmaceut.2c00805] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Chinese yam polysaccharides (CYPs) have received wide attention for their immunomodulatory activity. Our previous studies had discovered that the Chinese yam polysaccharide PLGA-stabilized Pickering emulsion (CYP-PPAS) can serve as an efficient adjuvant to trigger powerful humoral and cellular immunity. Recently, positively charged nano-adjuvants are easily taken up by antigen-presenting cells, potentially resulting in lysosomal escape, the promotion of antigen cross-presentation, and the induction of CD8 T-cell response. However, reports on the practical application of cationic Pickering emulsions as adjuvants are very limited. Considering the economic damage and public-health risks caused by the H9N2 influenza virus, it is urgent to develop an effective adjuvant for boosting humoral and cellular immunity against influenza virus infection. Here, we applied polyethyleneimine-modified Chinese yam polysaccharide PLGA nanoparticles as particle stabilizers and squalene as the oil core to fabricate a positively charged nanoparticle-stabilized Pickering emulsion adjuvant system (PEI-CYP-PPAS). The cationic Pickering emulsion of PEI-CYP-PPAS was utilized as an adjuvant for the H9N2 Avian influenza vaccine, and the adjuvant activity was compared with the Pickering emulsion of CYP-PPAS and the commercial adjuvant (aluminum adjuvant). The PEI-CYP-PPAS, with a size of about 1164.66 nm and a ζ potential of 33.23 mV, could increase the H9N2 antigen loading efficiency by 83.99%. After vaccination with Pickering emulsions based on H9N2 vaccines, PEI-CYP-PPAS generated higher HI titers and stronger IgG antibodies than CYP-PPAS and Alum and increased the immune organ index of the spleen and bursa of Fabricius without immune organ injury. Moreover, treatment with PEI-CYP-PPAS/H9N2 induced CD4+ and CD8+ T-cell activation, a high lymphocyte proliferation index, and increased cytokine expression of IL-4, IL-6, and IFN-γ. Thus, compared with the CYP-PPAS and aluminum adjuvant, the cationic nanoparticle-stabilized vaccine delivery system of PEI-CYP-PPAS was an effective adjuvant for H9N2 vaccination to elicit powerful humoral and cellular immune responses.
Collapse
Affiliation(s)
- Yue Zhang
- College of Veterinary Medicine, Institute of Traditional Chinese Veterinary Medicine, Nanjing, Jiangsu 210095, P. R. China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Tianyu Zhu
- College of Veterinary Medicine, Institute of Traditional Chinese Veterinary Medicine, Nanjing, Jiangsu 210095, P. R. China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Shuwen Xu
- College of Veterinary Medicine, Institute of Traditional Chinese Veterinary Medicine, Nanjing, Jiangsu 210095, P. R. China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Pengfei Gu
- College of Veterinary Medicine, Institute of Traditional Chinese Veterinary Medicine, Nanjing, Jiangsu 210095, P. R. China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Gaofeng Cai
- College of Veterinary Medicine, Institute of Traditional Chinese Veterinary Medicine, Nanjing, Jiangsu 210095, P. R. China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Song Peng
- College of Veterinary Medicine, Institute of Traditional Chinese Veterinary Medicine, Nanjing, Jiangsu 210095, P. R. China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Zhenguang Liu
- College of Veterinary Medicine, Institute of Traditional Chinese Veterinary Medicine, Nanjing, Jiangsu 210095, P. R. China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Yang Yang
- College of Veterinary Medicine, Institute of Traditional Chinese Veterinary Medicine, Nanjing, Jiangsu 210095, P. R. China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Yuanliang Hu
- College of Veterinary Medicine, Institute of Traditional Chinese Veterinary Medicine, Nanjing, Jiangsu 210095, P. R. China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Jiaguo Liu
- College of Veterinary Medicine, Institute of Traditional Chinese Veterinary Medicine, Nanjing, Jiangsu 210095, P. R. China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Deyun Wang
- College of Veterinary Medicine, Institute of Traditional Chinese Veterinary Medicine, Nanjing, Jiangsu 210095, P. R. China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P. R. China
| |
Collapse
|
23
|
Zhang Y, Chen J, Shi L, Ma F. Polymeric nanoparticle-based nanovaccines for cancer immunotherapy. MATERIALS HORIZONS 2023; 10:361-392. [PMID: 36541078 DOI: 10.1039/d2mh01358d] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Therapeutic cancer vaccines, which are designed to amplify tumor-specific T cell responses, have been envisioned as one of the most powerful tools for effective cancer immunotherapy. However, increasing the potency, quality and durability of the vaccine response remains a big challenge. In recent years, materials-based delivery systems focusing on the co-delivery of antigens and adjuvants to enhance cancer vaccination therapy have attracted increasing interest. Among various materials, polymeric nanoparticles (NPs) with different physicochemical properties which can incorporate multiple immunological cues are of great interest. In this review, the recent progress in the design and construction of both ex vivo subunit and in situ cancer vaccines using polymeric NPs is summarized. Especially, we will focus on how these NPs improve the adjuvanticity of vaccines. The design principles of polymeric NPs for ex vivo subunit cancer vaccines and in situ cancer vaccination are also discussed. Finally, we want to briefly discuss molecular chaperones in cancer immunity and the applications of our unique self-assembly mixed shell polymeric micelle-based nanochaperones for cancer vaccines.
Collapse
Affiliation(s)
- Yongxin Zhang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry and College of Chemistry, Nankai University, Tianjin, 300071, P. R. China.
| | - Jiajing Chen
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry and College of Chemistry, Nankai University, Tianjin, 300071, P. R. China.
| | - Linqi Shi
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry and College of Chemistry, Nankai University, Tianjin, 300071, P. R. China.
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin, 300192, P. R. China
| | - Feihe Ma
- State Key Laboratory of Separation Membranes and Membrane Processes, School of Materials Science and Engineering, Tiangong University, Tianjin, 300387, P. R. China.
| |
Collapse
|
24
|
Huo J, Zhang A, Wang S, Cheng H, Fan D, Huang R, Wang Y, Wan B, Zhang G, He H. Splenic-targeting biomimetic nanovaccine for elevating protective immunity against virus infection. J Nanobiotechnology 2022; 20:514. [PMID: 36463277 PMCID: PMC9719655 DOI: 10.1186/s12951-022-01730-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND The prevalence of viral infectious diseases has become a serious threat to public safety, economic and social development. Vaccines have been served as the most effective platform to prevent virus transmission via the activation of host immune responses, while the low immunogenicity or safety, the high cost of production, storage, transport limit their effective clinical application. Therefore, there is a need to develop a promising strategy to improve the immunogenicity and safety of vaccines. METHODS We developed a splenic-targeting biomimetic nanovaccine (NV) that can boost protective humoral and cellular immunity against african swine fever virus (ASFV) infection. The universal PLGA nanoparticles (CMR-PLGA/p54 NPs) coated with mannose and CpG (TLR9 agonist) co-modified red blood cell (RBC) membrane were prepared, which comprised a viral antigen (p54) and can be served as a versatile nanovaccine for elevating protective immunity. RESULTS CMR-PLGA/p54 NVs could be effectively uptaken by BMDC and promoted BMDC maturation in vitro. After subcutaneous immunization, antigen could be effectively delivered to the splenic dendritic cells (DCs) due to the splenic homing ability of RBC and DC targeting capacity of mannose, which promoted antigen presentation and DCs maturation, and further elicited higher levels of cytokines secretion and specific IgG titers, CD4+ and CD8+ T cells activation and B maturation. Moreover, NVs demonstrated notable safety during the immunization period. CONCLUSIONS This study demonstrates the high potential of CMR-PLGA NPs as vaccine delivery carriers to promote humoral and cellular immune responses, and it provides a promising strategy to develop safe and effective vaccines against viral infectious diseases.
Collapse
Affiliation(s)
- Jian Huo
- grid.108266.b0000 0004 1803 0494College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Engineering Laboratory of Animal Biological Products, Longhu Laboratory, Henan Agricultural University, Zhengzhou, 450046 China
| | - Angke Zhang
- grid.108266.b0000 0004 1803 0494College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Engineering Laboratory of Animal Biological Products, Longhu Laboratory, Henan Agricultural University, Zhengzhou, 450046 China
| | - Shuqi Wang
- grid.108266.b0000 0004 1803 0494College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Engineering Laboratory of Animal Biological Products, Longhu Laboratory, Henan Agricultural University, Zhengzhou, 450046 China
| | - Hanghang Cheng
- grid.108266.b0000 0004 1803 0494College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Engineering Laboratory of Animal Biological Products, Longhu Laboratory, Henan Agricultural University, Zhengzhou, 450046 China
| | - Daopeng Fan
- grid.108266.b0000 0004 1803 0494College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Engineering Laboratory of Animal Biological Products, Longhu Laboratory, Henan Agricultural University, Zhengzhou, 450046 China
| | - Ran Huang
- grid.108266.b0000 0004 1803 0494College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Engineering Laboratory of Animal Biological Products, Longhu Laboratory, Henan Agricultural University, Zhengzhou, 450046 China
| | - Yanan Wang
- grid.108266.b0000 0004 1803 0494College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Engineering Laboratory of Animal Biological Products, Longhu Laboratory, Henan Agricultural University, Zhengzhou, 450046 China
| | - Bo Wan
- grid.108266.b0000 0004 1803 0494College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Engineering Laboratory of Animal Biological Products, Longhu Laboratory, Henan Agricultural University, Zhengzhou, 450046 China
| | - Gaiping Zhang
- grid.108266.b0000 0004 1803 0494College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Engineering Laboratory of Animal Biological Products, Longhu Laboratory, Henan Agricultural University, Zhengzhou, 450046 China
| | - Hua He
- grid.108266.b0000 0004 1803 0494College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Engineering Laboratory of Animal Biological Products, Longhu Laboratory, Henan Agricultural University, Zhengzhou, 450046 China
| |
Collapse
|
25
|
Nahar UJ, Toth I, Skwarczynski M. Mannose in vaccine delivery. J Control Release 2022; 351:284-300. [PMID: 36150579 DOI: 10.1016/j.jconrel.2022.09.038] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/15/2022] [Accepted: 09/16/2022] [Indexed: 11/16/2022]
Abstract
Adjuvants and vaccine delivery systems are used widely to improve the efficacy of vaccines. Their primary roles are to protect antigen from degradation and allow its delivery and uptake by antigen presenting cells (APCs). Carbohydrates, including various structures/forms of mannose, have been broadly utilized to target carbohydrate binding receptors on APCs. This review summarizes basic functions of the immune system, focusing on the role of mannose receptors in antigen recognition by APCs. The most popular strategies to produce mannosylated vaccines via conjugation and formulation are presented. The efficacy of mannosylated vaccines is discussed in detail, taking into consideration factors, such as valency and number of mannose in mannose ligands, mannose density, length of spacers, special arrangement of mannose ligands, and routes of administration of mannosylated vaccines. The advantages and disadvantages of mannosylation strategy and future directions in the development of mannosylated vaccines are also debated.
Collapse
Affiliation(s)
- Ummey Jannatun Nahar
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia; 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.
| |
Collapse
|
26
|
Rostamabadi MM, Falsafi SR, Nishinari K, Rostamabadi H. Seed gum-based delivery systems and their application in encapsulation of bioactive molecules. Crit Rev Food Sci Nutr 2022; 63:9937-9960. [PMID: 35587167 DOI: 10.1080/10408398.2022.2076065] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Now-a-days, the food/pharma realm faces with great challenges for the application of bioactive molecules when applying them in free form due to their instability in vitro/in vivo. For promoting the biological and functional properties of bioactive molecules, efficient delivery systems have played a pivotal role offering a controlled delivery and improved bioavailability/solubility of bioactives. Among different carbohydrate-based delivery systems, seed gum-based vehicles (SGVs) have shown great promise, facilitating the delivery of a high concentration of bioactive at the site of action, a controlled payload release, and less bioactive loss. SGVs are potent structures to promote the bioavailability, beneficial properties, and in vitro/in vivo stability of bioactive components. Here, we offer a comprehensive overview of seed gum-based nano- and microdevices as delivery systems for bioactive molecules. We have a focus on structural/functional attributes and health-promoting benefits of seed gums, but also strategies involving modification of these biopolymers are included. Diverse SGVs (nano/microparticles, functional films, hydrogels/nanogels, particles for Pickering nanoemulsions, multilayer carriers, emulsions, and complexes/conjugates) are reviewed and important parameters for bioactive delivery are highlighted (e.g. bioactive-loading capacity, control of bioactive release, (bio)stability, and so on). Future challenges for these biopolymer-based carriers have also been discussed. HighlightsSeed gum-based polymers are promising materials to design different bioactive delivery systems.Seed gum-based delivery systems are particles, fibers, complexes, conjugates, hydrogels, etc.Seed gum-based vehicles are potent structures to promote the bioavailability, beneficial properties, and in vitro/in vivo stability of bioactive components.
Collapse
Affiliation(s)
- Mohammad Mahdi Rostamabadi
- Department of Food Science and Technology, College of Agriculture, Isfahan University of Technology, Isfahan, Iran
| | - Seid Reza Falsafi
- Faculty of Food Science and Technology, Gorgan University of Agricultural Sciences and Natural Resources, Gorgan, Iran
| | - Katsuyoshi Nishinari
- Glyn O. Phillips Hydrocolloid Research Centre, Department of Bioengineering and Food Science, Hubei University of Technology, Wuhan, China
- Food Hydrocolloid International Science and Technology, Cooperation Base of Hubei Province, Hubei University of Technology, Wuhan, China
| | - Hadis Rostamabadi
- Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
27
|
Protein corona-driven nanovaccines improve antigen intracellular release and immunotherapy efficacy. J Control Release 2022; 345:601-609. [PMID: 35346769 DOI: 10.1016/j.jconrel.2022.03.048] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 03/18/2022] [Accepted: 03/23/2022] [Indexed: 01/18/2023]
Abstract
During vaccine delivery in vivo, the vaccine carrier dynamically adsorbs the surrounding proteins or biomacromolecules to form a protein corona layer, which determines the physiological and therapeutic responses of the vaccine. Although the importance of the protein corona effect in drug delivery is widely accepted, understanding of the rational use of the protein corona to improve antigen controlled release is still sparse. Here, we constructed a protein corona-driven nanovaccine (PCNV), which has the dual effects of resisting the protein corona-induced antigen extracellular release and promoting protein corona-triggered antigen cytosolic release under reductive conditions. Specifically, the nanovaccine was formulated via the assembly of fluorinated dendrigraft-poly-lysine and cleavable antigen-CpG conjugate. Before entering antigen-presenting cells (APCs), the anchoring effect of CpG was used to avoid the dissociation of antigens from the carrier even under the protein corona effect. While nanovaccine enters the APCs, the intracellular reducing conditions can induce a break in the disulfide bond between CpG and antigen. Notably, at the same time, the intracellular protein corona effect triggers antigen release from the carrier and achieves efficient antigen presentation. In addition, the PCNV produced a significant prophylactic and therapeutic antitumor response in the mouse model. Therefore, the rational use of the protein corona effect provides an effective strategy for vaccine delivery.
Collapse
|