1
|
Wan H, Yang Y, Tu Z, Tang M, Jing B, Feng Y, Xie J, Gao H, Song X, Zhao X. Enhanced mucosal immune response through nanoparticle delivery system based on chitosan-catechol and a recombinant antigen targeted towards M cells. Int J Biol Macromol 2025; 306:141345. [PMID: 40010449 DOI: 10.1016/j.ijbiomac.2025.141345] [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: 11/20/2024] [Revised: 02/12/2025] [Accepted: 02/19/2025] [Indexed: 02/28/2025]
Abstract
In mucosal vaccination, the targeted delivery of antigens through M (microfold) cells is essential for initiating a robust antigen-specific immune response. In the present study, we devised a nano-delivery platform to target M cells. This platform involved coating mesoporous silica nanoparticles (MSN) with a mucoadhesive chitosan-catechol (Chic) layer, incorporating a recombinant antigen to form nanoparticles that enhance the immune response. The collagenase equivalent domain (COE) of porcine epidemic diarrhea virus (PEDV) terminated with the M cell-targeting sequence RGD (COER), was initially expressed by Escherichia coli (E. coli) and subsequently conjugated to the surface of MSN-Chic, forming the MSN-Chic-COER nanoparticles. MSN-Chic-COER with strong mucoadhesive properties and a propensity for M cell targeting, demonstrated enhanced uptake by dendritic cells (DCs) and trafficking to lymph nodes, compared to COE/COER after intranasal administration. MSN-Chic-COER recruited more dendritic cells to the antigen-located site via stimulating chemokine CCL20 secretion was evidenced by cell co-culture model. Additionally, it enhanced antigen permeability by disrupting the distribution of the ZO-1 protein in epithelial cells. Notably, MSN-Chic-COER elicited a higher level of cellular immunity, humoral immunity, and PEDV neutralizing antibody production. These findings underscore the potential of MSN-Chic-COER as a promising intranasal vaccine delivery system.
Collapse
Affiliation(s)
- Hongping Wan
- Center for Infectious Diseases Control (CIDC), Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Center for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu 611130, China.
| | - Yunhan Yang
- Center for Infectious Diseases Control (CIDC), Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Center for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu 611130, China
| | - Zhiwen Tu
- Center for Infectious Diseases Control (CIDC), Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Center for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu 611130, China
| | - Mingrun Tang
- Center for Infectious Diseases Control (CIDC), Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Center for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu 611130, China
| | - Bo Jing
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yumei Feng
- Instrumental analysis center, Sichuan Agricultural University, Chengdu 611130, China
| | - Jiulong Xie
- Instrumental analysis center, Sichuan Agricultural University, Chengdu 611130, China
| | - Huile Gao
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu 610041, China.
| | - Xu Song
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Xinghong Zhao
- Center for Infectious Diseases Control (CIDC), Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Center for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
2
|
Zhang J, Yang X, Chang Z, Zhu W, Ma Y, He H. Polymeric nanocarriers for therapeutic gene delivery. Asian J Pharm Sci 2025; 20:101015. [PMID: 39931356 PMCID: PMC11808530 DOI: 10.1016/j.ajps.2025.101015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 11/12/2024] [Accepted: 11/19/2024] [Indexed: 02/13/2025] Open
Abstract
The recent commercialization of gene products has sparked significant interest in gene therapy, necessitating efficient and precise gene delivery via various vectors. Currently, viral vectors and lipid-based nanocarriers are the predominant choices and have been extensively investigated and reviewed. Beyond these vectors, polymeric nanocarriers also hold the promise in therapeutic gene delivery owing to their versatile functionalities, such as improving the stability, cellar uptake and endosomal escape of nucleic acid drugs, along with precise delivery to targeted tissues. This review presents a brief overview of the status quo of the emerging polymeric nanocarriers for therapeutic gene delivery, focusing on key cationic polymers, nanocarrier types, and preparation methods. It also highlights targeted diseases, strategies to improve delivery efficiency, and potential future directions in this research area. The review is hoped to inspire the development, optimization, and clinical translation of highly efficient polymeric nanocarriers for therapeutic gene delivery.
Collapse
Affiliation(s)
- Jiayuan Zhang
- Key Laboratory of Smart Drug Delivery of Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
- Key Laboratory for Tibet Plateau Phytochemistry of Qinghai Province, School of Pharmacy, Qinghai Minzu University, Xining 810007, China
| | - Xinyu Yang
- Key Laboratory of Smart Drug Delivery of Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Zhichao Chang
- Key Laboratory of Smart Drug Delivery of Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Wenwei Zhu
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yuhua Ma
- Key Laboratory for Tibet Plateau Phytochemistry of Qinghai Province, School of Pharmacy, Qinghai Minzu University, Xining 810007, China
| | - Haisheng He
- Key Laboratory of Smart Drug Delivery of Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| |
Collapse
|
3
|
Saleh M, El-Moghazy A, Elgohary AH, Saber WIA, Helmy YA. Revolutionizing Nanovaccines: A New Era of Immunization. Vaccines (Basel) 2025; 13:126. [PMID: 40006673 PMCID: PMC11860605 DOI: 10.3390/vaccines13020126] [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: 11/16/2024] [Revised: 01/13/2025] [Accepted: 01/17/2025] [Indexed: 02/27/2025] Open
Abstract
Infectious diseases continue to pose a significant global health threat. To combat these challenges, innovative vaccine technologies are urgently needed. Nanoparticles (NPs) have unique properties and have emerged as a promising platform for developing next-generation vaccines. Nanoparticles are revolutionizing the field of vaccine development, offering a new era of immunization. They allow the creation of more effective, stable, and easily deliverable vaccines. Various types of NPs, including lipid, polymeric, metal, and virus-like particles, can be employed to encapsulate and deliver vaccine components, such as mRNA or protein antigens. These NPs protect antigens from degradation, target them to specific immune cells, and enhance antigen presentation, leading to robust and durable immune responses. Additionally, NPs can simultaneously deliver multiple vaccine components, including antigens, and adjuvants, in a single formulation, simplifying vaccine production and administration. Nanovaccines offer a promising approach to combat food- and water-borne bacterial diseases, surpassing traditional formulations. Further research is needed to address the global burden of these infections. This review highlights the potential of NPs to revolutionize vaccine platforms. We explore their mechanisms of action, current applications, and emerging trends. The review discusses the limitations of nanovaccines, innovative solutions and the potential role of artificial intelligence in developing more effective and accessible nanovaccines to combat infectious diseases.
Collapse
Affiliation(s)
- Mohammed Saleh
- Department of Veterinary Science, Martin-Gatton College of Agriculture, Food, and Environment, University of Kentucky, Lexington, KY 40546, USA
| | - Ahmed El-Moghazy
- Department of Microbiology and Plant Pathology, University of California, Riverside, CA 92521, USA
| | - Adel H. Elgohary
- Department of Hygiene and Zoonoses, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| | - WesamEldin I. A. Saber
- Microbial Activity Unit, Department of Microbiology, Soils, Water and Environment Research Institute, Agricultural Research Center, Giza 12619, Egypt
| | - Yosra A. Helmy
- Department of Veterinary Science, Martin-Gatton College of Agriculture, Food, and Environment, University of Kentucky, Lexington, KY 40546, USA
| |
Collapse
|
4
|
Wijesundara YH, Arora N, Ehrman RN, Howlett TS, Weyman TM, Trashi I, Trashi O, Kumari S, Diwakara SD, Tang W, Senarathna MC, Drewniak KH, Wang Z, Smaldone RA, Gassensmith JJ. A Self-Adjuvanting Large Pore 2D Covalent Organic Framework as a Vaccine Platform. Angew Chem Int Ed Engl 2025; 64:e202413020. [PMID: 39621809 DOI: 10.1002/anie.202413020] [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: 07/10/2024] [Revised: 12/01/2024] [Indexed: 12/17/2024]
Abstract
Vaccines are one of the greatest human achievements in public health, as they help prevent the spread of diseases, reduce illness and death rates, saving thousands of lives with few side effects. Traditional vaccine development is centered around using live attenuated or inactivated pathogens, which is expensive and has resulted in vaccine-associated illnesses. Advancements have led to the development of safer subunit vaccines, which contain recombinant proteins isolated from pathogens. Their short half-life and small size make most subunit vaccines less immunogenic. Here, we introduce a large pore 2D covalent organic framework (COF), PyCOFamide, as a promising solution for an effective subunit platform. Our study demonstrates that simple adsorption of a model antigen, ovalbumin (OVA), onto PyCOFamide (OVA@COF) significantly enhances humoral and cell-mediated immune response compared to free OVA. OVA@COF exhibited heightened immune cell activation and acts as an antigen reservoir, facilitating antigen-presenting cell trafficking to the draining lymph nodes, amplifying the humoral immune response. Additionally, the breakdown of the COF releases monomers that adjuvant the activation of immune cells vital to creating strong immunity. This platform offers a potential avenue for safer, more effective subunit vaccines.
Collapse
Affiliation(s)
- Yalini H Wijesundara
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, 800 West Campbell Rd, Richardson, 75080, TX, USA
| | - Niyati Arora
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, 800 West Campbell Rd, Richardson, 75080, TX, USA
| | - Ryanne N Ehrman
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, 800 West Campbell Rd, Richardson, 75080, TX, USA
| | - Thomas Sinclair Howlett
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, 800 West Campbell Rd, Richardson, 75080, TX, USA
| | - Trevor M Weyman
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, 800 West Campbell Rd, Richardson, 75080, TX, USA
| | - Ikeda Trashi
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, 800 West Campbell Rd, Richardson, 75080, TX, USA
| | - Orikeda Trashi
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, 800 West Campbell Rd, Richardson, 75080, TX, USA
| | - Sneha Kumari
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, 800 West Campbell Rd, Richardson, 75080, TX, USA
| | - Shashini D Diwakara
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, 800 West Campbell Rd, Richardson, 75080, TX, USA
| | - Wendy Tang
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, 800 West Campbell Rd, Richardson, 75080, TX, USA
| | - Milinda C Senarathna
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, 800 West Campbell Rd, Richardson, 75080, TX, USA
| | - Katarzyna H Drewniak
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, 800 West Campbell Rd, Richardson, 75080, TX, USA
| | - Ziqi Wang
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, 800 West Campbell Rd, Richardson, 75080, TX, USA
| | - Ronald A Smaldone
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, 800 West Campbell Rd, Richardson, 75080, TX, USA
| | - Jeremiah J Gassensmith
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, 800 West Campbell Rd, Richardson, 75080, TX, USA
- Department of Bioengineering, The University of Texas at Dallas, 800 West Campbell Rd, Richardson, 75080, TX, USA
| |
Collapse
|
5
|
He J, Zhu T, Mao N, Jiang W, Lin F, Lu X, Gao Z, Yang Y, Wang D. Cistanche deserticola polysaccharide-functionalized dendritic fibrous nano-silica as oral delivery system for H 9N 2 vaccine to promote systemic and mucosal immune response. Int J Biol Macromol 2024; 282:136690. [PMID: 39433190 DOI: 10.1016/j.ijbiomac.2024.136690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 10/10/2024] [Accepted: 10/17/2024] [Indexed: 10/23/2024]
Abstract
Most infectious diseases are caused by pathogens that invade the body tissues through mucosal tract. Therefore, it is essential to develop effective vaccines administered through the mucosa as a first-line of defense against major infectious diseases. Oral delivery of vaccines is currently of great interest due to its potential to elicit both mucosal and systemic immune responses, high compliance rate and non-invasive nature. However, their development is limited by the challenging gastrointestinal (GI) environment, the low permeability of the mucus barrier, and the lack of effective and safe mucosal adjuvants. Currently, nanoparticle-based strategies show significant potential for improving oral vaccine delivery systems. Herein, the dendritic fibrous nano-silica (DFNS) grafted with Cistanche deserticola polysaccharide (CDP) nanoparticles (CDP-DFNS) were developed for oral delivery of H9N2 antigen. CDP-DFNS induced the activation of macrophages, thereby enhancing antigen uptake in vitro. Additionally, CDP-DFNS/H9N2 significantly activated the dendritic cells (DCs) in Peyer's patches (PPs), and T/B cells in mesenteric lymph nodes (MLNs). Moreover, CDP-DFNS/H9N2 enhanced the HI titers and levels of H9N2-specific antibody IgG, secretory IgA (SIgA) and H9N2-specific IgA in intestinal and respiratory mucosa, as well as Th-associated cytokines. Our results indicate that CDP-DFNS could be a promising oral vaccine adjuvant delivery system.
Collapse
Affiliation(s)
- Jin He
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Tianyu Zhu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Ningning Mao
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Wenming Jiang
- China Animal Health and Epidemiology Center, Qingdao, PR China
| | - Fangzhu Lin
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Xuanqi Lu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Zhenzhen Gao
- College of Animal Husbandry and Veterinary Medicine, Jiangsu Vocational College of Agriculture and Forestry, Jurong, Jiangsu 212499, PR China
| | - Yang Yang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Deyun Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China.
| |
Collapse
|
6
|
Ontiveros-Padilla L, Bachelder EM, Ainslie KM. Microparticle and nanoparticle-based influenza vaccines. J Control Release 2024; 376:880-898. [PMID: 39427775 DOI: 10.1016/j.jconrel.2024.10.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/12/2024] [Accepted: 10/15/2024] [Indexed: 10/22/2024]
Abstract
Influenza infections are a health public problem worldwide every year with the potential to become the next pandemic. Vaccination is the most effective strategy to prevent future influenza outbreaks, however, influenza vaccines need to be reformulated each year to provide protection due to viral antigenic drift and shift. As more efficient influenza vaccines are needed, it is relevant to recapitulate strategies to improve the immunogenicity and broad reactivity of the current vaccines. Here, we review the current approved vaccines in the U.S. market and the platform used for their production. We discuss the different approaches to develop a broadly reactive vaccine as well as reviewing the adjuvant systems that are under study for being potentially included in future influenza vaccine formulations. The main components of the immune system involved in achieving a protective immune response are reviewed and how they participate in the trafficking of particles systemically and in the mucosa. Finally, we describe and classify, according to their physicochemical properties, some of the potential micro and nano-particulate platforms that can be used as delivery systems for parenteral and mucosal vaccinations.
Collapse
Affiliation(s)
- Luis Ontiveros-Padilla
- Division of Pharmacoengineering & Molecular Pharmaceutics, Eshelman School of Pharmacy, UNC, Chapel Hill, NC, USA
| | - Eric M Bachelder
- Division of Pharmacoengineering & Molecular Pharmaceutics, Eshelman School of Pharmacy, UNC, Chapel Hill, NC, USA
| | - Kristy M Ainslie
- Division of Pharmacoengineering & Molecular Pharmaceutics, Eshelman School of Pharmacy, UNC, Chapel Hill, NC, USA; Department of Biomedical Engineering, NC State/UNC, Chapel Hill, NC, USA; Department of Microbiology and Immunology, School of Medicine, UNC, Chapel Hill, NC, USA.
| |
Collapse
|
7
|
Wan D, Bai Z, Zhang Y, Chen L, Que H, Lan T, Hong W, Huang J, He C, Wei Y, Pu Q, Wei X. Simultaneous enhancement of cellular and humoral immunity by the lymph node-targeted cholesterolized TLR7 agonist liposomes. Acta Pharm Sin B 2024; 14:4577-4590. [PMID: 39525596 PMCID: PMC11544185 DOI: 10.1016/j.apsb.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 05/14/2024] [Accepted: 05/16/2024] [Indexed: 11/16/2024] Open
Abstract
Toll-like receptor (TLR) agonists, as promising adjuvants and immunotherapeutic agents, have the potential to enhance immune responses and modulate antigen-dependent T-cell immune memory through activation of distinct signaling pathways. However, their clinical application is hindered by uncontrolled systemic inflammatory reactions. Therefore, it is imperative to create a vaccine adjuvant for TLR receptors that ensures both safety and efficacy. In this study, we designed lymph node-targeted cholesterolized TLR7 agonist cationic liposomes (1V209-Cho-Lip+) to mitigate undesired side effects. Co-delivery of the model antigen OVA and cholesterolized TLR7 agonist facilitated DC maturation through TLR activation while ensuring optimal presentation of the antigen to CD8+ T cells. The main aim of the present study is to evaluate the adjuvant effectiveness of 1V209-Cho-Lip+ in tumor vaccines. Following immunization with 1V209-Cho-Lip++OVA, we observed a pronounced "depot effect" and enhanced trafficking to secondary lymphoid organs. Prophylactic vaccination with 1V209-Cho-Lip++OVA significantly delays tumor development, prolongs mouse survival, and establishes durable immunity against tumor recurrence. Additionally, 1V209-Cho-Lip++OVA, while used therapeutic tumor vaccine, has demonstrated its efficacy in inhibiting tumor progression, and when combined with anti-PD-1, it further enhances antitumor effects. Therefore, the co-delivery of antigen and lymph node-targeted cholesterolized TLR7 agonist shows great promise as a cancer vaccine.
Collapse
Affiliation(s)
- Dandan Wan
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ziyi Bai
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yu Zhang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Li Chen
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Haiying Que
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tianxia Lan
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Weiqi Hong
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiayu Huang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Cai He
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiang Pu
- Department of Thoracic Surgery, National Frontier Center of Disease Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
8
|
Jia H, Zheng Z, Qu J, Feng T, Jiang X, Yu H, Zhu Z, Su F, Yang Y, Lu Q, Jie Q. Study on the synthesis of iron-based nanomedicine assisted by angelica sinensis polysaccharide with enhanced retention performance and its application in anemia treatment. Int J Biol Macromol 2024; 280:135969. [PMID: 39322144 DOI: 10.1016/j.ijbiomac.2024.135969] [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: 07/25/2024] [Revised: 09/20/2024] [Accepted: 09/21/2024] [Indexed: 09/27/2024]
Abstract
Inappropriate treatment of chronic inflammation and infection can lead to serious consequences, with anemia being the most common secondary disease that often requires systematic treatment. However, the complex pathology and gastrointestinal irritation associated with oral iron supplements limit their effectiveness. To address this, a bioactive ingredient derived from natural herbs, Angelica sinensis polysaccharide (ASP), was utilized as an ideal adjuvant for regulating the size and stability of iron oxide nanoparticles (IONPs). Highly hydrophilic ASP-modified IONPs (IONPs@ASP) with a mesoporous structure were developed under the induction of microemulsion.The as-prepared IONPs@ASP exhibited enhanced stability, retention performance and controlled degradation in blood and lysosomal environments, respectively, which is beneficial for long-term intravenous iron maintenance in anemia treatment. After confirming the biosafety of IONPs@ASP, pharmacodynamic results showed that hemoglobin levels increased significantly and rapidly returned to normal levels in anemia model rats treated with IONPs@ASP, even surpassing the effects of IONPs or ASP monotherapy. Additionally, analysis of inflammatory factors in rat serum suggested that ASP effectively upregulated the expression of anti-inflammatory factors, indicating synergistic effects of iron-based nanomedicine and immune regulation in anemia treatment. These findings represent a significant advancement in anemia treatment and open new possibilities for developing versatile nanoparticles based on ASP.
Collapse
Affiliation(s)
- Haoruo Jia
- Pediatric Orthopaedic Hospital, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, China; Clinical Research Center for Pediactric Skeletal Deformity and Injury of Shaanxi Province, Xi'an 710054, China; Xi'an Key Laboratory of Skeletal Developmental Deformity and Injury Repain, Xi'an 710054, China
| | - Ziyuan Zheng
- School of Environmental Science and Engineering, Changzhou University, Changzhou 213164, China
| | - Jining Qu
- Pediatric Orthopaedic Hospital, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, China; Clinical Research Center for Pediactric Skeletal Deformity and Injury of Shaanxi Province, Xi'an 710054, China; Xi'an Key Laboratory of Skeletal Developmental Deformity and Injury Repain, Xi'an 710054, China
| | | | - Xin Jiang
- Xi'an Medical University, Xian 710068, China
| | - Hongtao Yu
- First Affiliated Hospital, Shihezi University, Shihezi 832008, China
| | - Zhoujun Zhu
- Department of Joint Surgery, Sixth Affiliated Hospital, Xinjiang Medical University, Urumqi 830092, China
| | - Fei Su
- Pediatric Orthopaedic Hospital, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, China; Clinical Research Center for Pediactric Skeletal Deformity and Injury of Shaanxi Province, Xi'an 710054, China; Xi'an Key Laboratory of Skeletal Developmental Deformity and Injury Repain, Xi'an 710054, China
| | - Yating Yang
- Pediatric Orthopaedic Hospital, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, China; Clinical Research Center for Pediactric Skeletal Deformity and Injury of Shaanxi Province, Xi'an 710054, China; Xi'an Key Laboratory of Skeletal Developmental Deformity and Injury Repain, Xi'an 710054, China
| | - Qingda Lu
- Pediatric Orthopaedic Hospital, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, China; Clinical Research Center for Pediactric Skeletal Deformity and Injury of Shaanxi Province, Xi'an 710054, China; Xi'an Key Laboratory of Skeletal Developmental Deformity and Injury Repain, Xi'an 710054, China
| | - Qiang Jie
- Pediatric Orthopaedic Hospital, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, China; Clinical Research Center for Pediactric Skeletal Deformity and Injury of Shaanxi Province, Xi'an 710054, China; Xi'an Key Laboratory of Skeletal Developmental Deformity and Injury Repain, Xi'an 710054, China.
| |
Collapse
|
9
|
He J, Lu X, Mao N, Zhu T, Yu L, Yu Y, Peng S, Deng X, Hu B, Jiang W, Lu Y, Wang D. Cistanche deserticola polysaccharide- functionalized dendritic fibrous nano-silica -based adjuvant for H 9N 2 oral vaccine enhance systemic and mucosal immunity in chickens. Int J Pharm 2024; 660:124318. [PMID: 38852750 DOI: 10.1016/j.ijpharm.2024.124318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/29/2024] [Accepted: 06/05/2024] [Indexed: 06/11/2024]
Abstract
Avian influenza virus subtype H9N2 has the ability to infect birds and humans, further causing significant losses to the poultry industry and even posing a great threat to human health. Oral vaccine received particular interest for preventing majority infection due to its ability to elicit both mucosal and systemic immune responses, but their development is limited by the bad gastrointestinal (GI) environment, compact epithelium and mucus barrier, and the lack of effective mucosal adjuvants. Herein, we developed the dendritic fibrous nano-silica (DFNS) grafted with Cistanche deserticola polysaccharide (CDP) nanoparticles (CDP-DFNS) as an adjuvant for H9N2 vaccine. Encouragingly, CDP-DFNS facilitated the proliferation of T and B cells, and further induced the activation of T lymphocytes in vitro. Moreover, CDP-DFNS/H9N2 significantly promoted the antigen-specific antibodies levels in serum and intestinal mucosal of chickens, indicating the good ability to elicit both systemic and mucosal immunity. Additional, CDP-DFNS facilitate the activation of CD4 + and CD8 + T cells both in spleen and intestinal mucosal, and the indexes of immune organs. This study suggested that CDP-DFNS may be a new avenue for development of oral vaccine against pathogens that are transmitted via mucosal route.
Collapse
Affiliation(s)
- Jin He
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Xuanqi Lu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Ningning Mao
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Tianyu Zhu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Lin Yu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Yaming Yu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Song Peng
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiangwen Deng
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Bing Hu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Wenming Jiang
- China Animal Health and Epidemiology Center, Qingdao, China
| | - Yu Lu
- Institute of Veterinary Immunology & Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Deyun Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
10
|
Liu T, Li M, Tian Y, Dong Y, Liu N, Wang Z, Zhang H, Zheng A, Cui C. Immunogenicity and safety of a self-assembling ZIKV nanoparticle vaccine in mice. Int J Pharm 2024; 660:124320. [PMID: 38866086 DOI: 10.1016/j.ijpharm.2024.124320] [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: 02/14/2024] [Revised: 05/07/2024] [Accepted: 06/06/2024] [Indexed: 06/14/2024]
Abstract
Zika virus (ZIKV) is a mosquito-borne flavivirus that highly susceptibly causes Guillain-Barré syndrome and microcephaly in newborns. Vaccination is one of the most effective measures for preventing infectious diseases. However, there is currently no approved vaccine to prevent ZIKV infection. Here, we developed nanoparticle (NP) vaccines by covalently conjugating self-assembled 24-subunit ferritin to the envelope structural protein subunit of ZIKV to achieve antigen polyaggregation. The immunogenicityof the NP vaccine was evaluated in mice. Compared to monomer vaccines, the NP vaccine achieved effective antigen presentation, promoted the differentiation of follicular T helper cells in lymph nodes, and induced significantly greater antigen-specific humoral and cellular immune responses. Moreover, the NP vaccine enhanced high-affinity antigen-specific IgG antibody levels, increased secretion of the cytokines IL-4 and IFN-γ by splenocytes, significantly activated T/B lymphocytes, and improved the generation of memory T/B cells. In addition, no significant adverse reactions occurred when NP vaccine was combined with adjuvants. Overall, ferritin-based NP vaccines are safe and effective ZIKV vaccine candidates.
Collapse
Affiliation(s)
- Ting Liu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China; Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing 100069, China; Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing 100069, China; Beijing Laboratory of Biomedical Materials, Beijing 100069, China
| | - Meng Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Yang Tian
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China; Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing 100069, China; Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing 100069, China; Beijing Laboratory of Biomedical Materials, Beijing 100069, China
| | - Yuhan Dong
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Nan Liu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Zengming Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Hui Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Aiping Zheng
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Chunying Cui
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China; Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing 100069, China; Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing 100069, China; Beijing Laboratory of Biomedical Materials, Beijing 100069, China.
| |
Collapse
|
11
|
Yu H, Chen G, Li L, Wei G, Li Y, Xiong S, Qi X. Spider minor ampullate silk protein nanoparticles: an effective protein delivery system capable of enhancing systemic immune responses. MedComm (Beijing) 2024; 5:e573. [PMID: 38882211 PMCID: PMC11179522 DOI: 10.1002/mco2.573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 03/17/2024] [Accepted: 04/05/2024] [Indexed: 06/18/2024] Open
Abstract
Spider silk proteins (spidroins) are particularly attractive due to their excellent biocompatibility. Spider can produce up to seven different types of spidroins, each with unique properties and functions. Spider minor ampullate silk protein (MiSp) might be particularly interesting for biomedical applications, as the constituent silk is mechanically strong and does not super-contract in water, attributed to its amino acid composition. In this study, we evaluate the potential of recombinant nanoparticles derived from Araneus ventricosus MiSp as a protein delivery carrier. The MiSp-based nanoparticles were able to serve as an effective delivery system, achieving nearly 100% efficiency in loading the model protein lysozyme, and displayed a sustained release profile at physiological pH. These nanoparticles could significantly improve the delivery efficacy of the model proteins through different administration routes. Furthermore, nanoparticles loaded with model protein antigen lysozyme after subcutaneous or intramuscular administration could enhance antigen-specific immune responses in mouse models, through a mechanism involving antigen-depot effects at the injection site, long-term antigen persistence, and efficient uptake by dendritic cells as well as internalization by lymph nodes. These findings highlight the transnational potential of MiSp-based nanoparticle system for protein drug and vaccine delivery.
Collapse
Affiliation(s)
- Hairui Yu
- The Jiangsu Key Laboratory of Infection and Immunity Institutes of Biology and Medical Sciences Soochow University Suzhou China
| | - Gefei Chen
- Department of Biosciences and Nutrition Karolinska Institutet Huddinge Sweden
| | - Linchao Li
- The Jiangsu Key Laboratory of Infection and Immunity Institutes of Biology and Medical Sciences Soochow University Suzhou China
| | - Guoqiang Wei
- The Jiangsu Key Laboratory of Infection and Immunity Institutes of Biology and Medical Sciences Soochow University Suzhou China
| | - Yanan Li
- Department of Neurosurgery Changhai Hospital Naval Medical University Shanghai China
| | - Sidong Xiong
- The Jiangsu Key Laboratory of Infection and Immunity Institutes of Biology and Medical Sciences Soochow University Suzhou China
| | - Xingmei Qi
- The Jiangsu Key Laboratory of Infection and Immunity Institutes of Biology and Medical Sciences Soochow University Suzhou China
| |
Collapse
|
12
|
Gu P, Xu P, Zhu Y, Zhao Q, Zhao X, Fan Y, Wang X, Ma N, Bao Y, Shi W. Structural characterization and adjuvant activity of a water soluble polysaccharide from Poria cocos. Int J Biol Macromol 2024; 273:133067. [PMID: 38866287 DOI: 10.1016/j.ijbiomac.2024.133067] [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: 02/16/2024] [Revised: 04/21/2024] [Accepted: 06/08/2024] [Indexed: 06/14/2024]
Abstract
Adjuvants, as the essential component of vaccines, are crucial in enhancing the magnitude, breadth and durability of immune responses. Unfortunately, commonly used Alum adjuvants predominantly provoke humoral immune response, but fail to evoke cellular immune response, which is crucial for the prevention of various chronic infectious diseases and cancers. Thus, it is necessary to develop effective adjuvants to simultaneously induce humoral and cellular immune response. In this work, we obtained a water soluble polysaccharide isolated and purified from Poria cocos, named as PCP, and explored the possibility of PCP as a vaccine adjuvant. The PCP, with Mw of 20.112 kDa, primarily consisted of →6)-α-D-Galp-(1→, with a small amount of →3)-β-D-Glcp-(1 → and →4)-β-D-Glcp-(1→. Our results demonstrated that the PCP promoted the activation of dendritic cells (DCs) and macrophages in vitro. As the adjuvant to ovalbumin, the PCP facilitated the activation of DCs in lymph nodes, and evoked strong antibody response with a combination of Th1 and Th2 immune responses. Moreover, compared to Alum adjuvant, the PCP markedly induced a potent cellular response, especially the cytotoxic T lymphocytes response. Therefore, we confirmed that the PCP has great potential to be an available adjuvant for simultaneously inducing humoral and cellular immune responses.
Collapse
Affiliation(s)
- Pengfei Gu
- 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
| | - Yixuan Zhu
- 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
| | - Yingsai Fan
- 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
| | - Ning Ma
- 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
|
13
|
Ye JJ, Bao P, Deng K, Dong X, He J, Xia Y, Wang Z, Liu X, Tang Y, Feng J, Zhang XZ. Engineering cancer cell membranes with endogenously upregulated HSP70 as a reinforced antigenic repertoire for the construction of material-free prophylactic cancer vaccines. Acta Biomater 2024; 174:386-399. [PMID: 38016511 DOI: 10.1016/j.actbio.2023.11.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 11/20/2023] [Accepted: 11/22/2023] [Indexed: 11/30/2023]
Abstract
Immune cells distinguish cancer cells mainly relying on their membrane-membrane communication. The major challenge of cancer vaccines exists in difficult identification of cancer neoantigens and poor understanding over immune recognition mechanisms against cancer cells, particularly the combination among multiple antigens and the cooperation between antigens and immune-associated proteins. We exploit cancer cell membranes as the whole cancer antigen repertoire and reinforce its immunogenicity by cellular engineering to modulate the cytomembrane's immune-associated functions. This study reports a vaccine platform based on radiation-engineered cancer cells, of which the membrane HSP70 protein as the immune chaperon/traitor is endogenously upregulated. The resulting positive influences are shown to cover immunogenic steps occurring in antigen-presenting cells, including the uptake and the cross-presentation of the cancer antigens, thus amplifying cancer-specific immunogenicity. Membrane vaccines offer chances to introduce desired metal ions through membrane-metal complexation. Using Mn2+ ion as the costimulatory interferon genes agonist, immune activity is enhanced to further boost adaptive cancer immunogenicity. Results have evidenced that this artificially engineered membrane vaccine with favorable bio-safety could considerably reduce tumorigenicity and inhibit tumor growth. This study provides a universally applicable and facilely available cancer vaccine platform by artificial engineering of cancer cells to inherit and amplify the natural merits of cancer cell membranes. STATEMENT OF SIGNIFICANCE: The major challenge of cancer vaccines exists in difficult identification of cancer neoantigens and poor understanding over immune recognition mechanisms against cancer cells, particularly the combination among multiple antigens and the cooperation between antigens and immune-associated proteins. Cancer cell membrane presents superior advantages as the whole cancer antigen repertoire, including the reported and the unidentified antigens, but its immunogenicity is far from satisfactory. Cellular engineering approaches offer chances to endogenously modulate the immune-associated functions of cell membranes. Such a reinforced vaccine based on the engineered cancer cell membranes matches better the natural immune recognition pathway than the conventional vaccines.
Collapse
Affiliation(s)
- Jing-Jie Ye
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Peng Bao
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Kai Deng
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, PR China
| | - Xue Dong
- The Institute for Advanced Studies, Wuhan University, Wuhan 430072, PR China
| | - Jinlian He
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Yu Xia
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Ziyang Wang
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Xinhua Liu
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Ying Tang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine of Ministry of Education (KLOBM) School and Hospital of Stomatology, Wuhan University, Wuhan 430079, PR China
| | - Jun Feng
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, PR China.
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| |
Collapse
|
14
|
Xiong H, Han X, Cai L, Zheng H. Natural polysaccharides exert anti-tumor effects as dendritic cell immune enhancers. Front Oncol 2023; 13:1274048. [PMID: 37876967 PMCID: PMC10593453 DOI: 10.3389/fonc.2023.1274048] [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: 08/07/2023] [Accepted: 09/18/2023] [Indexed: 10/26/2023] Open
Abstract
With the development of immunotherapy, the process of tumor treatment is also moving forward. Polysaccharides are biological response modifiers widely found in plants, animals, fungi, and algae and are mainly composed of monosaccharides covalently linked by glycosidic bonds. For a long time, polysaccharides have been widely used clinically to enhance the body's immunity. However, their mechanisms of action in tumor immunotherapy have not been thoroughly explored. Dendritic cells (DCs) are a heterogeneous population of antigen presenting cells (APCs) that play a crucial role in the regulation and maintenance of the immune response. There is growing evidence that polysaccharides can enhance the essential functions of DCs to intervene the immune response. This paper describes the research progress on the anti-tumor immune effects of natural polysaccharides on DCs. These studies show that polysaccharides can act on pattern recognition receptors (PRRs) on the surface of DCs and activate phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT), mitogen-activated protein kinase (MAPK), nuclear factor-κB (NF-κB), Dectin-1/Syk, and other signalling pathways, thereby promoting the main functions of DCs such as maturation, metabolism, antigen uptake and presentation, and activation of T cells, and then play an anti-tumor role. In addition, the application of polysaccharides as adjuvants for DC vaccines, in combination with adoptive immunotherapy and immune checkpoint inhibitors (ICIs), as well as their co-assembly with nanoparticles (NPs) into nano drug delivery systems is also introduced. These results reveal the biological effects of polysaccharides, provide a new perspective for the anti-tumor immunopharmacological research of natural polysaccharides, and provide helpful information for guiding polysaccharides as complementary medicines in cancer immunotherapy.
Collapse
Affiliation(s)
- Hongtai Xiong
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xinpu Han
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Liu Cai
- The First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Honggang Zheng
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
15
|
Xu S, Li W, Jiao C, Cao Z, Wu F, Yan F, Wang H, Feng N, Zhao Y, Yang S, Wang J, Xia X. A Bivalent Bacterium-like Particles-Based Vaccine Induced Potent Immune Responses against the Sudan Virus and Ebola Virus in Mice. Transbound Emerg Dis 2023; 2023:9248581. [PMID: 40303775 PMCID: PMC12017122 DOI: 10.1155/2023/9248581] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 03/04/2023] [Accepted: 03/06/2023] [Indexed: 05/02/2025]
Abstract
Ebola virus disease (EVD) is an acute viral hemorrhagic fever disease causing thousands of deaths. The large Ebola outbreak in 2014-2016 posed significant threats to global public health, requiring the development of multiple medical measures for disease control. Sudan virus (SUDV) and Zaire virus (EBOV) are responsible for severe disease and occasional deadly outbreaks in West Africa and Middle Africa. This study shows that bivalent bacterium-like particles (BLPs)-based vaccine, SUDV-EBOV BLPs (S/ZBLP + 2 + P), generated by mixing SUDV-BLPs and EBOV-BLPs at a 1 : 1 ratio, is immunogenic in mice. The SUDV-EBOV BLPs induced potent immune responses against SUDV and EBOV and elicited both T-helper 1 (Th1) and T-helper 2 (Th2) immune responses. The results indicated that SUDV-EBOV BLPs-based vaccine has the potential to be a promising candidate against SUDV and EBOV infections and provide a strategy to develop universal vaccines for EVD.
Collapse
Affiliation(s)
- Shengnan Xu
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
- Shandong Agricultural University, Taian, China
| | - Wujian Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Cuicui Jiao
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Zengguo Cao
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Fangfang Wu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Feihu Yan
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Hualei Wang
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Na Feng
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Yongkun Zhao
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Songtao Yang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Jianzhong Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
| | - Xianzhu Xia
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
- Collaborative Innovation Center for Healthy Sheep Breeding and Zoonoses Prevention and Control, Shihezi University, Shihezi, Xinjiang, China
| |
Collapse
|
16
|
Agallou M, Margaroni M, Tsanaktsidou E, Badounas F, Kammona O, Kiparissides C, Karagouni E. A liposomal vaccine promotes strong adaptive immune responses via dendritic cell activation in draining lymph nodes. J Control Release 2023; 356:386-401. [PMID: 36893900 DOI: 10.1016/j.jconrel.2023.03.006] [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: 10/13/2022] [Revised: 02/14/2023] [Accepted: 03/03/2023] [Indexed: 03/11/2023]
Abstract
Subunit proteins provide a safe source of antigens for vaccine development especially for intracellular infections which require the induction of strong cellular immune responses. However, those antigens are often limited by their low immunogenicity. In order to achieve effective immune responses, they should be encapsulated into a stable antigen delivery system combined with an appropriate adjuvant. As such cationic liposomes provide an efficient platform for antigen delivery. In the present study, we describe a liposomal vaccine platform for co-delivery of antigens and adjuvants able to elicit strong antigen-specific adaptive immune responses. Liposomes are composed of the cationic lipid dimethyl dioctadecylammonium bromide (DDAB), cholesterol (CHOL) and oleic acid (OA). Physicochemical characterization of the formulations showed that their size was in the range of ∼250 nm with a positive zeta potential which was affected in some cases by the enviromental pH facilitating endosomal escape of potential vaccine cargo. In vitro, liposomes were effectively taken up by bone marrow dendritic cells (BMDCs) and when encapsulated IMQ they promoted BMDCs maturation and activation. Upon in vivo intramuscular administration, liposomes' active drainage to lymph nodes was mediated by DCs, B cells and macrophages. Thus, mice immunization with liposomes having encapsulated LiChimera, a previously characterized anti-leishmanial antigen, and IMQ elicited infiltration of CD11blow DCs populations in draining LNs followed by increased antigen-specific IgG, IgG2a and IgG1 levels production as well as indcution of antigen-specific CD4+ and CD8+ T cells. Collectively, the present work provides a proof-of-concept that cationic liposomes composed of DDAB, CHOL and OA adjuvanted with IMQ provide an efficient delivery platform for protein antigens able to induce strong adaptive immune responses via DCs targeting and induction of maturation.
Collapse
Affiliation(s)
- Maria Agallou
- Immunology of Infection Laboratory, Hellenic Pasteur Institute, Athens 125 21, Greece
| | - Maritsa Margaroni
- Immunology of Infection Laboratory, Hellenic Pasteur Institute, Athens 125 21, Greece
| | - Evgenia Tsanaktsidou
- Chemical Process & Energy Resources Institute, Centre for Research and Technology Hellas, P.O. Box 60361, Thessaloniki 57 001, Greece
| | - Fotis Badounas
- Molecular Genetics Laboratory, Department of Immunology, Transgenic Technology Laboratory, Hellenic Pasteur Institute, Athens 125 21, Greece
| | - Olga Kammona
- Chemical Process & Energy Resources Institute, Centre for Research and Technology Hellas, P.O. Box 60361, Thessaloniki 57 001, Greece
| | - Costas Kiparissides
- Chemical Process & Energy Resources Institute, Centre for Research and Technology Hellas, P.O. Box 60361, Thessaloniki 57 001, Greece; Department of Chemical Engineering, Aristotle University of Thessaloniki, P.O. Box 472, Thessaloniki 54 124, Greece
| | - Evdokia Karagouni
- Immunology of Infection Laboratory, Hellenic Pasteur Institute, Athens 125 21, Greece.
| |
Collapse
|
17
|
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
|
18
|
Hashemi Goradel N, Nemati M, Bakhshandeh A, Arashkia A, Negahdari B. Nanovaccines for cancer immunotherapy: Focusing on complex formation between adjuvant and antigen. Int Immunopharmacol 2023; 117:109887. [PMID: 36841155 DOI: 10.1016/j.intimp.2023.109887] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 01/29/2023] [Accepted: 02/10/2023] [Indexed: 02/27/2023]
Abstract
As an interesting cancer immunotherapy approach, cancer vaccines have been developed to deliver tumor antigens and adjuvants to antigen-presenting cells (APCs). Although the safety and easy production shifted the vaccine designing platforms toward the subunit vaccines, their efficacy is limited due to inefficient vaccine delivery. Nanotechnology-based vaccines, called nanovaccines, address the delivery limitations through co-delivery of antigens and adjuvants into lymphoid organs and APCs and their intracellular release, leading to cross-presentation of antigens and induction of potent anti-tumor immune responses. Although the nanovaccines, either as encapsulating agents or biomimetic nanoparticles, exert the desired anti-tumor activities, there is evidence that the mixing formulation to form nanocomplexes between antigens and adjuvants based on the electrostatic interactions provokes high levels of immune responses owing to Ags' availability and faster release. Here, we summarized the various platforms for developing cancer vaccines and the advantages of using delivery systems. The cancer nanovaccines, including nanoparticle-based and biomimetic-based nanovaccines, are discussed in detail. Finally, we focused on the nanocomplexes formation between antigens and adjuvants as promising cancer nanovaccine platforms.
Collapse
Affiliation(s)
- Nasser Hashemi Goradel
- Department of Medical Biotechnology, Maragheh University of Medical Sciences, Maragheh, Iran.
| | - Mahnaz Nemati
- Amir Oncology Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Azam Bakhshandeh
- Department of Industrial Engineering and Management Systems, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Arash Arashkia
- Department of Molecular Virology, Pasteur Institute of Iran, Tehran, Iran
| | - Babak Negahdari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
19
|
Yin Q, Wang Y, Xiang Y, Xu F. Nanovaccines: Merits, and diverse roles in boosting antitumor immune responses. Hum Vaccin Immunother 2022; 18:2119020. [PMID: 36170662 DOI: 10.1080/21645515.2022.2119020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
An attractive type of cancer immunotherapy is cancer therapeutic vaccines that induce antitumor immunity effectively. Although supportive results in the recent vaccine studies, there are still numerous drawbacks, such as poor stability, weak immunogenicity and strong toxicity, to be tackled for promoting the potency and durability of antitumor efficacy. NPs (Nanoparticles)-based vaccines offer unique opportunities to breakthrough the current bottleneck. As a rule, nanovaccines are new the generations of vaccines that use NPs as carriers and/or adjuvants. Several advantages of nanovaccines are constantly explored, including optimal nanometer size, high stability, plenty of antigen loading, enhanced immunogenicity, tunable antigen presentation, more retention in lymph nodes and promote patient compliance by a lower frequency of dosing. Here, we summarized the merits and highlight the diverse role nanovaccines play in improving antitumor responses.
Collapse
Affiliation(s)
- Qiliang Yin
- Department of Cadre Ward, The First Hospital of Jilin University, Changchun, China
| | - Ying Wang
- Academy of Health Management, Changchun University of Chinese Medicine, Changchun, China
| | - Yipeng Xiang
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Feng Xu
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
20
|
Dai H, Huang Y, Guo J, Li L, Ke Y, Cen L, Meng F, Chen X, Liu B, Qian X. Engineering a HemoMap Nanovaccine for Inducing Immune Responses against Melanoma. ACS APPLIED MATERIALS & INTERFACES 2022; 14:52634-52642. [PMID: 36383430 DOI: 10.1021/acsami.2c14379] [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: 06/16/2023]
Abstract
Neoantigen vaccines have opened a new paradigm for cancer immunotherapy. Here, we constructed a neoantigen nanovaccine-HemoMap, with the ability to target lymph nodes and activate immune cells. We propose a HemoMap nanovaccine consisting of the mouse melanoma highly expressed antigenic peptide Tyrp1 and a magnesium nanoadjuvant-HemoM. By immunofluorescence labeling of the nanovaccine, the lymph node targeting of the vaccine was observed and verified by a mouse near-infrared imaging system. About two-fold higher effective retention of HemoMap induces the internalization of Tyrp1 in DCs than that of free Tyrp1 in draining lymph nodes (DLNs) for 48 h. A mouse melanoma subcutaneous model was established to evaluate neoantigen-specific antitumor immune responses. In comparison to the control group, the tumor growth rate was dramatically slowed down by HemoMap treatment, and the median survival time was extended by 7 days. We discovered that effective co-delivery of Tyrp1 antigen and magnesium (Mg2+) to lymph nodes (LNs) boosted cellular internalization and activated immune cells, such as CD11c+ DCs and CD8+ T lymphocytes. Spleen lymphocytes from the HemoMap group displayed much more antitumor activity than those from the other groups. Our findings highlight that HemoMap is promising to trigger T cell responses and to provide novel nanoadjuvants strategies for cancer immunotherapy.
Collapse
Affiliation(s)
- Hengheng Dai
- The Comprehensive Cancer Centre, China Pharmaceutical University Nanjing Drum Tower Hospital, 321 Zhongshan Road, Nanjing 210008, China
| | - Ying Huang
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing 210008, China
| | - Jingyi Guo
- The Comprehensive Cancer Centre, China Pharmaceutical University Nanjing Drum Tower Hospital, 321 Zhongshan Road, Nanjing 210008, China
| | - Lin Li
- Department of Pathology, Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, China
| | - Yaohua Ke
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, China
| | - Lanqi Cen
- The Comprehensive Cancer Centre, China Pharmaceutical University Nanjing Drum Tower Hospital, 321 Zhongshan Road, Nanjing 210008, China
| | - Fanyan Meng
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, China
| | - Xinjie Chen
- The Comprehensive Cancer Centre, China Pharmaceutical University Nanjing Drum Tower Hospital, 321 Zhongshan Road, Nanjing 210008, China
| | - Baorui Liu
- The Comprehensive Cancer Centre, China Pharmaceutical University Nanjing Drum Tower Hospital, 321 Zhongshan Road, Nanjing 210008, China
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, China
| | - Xiaoping Qian
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, China
| |
Collapse
|
21
|
Flower-like mesoporous silica nanoparticles as an antigen delivery platform to promote systemic immune response. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2022; 42:102541. [PMID: 35181525 DOI: 10.1016/j.nano.2022.102541] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 12/06/2021] [Accepted: 02/04/2022] [Indexed: 12/31/2022]
Abstract
Virus-like particles (VLPs), a kind of superior subunit vaccine, are assembled from the viral structural proteins with similar capsids to viruses. However, the efficiency of cell uptake is not satisfactory. We prepared flower-like mesoporous silica nanoparticles (SiNPs) with large pore channels and interior cavities to solve the problem. The highly loaded VLPs-SiNPs composites not only enhanced the stability of VLPs, but also delivered antigen to cells and improved the cellular uptake efficiency. Compared with naked VLPs, mice intramuscularly immunized with the VLPs-SiNPs composite induced higher specific antibodies, greater lymphocyte activation and higher level of cytokine secretion. Moreover, the VLPs-SiNPs composite as vaccine also promoted mucosal immune response through intranasal immune pathway. Therefore, the VLPs-SiNPs enable to induce strong cellular, humoral, and slight mucosal immune response through different immunization routes. These results are potentially useful for vaccine formulations and may provide further reference for vaccine design and delivery systems.
Collapse
|
22
|
Surwase SS, Shahriar SMS, An JM, Ha J, Mirzaaghasi A, Bagheri B, Park JH, Lee YK, Kim YC. Engineered Nanoparticles inside a Microparticle Oral System for Enhanced Mucosal and Systemic Immunity. ACS APPLIED MATERIALS & INTERFACES 2022; 14:11124-11143. [PMID: 35227057 DOI: 10.1021/acsami.1c24982] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Antigen delivery through an oral route requires overcoming multiple challenges, including gastrointestinal enzymes, mucus, and epithelial tight junctions. Although each barrier has a crucial role in determining the final efficiency of the oral vaccination, transcytosis of antigens through follicle-associated epithelium (FAE) represents a major challenge. Most of the research is focused on delivering an antigen to the M-cell for FAE transcytosis because M-cells can easily transport the antigen from the luminal site. However, the fact is that the M-cell population is less than 1% of the total gastrointestinal cells, and most of the oral vaccines have failed to show any effect in clinical trials. To challenge the current dogma of M-cell targeting, in this study, we designed a novel tandem peptide with a FAE-targeting peptide at the front position and a cell-penetrating peptide at the back position. The tandem peptide was attached to a smart delivery system, which overcomes the enzymatic barrier and the mucosal barrier. The result showed that the engineered system could target the FAE (enterocytes and M-cells) and successfully penetrate the enterocytes to reach the dendritic cells located at the subepithelium dome. There was successful maturation and activation of dendritic cells in vitro confirmed by a significant increase in maturation markers such as CD40, CD86, presentation marker MHC I, and proinflammatory cytokines (TNF-α, IL-6, and IL-10). The in vivo results showed a high production of CD4+ T-lymphocytes (helper T-cell) and a significantly higher production of CD8+ T-lymphocytes (killer T-cell). Finally, the production of mucosal immunity (IgA) in the trachea, intestine, and fecal extracts and systemic immunity (IgG, IgG1, and IgG2a) was successfully confirmed. To the best of our knowledge, this is the first study that designed a novel tandem peptide to target the FAE, which includes M-cells and enterocytes rather than M-cell targeting and showed that a significant induction of both the mucosal and systemic immune response was achieved compared to M-cell targeting.
Collapse
Affiliation(s)
- Sachin S Surwase
- Department of Chemical & Biomolecular Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - S M Shatil Shahriar
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198-5940, United States
- KB Biomed Inc., Chungju 27469, Republic of Korea
- Department of Chemical & Biological Engineering, Korea National University of Transportation, Chungju 27469, Republic of Korea
| | - Jeong Man An
- Department of Chemical & Biological Engineering, Korea National University of Transportation, Chungju 27469, Republic of Korea
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, Republic of Korea
| | - JongHoon Ha
- Department of Chemical & Biomolecular Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Amin Mirzaaghasi
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Babak Bagheri
- Department of Chemical & Biomolecular Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Ji-Ho Park
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Yong-Kyu Lee
- KB Biomed Inc., Chungju 27469, Republic of Korea
- Department of Chemical & Biological Engineering, Korea National University of Transportation, Chungju 27469, Republic of Korea
| | - Yeu-Chun Kim
- Department of Chemical & Biomolecular Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| |
Collapse
|
23
|
Yang Y, Sun Z, Li H, Tian J, Chen M, Liu T. Preparation and Immune Effect of HEV ORF2 P206@PLGA Nanoparticles. NANOMATERIALS 2022; 12:nano12040595. [PMID: 35214924 PMCID: PMC8878542 DOI: 10.3390/nano12040595] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/28/2022] [Accepted: 02/03/2022] [Indexed: 12/12/2022]
Abstract
The hepatitis E virus (HEV) is an important pathogen that threatens global public health. One-third of the world’s population lives in the epidemic area of HEV, causing 20 million infections and 70,000 deaths annually. In China, HEV transmission has changed from human-to-human transmission of HEV1 to zoonotic transmission of HEV4, causing hepatitis outbreaks throughout the country. Protecting vulnerable groups, such as practitioners related to animal husbandry and downstream consumers who are immune deficient or pregnant, from HEV infections is an urgent task. At present, the commercial human vaccine, Hecolin® (HEV 239 vaccine), is licensed for use only in China. HEV 239 vaccine is a human vaccine developed for HEV1. Although it has a cross-protective effect on HEV4, the level of immune protection is still different. To address the transformation of domestic HEV transmission modes, there is an urgent need to develop a new vaccine against zoonotic HEV4. P206@PLGA is a vaccine candidate in which nanomaterials are used to encapsulate viral capsid proteins for the immunization of livestock animals. Our experiments show that P206@PLGA has excellent biocompatibility and safety. In addition, P206@PLGA can effectively induce animals to produce a high titer of antibodies against HEV4, and thus has the potential to become a veterinary vaccine for the prevention of HEV. This approach provides a new concept for HE prevention to reduce the transmission of HEV in farms and protect susceptible populations.
Collapse
Affiliation(s)
| | | | | | | | - Mingyong Chen
- Correspondence: (M.C.); (T.L.); Tel.: +86-010-62733398 (T.L.)
| | - Tianlong Liu
- Correspondence: (M.C.); (T.L.); Tel.: +86-010-62733398 (T.L.)
| |
Collapse
|
24
|
Hendy DA, Amouzougan EA, Young IC, Bachelder EM, Ainslie KM. Nano/microparticle Formulations for Universal Influenza Vaccines. AAPS J 2022; 24:24. [PMID: 34997352 PMCID: PMC8741137 DOI: 10.1208/s12248-021-00676-9] [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: 10/08/2021] [Accepted: 12/17/2021] [Indexed: 11/30/2022] Open
Abstract
Influenza affects millions of people worldwide and can result in severe sickness and even death. The best method of prevention is vaccination; however, the seasonal influenza vaccine often suffers from low efficacy and requires yearly vaccination due to changes in strain and viral mutations. More conserved universal influenza antigens like M2 ectodomain (M2e) and the stalk region of hemagglutinin (HA stalk) have been used clinically but often suffer from low antigenicity. To increase antigenicity, universal antigens have been formulated using nano/microparticles as vaccine carriers against influenza. Utilizing polymers, liposomes, metal, and protein-based particles, indicators of immunity and protection in mouse, pig, ferrets, and chicken models of influenza have been shown. In this review, seasonal and universal influenza vaccine formulations comprised of these materials including their physiochemical properties, fabrication, characterization, and biologic responses in vivo are highlighted. The review is concluded with future perspectives for nano/microparticles as carrier systems and other considerations within the universal influenza vaccine delivery landscape. Graphical Abstract ![]()
Collapse
Affiliation(s)
- Dylan A Hendy
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, 4012 Marsico Hall, 125 Mason Farm Road, Chapel Hill, North Carolina, 27599, USA
| | - Eva A Amouzougan
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, 4012 Marsico Hall, 125 Mason Farm Road, Chapel Hill, North Carolina, 27599, USA
| | - Isabella C Young
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, 4012 Marsico Hall, 125 Mason Farm Road, Chapel Hill, North Carolina, 27599, USA
| | - Eric M Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, 4012 Marsico Hall, 125 Mason Farm Road, Chapel Hill, North Carolina, 27599, USA
| | - Kristy M Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, 4012 Marsico Hall, 125 Mason Farm Road, Chapel Hill, North Carolina, 27599, USA. .,Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina, USA. .,Department of Microbiology and Immunology, UNC School of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA.
| |
Collapse
|
25
|
Han S, Ma W, Jiang D, Sutherlin L, Zhang J, Lu Y, Huo N, Chen Z, Engle JW, Wang Y, Xu X, Kang L, Cai W, Wang L. Intracellular signaling pathway in dendritic cells and antigen transport pathway in vivo mediated by an OVA@DDAB/PLGA nano-vaccine. J Nanobiotechnology 2021; 19:394. [PMID: 34838057 PMCID: PMC8626881 DOI: 10.1186/s12951-021-01116-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 11/02/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Poly(D, L-lactic-co-glycolic acid) (PLGA) nanoparticles have potential applications as a vaccine adjuvant and delivery system due to its unique advantages as biodegradability and biocompatibility. EXPERIMENTAL We fabricated cationic solid lipid nanoparticles using PLGA and dimethyl-dioctadecyl-ammonium bromide (DDAB), followed by loading of model antigen OVA (antigen ovalbumin, OVA257-264) to form an OVA@DDAB/PLGA nano-vaccine. And we investigated the intracellular signaling pathway in dendritic cells in vitro and antigen transport pathway and immune response in vivo mediated by an OVA@DDAB/PLGA nano-vaccine. RESULTS In vitro experiments revealed that the antigen uptake of BMDCs after nanovaccine incubation was two times higher than pure OVA or OVA@Al at 12 h. The BMDCs were well activated by p38 MAPK signaling pathway. Furthermore, the nano-vaccine induced antigen escape from lysosome into cytoplasm with 10 times increased cross-presentation activity than those of OVA or OVA@Al. Regarding the transport of antigen into draining lymph nodes (LNs), the nano-vaccine could rapidly transfer antigen to LNs by passive lymphatic drainage and active DC transport. The antigen+ cells in inguinal/popliteal LNs for the nano-vaccine were increased over two folds comparing to OVA@Al and OVA at 12 h. Moreover, the antigen of nano-vaccine stayed in LNs for over 7 days, germinal center formation over two folds higher than those of OVA@Al and OVA. After immunization, the nano-vaccine induced a much higher ratio of IgG2c/IgG1 than OVA@Al. It also effectively activated CD4+ T, CD8+ T and B cells for immune memory with a strong cellular response. CONCLUSION These results indicated that DDAB/PLGA NP was a potent platform to improve vaccine immunogenicity by p38 signaling pathway in BMDCs, enhancing transport of antigens to LNs, and higher immunity response.
Collapse
Affiliation(s)
- Shulan Han
- Key Laboratory of Green Process and Engineering, State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, People's Republic of China
| | - Wenyan Ma
- Key Laboratory of Green Process and Engineering, State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
- Tianjin University of Science and Technology, Tianjin, 300222, People's Republic of China
| | - Dawei Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Logan Sutherlin
- Departments of Radiology and Medical Physics, University of Wisconsin - Madison, Madison, WI, 53705, USA
| | - Jing Zhang
- Key Laboratory of Green Process and Engineering, State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Yu Lu
- Institute of Veterinary Immunology &Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, People's Republic of China
| | - Nan Huo
- Department of Genetic Engineering Laboratory, Beijing Institute of Biotechnology, Beijing, 100850, People's Republic of China
| | - Zhao Chen
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, 100034, People's Republic of China
| | - Jonathan W Engle
- Departments of Radiology and Medical Physics, University of Wisconsin - Madison, Madison, WI, 53705, USA
| | - Yanping Wang
- Tianjin University of Science and Technology, Tianjin, 300222, People's Republic of China.
| | - Xiaojie Xu
- Department of Genetic Engineering Laboratory, Beijing Institute of Biotechnology, Beijing, 100850, People's Republic of China.
| | - Lei Kang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, 100034, People's Republic of China.
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin - Madison, Madison, WI, 53705, USA.
| | - Lianyan Wang
- Key Laboratory of Green Process and Engineering, State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, People's Republic of China.
- University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China.
| |
Collapse
|
26
|
Howard GP, Bender NG, Khare P, López-Gutiérrez B, Nyasembe V, Weiss WJ, Simecka JW, Hamerly T, Mao HQ, Dinglasan RR. Immunopotentiation by Lymph-Node Targeting of a Malaria Transmission-Blocking Nanovaccine. Front Immunol 2021; 12:729086. [PMID: 34512663 PMCID: PMC8432939 DOI: 10.3389/fimmu.2021.729086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/06/2021] [Indexed: 11/13/2022] Open
Abstract
A successful malaria transmission blocking vaccine (TBV) requires the induction of a high antibody titer that leads to abrogation of parasite traversal of the mosquito midgut following ingestion of an infectious bloodmeal, thereby blocking the cascade of secondary human infections. Previously, we developed an optimized construct UF6b that elicits an antigen-specific antibody response to a neutralizing epitope of Anopheline alanyl aminopeptidase N (AnAPN1), an evolutionarily conserved pan-malaria mosquito midgut-based TBV target, as well as established a size-controlled lymph node targeting biodegradable nanoparticle delivery system that leads to efficient and durable antigen-specific antibody responses using the model antigen ovalbumin. Herein, we demonstrate that co-delivery of UF6b with the adjuvant CpG oligodeoxynucleotide immunostimulatory sequence (ODN ISS) 1018 using this biodegradable nanoparticle vaccine delivery system generates an AnAPN1-specific immune response that blocks parasite transmission in a standard membrane feeding assay. Importantly, this platform allows for antigen dose-sparing, wherein lower antigen payloads elicit higher-quality antibodies, therefore less antigen-specific IgG is needed for potent transmission-reducing activity. By targeting lymph nodes directly, the resulting immunopotentiation of AnAPN1 suggests that the de facto assumption that high antibody titers are needed for a TBV to be successful needs to be re-examined. This nanovaccine formulation is stable at -20°C storage for at least 3 months, an important consideration for vaccine transport and distribution in regions with poor healthcare infrastructure. Together, these data support further development of this nanovaccine platform for malaria TBVs.
Collapse
Affiliation(s)
- Gregory P Howard
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, United States
| | - Nicole G Bender
- Emerging Pathogens Institute, Department of Infectious Diseases & Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - Prachi Khare
- Emerging Pathogens Institute, Department of Infectious Diseases & Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - Borja López-Gutiérrez
- Emerging Pathogens Institute, Department of Infectious Diseases & Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - Vincent Nyasembe
- Emerging Pathogens Institute, Department of Infectious Diseases & Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - William J Weiss
- Department of Pharmaceutical Sciences and UNTHSC Preclinical Services, University of North Texas System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Jerry W Simecka
- Department of Pharmaceutical Sciences and UNTHSC Preclinical Services, University of North Texas System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Timothy Hamerly
- Emerging Pathogens Institute, Department of Infectious Diseases & Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - Hai-Quan Mao
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, United States.,Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, United States.,Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore MD, United States
| | - Rhoel R Dinglasan
- Emerging Pathogens Institute, Department of Infectious Diseases & Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| |
Collapse
|
27
|
Chen J, Fang H, Hu Y, Wu J, Zhang S, Feng Y, Lin L, Tian H, Chen X. Combining mannose receptor mediated nanovaccines and gene regulated PD-L1 blockade for boosting cancer immunotherapy. Bioact Mater 2021; 7:167-180. [PMID: 34466725 PMCID: PMC8379363 DOI: 10.1016/j.bioactmat.2021.05.036] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 05/17/2021] [Accepted: 05/25/2021] [Indexed: 12/11/2022] Open
Abstract
Tumor nanovaccines have potential applications in the prevention and treatment of malignant tumors. However, it remains a longstanding challenge in exploiting efficient nanocarriers for inducing potent specifically cellular immune responses. Toward this objective, we herein explore an intensive tumor immunotherapeutic strategy by combining mannosylated nanovaccines and gene regulated PD-L1 blockade for immune stimulation and killing activity. Here, we fabricate a mannose modified PLL-RT (Man-PLL-RT) mediated nanovaccines with dendritic cells (DCs) targeting capacity. Man-PLL-RT is capable of co-encapsulating with antigen (ovalbumin, OVA) and adjuvant (unmethylated cytosine-phosphate-guanine, CpG) by electrostatic interaction. This positively charged Man-PLL-RT/OVA/CpG nanovaccines can facilitate the endocytosis, maturation and cross presentation in DCs. However, the nanovaccines arouse limited inhibition of tumor growth, which is mainly due to the immunosuppressed microenvironment of tumors. Combining tumor nanovaccines with gene regulated PD-L1 blockade leads to an obvious tumor remission in B16F10 melanoma bearing mice. The collaborative strategy provides essential insights to boost the benefits of tumor vaccines by regulating the checkpoint blockade with gene therapy. The nanovaccines are composed of polypeptides, which are bio-safe and biodegradable. The nanovaccines have APCs target function. Blocking PD-1/PD-L1 through gene therapy can reverse the tumor immune-tolerant microenvironment. The combined strategy provides a potentially effective strategy for the clinical treatment of tumors.
Collapse
Affiliation(s)
- Jie Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China.,University of Science and Technology of China, Hefei, 230026, PR China.,Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, PR China
| | - Huapan Fang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China.,University of Science and Technology of China, Hefei, 230026, PR China.,Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for CarbonBased Functional Materials & Devices, Soochow University, Suzhou, 215123, Jiangsu, PR China
| | - Yingying Hu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China.,University of Science and Technology of China, Hefei, 230026, PR China
| | - Jiayan Wu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China.,University of Science and Technology of China, Hefei, 230026, PR China
| | - Sijia Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China.,University of Science and Technology of China, Hefei, 230026, PR China
| | - Yuanji Feng
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China.,University of Science and Technology of China, Hefei, 230026, PR China
| | - Lin Lin
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China.,University of Science and Technology of China, Hefei, 230026, PR China.,Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, PR China
| | - Huayu Tian
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China.,University of Science and Technology of China, Hefei, 230026, PR China.,Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, PR China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China.,University of Science and Technology of China, Hefei, 230026, PR China.,Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, PR China
| |
Collapse
|
28
|
Zhu L, Lei Z, Xia X, Zhang Y, Chen Y, Wang B, Li J, Li G, Yang G, Cao G, Yin Z. Yeast Shells Encapsulating Adjuvant AS04 as an Antigen Delivery System for a Novel Vaccine against Toxoplasma Gondii. ACS APPLIED MATERIALS & INTERFACES 2021; 13:40415-40428. [PMID: 34470103 DOI: 10.1021/acsami.1c12366] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Toxoplasma gondii (T. gondii) infection causes severe zoonotic toxoplasmosis, which threatens the safety of almost one-third of the human population globally. However, there is no effective protective vaccine against human toxoplasmosis. This necessitates anti-T. gondii vaccine development, which is a main priority of public health. In this study, we optimized the adjuvant system 04 (AS04), a vaccine adjuvant constituted by 3-O-desacyl-4'-monophosphoryl lipid A (a TLR4 agonist) and aluminum salts, by packing it within natural extracts of β-glucan particles (GPs) from Saccharomyces cerevisiae to form a GP-AS04 hybrid adjuvant system. Through a simple mixing procedure, we loaded GP-AS04 particles with the total extract (TE) of T. gondii lysate, forming a novel anti-T. gondii vaccine GP-AS04-TE. Results indicated that the hybrid adjuvant can efficiently and stably load antigens, mediate antigen delivery, facilitate the dendritic uptake of antigens, boost dendritic cell maturation and stimulation, and increase the secretion of pro-inflammatory cytokines. In the mouse inoculation model, GP-AS04-TE significantly stimulated the function of dendritic cells, induced a very strong TE-specific humoral and cellular immune response, and finally showed a strong and effective protection against toxoplasma chronic and acute infections. This work proves the potential of GP-AS04 for exploitation as a vaccine against a range of pathogens.
Collapse
Affiliation(s)
- Leqing Zhu
- The First Affiliated Hospital, Faculty of Medical Science, Jinan University, Guangzhou 510632, China
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou 510632, China
| | - Zhiwei Lei
- Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou 510632, China
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511518, China
| | - Xichun Xia
- Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai 51900, China
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou 510632, China
| | - Yingying Zhang
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou 510632, China
| | - Yuyuan Chen
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou 510632, China
| | - Baocheng Wang
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, Shenzhen 518055, China
| | - Jiawei Li
- Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai 51900, China
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou 510632, China
| | - Guangqiang Li
- The First Affiliated Hospital, Faculty of Medical Science, Jinan University, Guangzhou 510632, China
| | - Guang Yang
- Department of Pathogen Biology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Guangchao Cao
- Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai 51900, China
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou 510632, China
| | - Zhinan Yin
- Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai 51900, China
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou 510632, China
| |
Collapse
|
29
|
He Z, Jia H, Zheng M, Wang H, Yang W, Gao L, Zhang Z, Xue J, Xu B, Yang W, Xing G, Gao X, Gao F. Trp2 Peptide-Assembled Nanoparticles with Intrinsically Self-Chelating 64Cu Properties for PET Imaging Tracking and Dendritic Cell-Based Immunotherapy against Melanoma. ACS APPLIED BIO MATERIALS 2021; 4:5707-5716. [PMID: 35006752 DOI: 10.1021/acsabm.1c00480] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Dendritic cell-based immunotherapy, in which the antigen is effectively delivered to dendritic cells and then the dendritic cells stimulated by the antigen migrate to draining lymph nodes (DLNs) to induce the CD8+ T-cell immune response, shows great promise for tumor immunotherapy. In this study, we used coassembled nanoparticles formed by Trp2 antigen and the conjugates of short-chain poly(ethylene glycol) (PEG) and pyropheophorbide-A (PPa) (Trp2/PPa-PEGm) to deliver Trp2 to DCs. Intrinsically self-chelating 64Cu of coassemblies could be used to sensitively image the migration of DCs in vivo by positron emission tomography (PET) imaging. The coassemblies of the Trp2 antigen were efficiently engulfed by DCs without causing DC cytotoxicity in vitro and induced DC maturation. After injection of DCs labeled by coassemblies of the Trp2 antigen, the homing of DCs to DLNs in vivo could be sensitively observed by PET imaging. The C57BL/6 mice injected with DCs containing the Trp2/PPa-PEGm NP showed antigen-specific immune responses including enhanced interferon-γ (IFN-γ) production, splenocyte proliferation, and percentage of IFN-γ-secreting CD8+ T cells. In addition, C57BL/6 mice inoculated with B16-F10 tumor cells showed delayed tumor growth after immunization with the Trp2/PPa-PEGm NP-labeled DC vaccine and enhanced infiltration of CD8+ T cells in tumors.
Collapse
Affiliation(s)
- Zhesheng He
- CAS Key Laboratory for the Biological Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huiju Jia
- CAS Key Laboratory for the Biological Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China.,School of Pharmacy, Hebei University, Baoding 071002, China
| | - Miaomiao Zheng
- CAS Key Laboratory for the Biological Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China.,School of Pharmacy, Hebei University, Baoding 071002, China
| | - Huangwei Wang
- CAS Key Laboratory for the Biological Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China.,School of Pharmacy, Hebei University, Baoding 071002, China
| | - Wenjiang Yang
- CAS Key Laboratory of Nuclear Radiation and Nuclear Energy Technology, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Liang Gao
- Department of Chemistry and Biology, Faculty of Environment and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Zhiyong Zhang
- CAS Key Laboratory for the Biological Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Jingquan Xue
- CAS Key Laboratory of Nuclear Radiation and Nuclear Energy Technology, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Baixuan Xu
- Department of Nuclear Medicine, Chinese PLA General Hospital, Beijing 100853, China
| | - Wenzhi Yang
- School of Pharmacy, Hebei University, Baoding 071002, China
| | - Gengmei Xing
- CAS Key Laboratory for the Biological Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Xueyun Gao
- Department of Chemistry and Biology, Faculty of Environment and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Fuping Gao
- CAS Key Laboratory for the Biological Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
30
|
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: 82] [Impact Index Per Article: 20.5] [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
|
31
|
Han L, Peng K, Qiu LY, Li M, Ruan JH, He LL, Yuan ZX. Hitchhiking on Controlled-Release Drug Delivery Systems: Opportunities and Challenges for Cancer Vaccines. Front Pharmacol 2021; 12:679602. [PMID: 34040536 PMCID: PMC8141731 DOI: 10.3389/fphar.2021.679602] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 04/28/2021] [Indexed: 12/11/2022] Open
Abstract
Cancer vaccines represent among the most promising strategies in the battle against cancers. However, the clinical efficacy of current cancer vaccines is largely limited by the lack of optimized delivery systems to generate strong and persistent antitumor immune responses. Moreover, most cancer vaccines require multiple injections to boost the immune responses, leading to poor patient compliance. Controlled-release drug delivery systems are able to address these issues by presenting drugs in a controlled spatiotemporal manner, which allows co-delivery of multiple drugs, reduction of dosing frequency and avoidance of significant systemic toxicities. In this review, we outline the recent progress in cancer vaccines including subunit vaccines, genetic vaccines, dendritic cell-based vaccines, tumor cell-based vaccines and in situ vaccines. Furthermore, we highlight the efforts and challenges of controlled or sustained release drug delivery systems (e.g., microparticles, scaffolds, injectable gels, and microneedles) in ameliorating the safety, effectiveness and operability of cancer vaccines. Finally, we briefly discuss the correlations of vaccine release kinetics and the immune responses to enlighten the rational design of the next-generation platforms for cancer therapy.
Collapse
Affiliation(s)
- Lu Han
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| | - Ke Peng
- School of pharmacy, Queen's University Belfast, Belfast, United Kingdom
| | - Li-Ying Qiu
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| | - Meng Li
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| | - Jing-Hua Ruan
- The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Li-Li He
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| | - Zhi-Xiang Yuan
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| |
Collapse
|
32
|
Gu P, Zhang Y, Cai G, Liu Z, Hu Y, Liu J, Wang D. Administration Routes of Polyethylenimine-Coated PLGA Nanoparticles Encapsulating Angelica Sinensis Polysaccharide Vaccine Delivery System Affect Immune Responses. Mol Pharm 2021; 18:2274-2284. [PMID: 33926191 DOI: 10.1021/acs.molpharmaceut.1c00090] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Nanoparticle vaccine delivery systems have been emerging strategies for inducing potent immune responses to prevent and treat infectious diseases and cancers. The properties of nanoparticle vaccine delivery systems, such as nanoparticle size, surface charge, and antigen release kinetics, have been extensively studied and proven to effectively influence the efficacy of vaccine responses. However, a few types of research have focused on the influence of administration routes of nanoparticle vaccines on immune responses. Herein, to investigate how the administration routes affect the immune responses of nanoparticles vaccines, we developed a nanoparticles system (NPs), in which the ovalbumin (OVA) and Angelica sinensis polysaccharide (ASP) were incorporated into poly(lactic-co-glycolic acid) (PLGA) nanoparticles and the polyethylenimine (PEI) was coated on the surface of nanoparticles. The NPs vaccine was intramuscularly and subcutaneously injected (im and sc) into mice, and the immune responses induced by these two delivery routes were compared. The results showed that both im and sc administration of NPs vaccines elicited strong antigen-specific IgG, IgG1, and IgG2a antibody responses, with no significant difference. In contrast, NP vaccines with sc administration significantly enhanced immune responses, such as enhancing the recruitment and activation of dendritic cells (DCs) in lymph nodes (LNs), promoting the antigen transport into draining lymph nodes, increasing the secretion of cytokines, improving the ratio of CD4+T cells to CD8+ T cells, activating cytotoxic T lymphocyte response, and inducing a strong cellular immune response. These results may provide a new insight onto the development of vaccine delivery systems.
Collapse
Affiliation(s)
- Pengfei Gu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Yue Zhang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Gaofeng Cai
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Zhenguang Liu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Yuanliang Hu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Jiaguo Liu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Deyun Wang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| |
Collapse
|
33
|
Li W, Jing Z, Wang S, Li Q, Xing Y, Shi H, Li S, Hong Z. P22 virus-like particles as an effective antigen delivery nanoplatform for cancer immunotherapy. Biomaterials 2021; 271:120726. [PMID: 33636548 DOI: 10.1016/j.biomaterials.2021.120726] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 02/09/2021] [Accepted: 02/15/2021] [Indexed: 12/11/2022]
Abstract
As a new strategy for cancer immunotherapy, therapeutic cancer vaccines have been greatly improved in recent years. However, addressing the needs to quickly and efficiently elicit a high-intensity immune response against neoantigen peptides, especially to induce an effective cytotoxic lymphocyte (CTL) reaction, remain challenges in this field. In this study, virus-like particles (VLPs) derived from the phage P22 were adopted to load peptide antigens on the surface, to test whether VLP technology can be used as a platform for efficient peptide antigen delivery by therapeutic cancer vaccines. The B and T epitopes (OVAB peptide and OVAT peptide) of ovalbumin (OVA) were used here as model antigens and fused individually at the C terminus of the coat protein (CP), which allowed display on the surface of P22 particles to form two types of vaccine particles (VLP-OVAB and VLP-OVAT). Subsequent experiments showed that VLP-OVAB induced an antibody titer against the peptide antigen as high as 5.0 × 105 and that VLP-OVAT induced highly effective cross-presentation and then strongly activated a T epitope-specific CTL response. Mouse tumor model experiments showed that VLP-OVAT could significantly inhibit tumor growth by increasing the proportions of CD4+ T cells, CD8+ T cells and effector memory T cells (TEM cells) and lowering the proportion of myeloid-derived suppressor cells (MDSCs) among tumor-infiltrating lymphocytes and splenocytes. Compared with other chemically synthesized nanomaterials, VLPs have obvious advantages as vaccine carriers due to their clear chemical composition, fixed spatial structure, excellent biocompatibility, and relatively high potential for clinical translation. Therefore, this platform may lay a solid foundation for the design and preparation of personalized therapeutic vaccines based on neoantigen peptides.
Collapse
Affiliation(s)
- Wenjing Li
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Zhe Jing
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Shuqing Wang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Qiyu Li
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Yutong Xing
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Haobo Shi
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Shuang Li
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, PR China.
| | - Zhangyong Hong
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, PR China.
| |
Collapse
|
34
|
Zhang Y, Gu P, Wusiman A, Xu S, Ni H, Qiu T, Liu Z, Hu Y, Liu J, Wang D. The Immunoenhancement Effects of Polyethylenimine-Modified Chinese Yam Polysaccharide-Encapsulated PLGA Nanoparticles as an Adjuvant. Int J Nanomedicine 2020; 15:5527-5543. [PMID: 32848386 PMCID: PMC7429225 DOI: 10.2147/ijn.s252515] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 07/15/2020] [Indexed: 01/13/2023] Open
Abstract
Background Poly(lactic-co-glycolic acid) (PLGA) has been extensively applied for sustained drug delivery and vaccine delivery system. However, vaccines delivered by PLGA nanoparticles alone could not effectively activate antigen-presenting cells (APCs) to induce strong immune responses. Purpose The aim of the present study was to design polyethylenimine (PEI)-modified Chinese yam polysaccharide (CYP)-encapsulated PLGA nanoparticles (CYPP-PEI) as a vaccine delivery system and evaluate the adjuvant activities in vitro and in vivo. Materials and Methods Cationic-modified nanoparticles exhibited high antigen absorption and could be efficiently taken by APCs to enhance the immune responses. Therefore, PEI-modified CYP-encapsulated PLGA nanoparticles (CYPP-PEI) were prepared. The storage stability and effective adsorption capacity for porcine circovirus-2 (PCV-2) antigen of these antigen-absorbed nanoparticles were measured for one month. Furthermore, the adjuvant activity of CYPP-PEI nanoparticles was evaluated on macrophages in vitro and through immune responses triggered by PCV-2 antigen in vivo. Results The PCV-2 absorbed CYPP-PEI nanoparticles showed excellent storage stability and high absorption efficiency of PCV-2 antigen. In vitro, CYPP-PEI nanoparticles promoted antigen uptake, enhanced surface molecular expressions of CD80 and CD86, and improved cytokine secretion of TNF-α, IFN-γ, and IL-12p70 in macrophages. After immunization with CYPP-PEI/PCV-2 formulation in mice, the expressions of surface activation markers on dendritic cells which located in draining lymph nodes were increased, such as MHCI, MHCII, and CD80. In addition, CYPP-PEI nanoparticles induced dramatically high PCV-2-specific IgG levels which could last for a long time and stimulated the secretion of subtype antibodies and cytokines. The results showed that CYPP-PEI could induce Th1/Th2 mixed but Th1-biased type immune responses. Conclusion Polyethylenimine-modified Chinese yam polysaccharide-encapsulated PLGA nanoparticle was a potential vaccine delivery system to trigger strong and persistent immune responses.
Collapse
Affiliation(s)
- Yue Zhang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing, People's Republic of China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Pengfei Gu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing, People's Republic of China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Adelijiang Wusiman
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing, People's Republic of China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Shuwen Xu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing, People's Republic of China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Haiyu Ni
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing, People's Republic of China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Tianxin Qiu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing, People's Republic of China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Zhenguang Liu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing, People's Republic of China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Yuanliang Hu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing, People's Republic of China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Jiaguo Liu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing, People's Republic of China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Deyun Wang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing, People's Republic of China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| |
Collapse
|
35
|
He J, Liu Z, Jiang W, Zhu T, Wusiman A, Gu P, Liu J, Wang D. Immune-adjuvant activity of lentinan-modified calcium carbonate microparticles on a H 5N 1 vaccine. Int J Biol Macromol 2020; 163:1384-1392. [PMID: 32758599 DOI: 10.1016/j.ijbiomac.2020.08.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 07/24/2020] [Accepted: 08/01/2020] [Indexed: 12/20/2022]
Abstract
In recent years, the high prevalence of avian influenza viruses especially H5N1 subtype isolated from poultry and human has become a major public health concern. Vaccination is still a major strategy for preventing H5N1 infections. Lentinan (LNT), a β-1,3-glucohexaose with β-1,6-branches, is extracted from Lentinus edodes and has been extensively studied for its immunoenhancement effects. In this study, we synthesized and characterized calcium carbonate (CaCO3) microparticles which modified with LNT as an adjuvant for H5N1 vaccine and investigated their ability to enhance immune responses. We prepared spherical and uniform CaCO3-LNT microparticles with a mean hydrodynamic size was around 2 μm. The H5N1 antigen-loaded CaCO3-LNT particles were injected into mice to evaluate their effectiveness as an adjuvant for H5N1 vaccines. The results demonstrated that CaCO3-LNT/H5N1 significantly enhanced the expression of MHC-II and CD86 in lymph node dendritic cells, and increased the ratio of CD4+ to CD8+ T cells in lymphocytes. Moreover, CaCO3-LNT/H5N1 surprisingly increased the HI titers and induced the secretion of IgG subtypes (IgG1 and IgG2b) and Th-associated cytokines (TNF-α, IFN-γ and IL-4) in immunized mice. Therefore, by combining with the immunostimulatory activity of LNT and the drug/antigen delivery capabilities of CaCO3, the CaCO3-LNT/H5N1 could induce a stronger cellular and humoral immune response and could be a potential adjuvant for the H5N1 vaccine.
Collapse
Affiliation(s)
- Jin He
- Institution of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 21005, PR China
| | - Zhenguang Liu
- Institution of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 21005, PR China
| | - Wenming Jiang
- China Animal Health and Epidemiology Center, Qingdao, PR China
| | - Tianyu Zhu
- Institution of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 21005, PR China
| | - Adelijiang Wusiman
- Institution of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 21005, PR China
| | - Pengfei Gu
- Institution of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 21005, PR China
| | - Jiaguo Liu
- Institution of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 21005, PR China
| | - Deyun Wang
- Institution of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 21005, PR China.
| |
Collapse
|
36
|
Liu Z, Yu L, Gu P, Bo R, Wusiman A, Liu J, Hu Y, Wang D. Preparation of lentinan-calcium carbonate microspheres and their application as vaccine adjuvants. Carbohydr Polym 2020; 245:116520. [PMID: 32718625 DOI: 10.1016/j.carbpol.2020.116520] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/22/2020] [Accepted: 05/25/2020] [Indexed: 11/19/2022]
Abstract
Adjuvants improve vaccine potency by enhancing immunogenicity and sustaining long-term immune responses. Lentinan (LNT), a β-1,3-glucohexaose with β-1,6-branches, is extracted from the mushroom Lentinus edodes and functions as an effective immunostimulatory drug. Previous studies have demonstrated the adjuvant activity of calcium carbonate (CaCO3) microspheres as well as their use as antigen delivery systems. In this study, we successfully loaded CaCO3 microspheres with LNT and evaluated their physicochemical characteristics prior to the adsorption of ovalbumin. Our experimental results demonstrated that LNT-CaCO3 significantly enhanced lymphocyte proliferation, and boosted the frequency of CD69 + B cells and the ratio of CD4+ to CD8 + T cells in spleen lymphocytes. Moreover, LNT-CaCO3 unexpectedly induced the secretion of IgG and Th-associated cytokines (IL-2, IL-4, IFN-γ, and TNF-α) in immunized mice. Therefore, LNT-CaCO3 microspheres induce robust cellular and humoral immune responses and have potential utility as vaccine delivery systems.
Collapse
Affiliation(s)
- Zhenguang Liu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Lin Yu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Pengfei Gu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Ruonan Bo
- School of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, PR China
| | - Adelijiang Wusiman
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Jiaguo Liu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Yuanliang Hu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China; 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, Nanjing Agricultural University, Nanjing, 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China.
| |
Collapse
|
37
|
Hong X, Zhong X, Du G, Hou Y, Zhang Y, Zhang Z, Gong T, Zhang L, Sun X. The pore size of mesoporous silica nanoparticles regulates their antigen delivery efficiency. SCIENCE ADVANCES 2020; 6:eaaz4462. [PMID: 32596445 PMCID: PMC7304990 DOI: 10.1126/sciadv.aaz4462] [Citation(s) in RCA: 157] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 05/08/2020] [Indexed: 05/19/2023]
Abstract
Subunit vaccines generally proceed through a 4-step in vivo cascade-the DUMP cascade-to generate potent cell-mediated immune responses: (1) drainage to lymph nodes; (2) uptake by dendritic cells (DCs); (3) maturation of DCs; and (4) Presentation of peptide-MHC I complexes to CD8+ T cells. How the physical properties of vaccine carriers such as mesoporous silica nanoparticles (MSNs) influence this cascade is unclear. We fabricated 80-nm MSNs with different pore sizes (7.8 nm, 10.3 nm, and 12.9 nm) and loaded them with ovalbumin antigen. Results demonstrated these MSNs with different pore sizes were equally effective in the first three steps of the DUMP cascade, but those with larger pores showed higher cross-presentation efficiency (step 4). Consistently, large-pore MSNs loaded with B16F10 tumor antigens yielded the strongest antitumor effects. These results demonstrate the promise of our lymph node-targeting large-pore MSNs as vaccine-delivery vehicles for immune activation and cancer vaccination.
Collapse
Affiliation(s)
- Xiaoyu Hong
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xiaofang Zhong
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Guangsheng Du
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yingying Hou
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yunting Zhang
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Tao Gong
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Ling Zhang
- College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Xun Sun
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
38
|
Dong Y, Gao J, Pei M, Wang X, Zhang C, Du Y, Jiang Y. Antigen-Conjugated Silica Solid Sphere as Nanovaccine for Cancer Immunotherapy. Int J Nanomedicine 2020; 15:2685-2697. [PMID: 32368049 PMCID: PMC7184137 DOI: 10.2147/ijn.s242463] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 04/08/2020] [Indexed: 12/19/2022] Open
Abstract
Background Nanocarriers could deliver significantly higher amounts of antigen to antigen-presenting cells (APCs), which have great potential to stimulate humoral and cellular response in cancer immunotherapy. Thereafter, silica solid nanosphere (SiO2) was prepared, and a model antigen (ovalbumin, OVA) was covalently conjugated on the surface of SiO2 to form nanovaccine (OVA@SiO2). And the application of OVA@SiO2 for cancer immunotherapy was evaluated. Materials and Methods SiO2 solid nanosphere was prepared by the Stöber method, then successively aminated by aminopropyltriethoxysilane and activated with glutaraldehyde. OVA was covalently conjugated on the surface of activated SiO2 to obtain nanovaccine (OVA@SiO2). Dynamic light scattering, scanning electron microscope, and transmission electron microscope were conducted to identify the size distribution, zeta potential and morphology of OVA@SiO2. The OVA loading capacity was investigated by varying glutaraldehyde concentration. The biocompatibility of OVA@SiO2 to DC2.4 and RAW246.7 cells was evaluated by a Cell Counting Kit-8 assay. The uptake of OVA@SiO2 by DC2.4 and its internalization pathway were evaluated in the absence or presence of different inhibitors. The activation and maturation of bone marrow-derived DC cells by OVA@SiO2 were also investigated. Finally, the in vivo transport of OVA@SiO2 and its toxicity to organs were appraised. Results All results indicated the successful covalent conjugation of OVA on the surface of SiO2. The as-prepared OVA@SiO2 possessed high antigen loading capacity, which had good biocompatibility to APCs and major organs. Besides, OVA@SiO2 facilitated antigen uptake by DC2.4 cells and its cytosolic release. Noteworthily, OVA@SiO2 significantly promoted the maturation of dendritic cells and up-regulation of cytokine secretion by co-administration of adjuvant CpG-ODN. Conclusion The as-prepared SiO2 shows promising potential for use as an antigen delivery carrier.
Collapse
Affiliation(s)
- Ying Dong
- School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin 300130, People's Republic of China
| | - Jing Gao
- School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin 300130, People's Republic of China
| | - Mengyue Pei
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, People's Republic of China
| | - Xiaoli Wang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, People's Republic of China
| | - Chuangnian Zhang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, People's Republic of China
| | - Yingjie Du
- School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin 300130, People's Republic of China
| | - Yanjun Jiang
- School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin 300130, People's Republic of China
| |
Collapse
|
39
|
Wu J, Ma G. Biomimic strategies for modulating the interaction between particle adjuvants and antigen-presenting cells. Biomater Sci 2020; 8:2366-2375. [DOI: 10.1039/c9bm02098e] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The design strategies of particle adjuvants by mimicking natural pathogens to strengthen their interaction with antigen-presenting cells.
Collapse
Affiliation(s)
- Jie Wu
- State Key Laboratory of Biochemical Engineering
- Institute of Process Engineering
- Chinese Academy of Sciences
- Beijing 100190
- P.R. China
| | - Guanghui Ma
- State Key Laboratory of Biochemical Engineering
- Institute of Process Engineering
- Chinese Academy of Sciences
- Beijing 100190
- P.R. China
| |
Collapse
|
40
|
Ji H, Wu G, Li Y, Wang K, Xue X, You S, Wu S, Ren T, He B, Shi X. Self-Albumin Camouflage of Carrier Protein Prevents Nontarget Antibody Production for Enhanced LDL-C Immunotherapy. Adv Healthc Mater 2020; 9:e1901203. [PMID: 31814301 DOI: 10.1002/adhm.201901203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 11/07/2019] [Indexed: 11/05/2022]
Abstract
Elevated low-density lipoprotein cholesterol (LDL-C) increases the risk of atherosclerotic cardiovascular disease. Peptide-based PCSK9 vaccines have shown a promising prospect of reducing LDL-C. In peptide vaccine (pVax) design, the peptide antigens need to conjugate with carrier protein (CP). However, CP incorporation can induce undesirable anti-CP antibodies, which sterically mask peptide epitopes from being recognized by specific B cells and impair subsequent therapeutically antibody production. This epitopic suppression has posed a barrier in clinical translation of conjugate vaccines all along. A model CP (keyhole limpet hemocyanin, KLH) is herein camouflaged with serum albumin (SA) into hybrid nanocarriers (SA@N), with PCSK9 peptide being anchored onto the surface to form nanovaccine (SA@NVax). Such camouflage of KLH via high "self" SA coverage is able to inhibit KLH from extracellular immune recognition and prevent detectable anti-KLH antibody production. Furthermore, the nanovaccine around 70 nm stabilized by intermolecular disulfide network is ideal for internalization and biodegradation by antigen presenting cells as well as better retention in draining lymph nodes and spleen. As expected, the SA@NVax efficiently primes higher anti-PCSK9 IgG antibody titer than PCSK9 pVax.
Collapse
Affiliation(s)
- Haiying Ji
- Department of Anesthesiology and SICUXinhua HospitalSchool of MedicineShanghai Jiao Tong University Shanghai 200092 China
| | - Guangxi Wu
- Department of Anesthesiology and SICUXinhua HospitalSchool of MedicineShanghai Jiao Tong University Shanghai 200092 China
| | - Yongyong Li
- Institute for Biomedical Engineering and Nano ScienceShanghai East HospitalTongji University School of Medicine Shanghai 200092 China
| | - Kun Wang
- Institute for Biomedical Engineering and Nano ScienceShanghai East HospitalTongji University School of Medicine Shanghai 200092 China
| | - Xiaomei Xue
- Department of Anesthesiology and SICUXinhua HospitalSchool of MedicineShanghai Jiao Tong University Shanghai 200092 China
| | - Shasha You
- Department of Anesthesiology and SICUXinhua HospitalSchool of MedicineShanghai Jiao Tong University Shanghai 200092 China
| | - Shengming Wu
- Institute for Biomedical Engineering and Nano ScienceShanghai East HospitalTongji University School of Medicine Shanghai 200092 China
| | - Tianbin Ren
- School of Materials Science and EngineeringTongji University Shanghai 200092 China
| | - Bin He
- Department of Anesthesiology and SICUXinhua HospitalSchool of MedicineShanghai Jiao Tong University Shanghai 200092 China
| | - Xueyin Shi
- Department of Anesthesiology and SICUXinhua HospitalSchool of MedicineShanghai Jiao Tong University Shanghai 200092 China
| |
Collapse
|
41
|
Gu P, Wusiman A, Zhang Y, Liu Z, Bo R, Hu Y, Liu J, Wang D. Rational Design of PLGA Nanoparticle Vaccine Delivery Systems To Improve Immune Responses. Mol Pharm 2019; 16:5000-5012. [DOI: 10.1021/acs.molpharmaceut.9b00860] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
42
|
Vladisavljević GT. Preparation of microemulsions and nanoemulsions by membrane emulsification. Colloids Surf A Physicochem Eng Asp 2019. [DOI: 10.1016/j.colsurfa.2019.123709] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
43
|
Song C, Li F, Wang S, Wang J, Wei W, Ma G. Recent Advances in Particulate Adjuvants for Cancer Vaccination. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201900115] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Cui Song
- State Key Laboratory of Biochemical EngineeringInstitute of Process EngineeringChinese Academy of Sciences Beijing 100190 P. R. China
- University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Feng Li
- State Key Laboratory of Biochemical EngineeringInstitute of Process EngineeringChinese Academy of Sciences Beijing 100190 P. R. China
- University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Shuang Wang
- State Key Laboratory of Biochemical EngineeringInstitute of Process EngineeringChinese Academy of Sciences Beijing 100190 P. R. China
| | - Jianghua Wang
- State Key Laboratory of Biochemical EngineeringInstitute of Process EngineeringChinese Academy of Sciences Beijing 100190 P. R. China
- University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Wei Wei
- State Key Laboratory of Biochemical EngineeringInstitute of Process EngineeringChinese Academy of Sciences Beijing 100190 P. R. China
- University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Guanghui Ma
- State Key Laboratory of Biochemical EngineeringInstitute of Process EngineeringChinese Academy of Sciences Beijing 100190 P. R. China
- University of Chinese Academy of Sciences Beijing 100049 P. R. China
| |
Collapse
|
44
|
Li E, Chi H, Huang P, Yan F, Zhang Y, Liu C, Wang Z, Li G, Zhang S, Mo R, Jin H, Wang H, Feng N, Wang J, Bi Y, Wang T, Sun W, Gao Y, Zhao Y, Yang S, Xia X. A Novel Bacterium-Like Particle Vaccine Displaying the MERS-CoV Receptor-Binding Domain Induces Specific Mucosal and Systemic Immune Responses in Mice. Viruses 2019; 11:E799. [PMID: 31470645 PMCID: PMC6784119 DOI: 10.3390/v11090799] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/26/2019] [Accepted: 08/27/2019] [Indexed: 02/06/2023] Open
Abstract
Middle East respiratory syndrome coronavirus (MERS-CoV), a new coronavirus that has been causing severe and fatal acute respiratory illnesses in humans since its outbreak in 2012, has raised public fear worldwide. The development of prophylactics and therapeutics is urgently needed to prevent and control MERS-CoV infections. In this study, a bacterium (Lactococcus lactis)-like particle (BLP) vaccine displaying the MERS-CoV receptor-binding domain (RBD) was developed, and gram-positive enhancer matrix (GEM) particles were used as substrates to externally bind to the MERS-CoV RBD through a protein anchor (PA). The designs included different numbers of lysin motif (LysM) repeats in the PAs linked by linkers (RBD-linker-PA2 (RLP2), RBD-linker-PA3 (RLP3) and RBD-PA3 (RP3)), and three LysM repeats and a linker in the fusion proteins increased the binding activity to the RBD. The specific immune responses were tested by intranasally immunizing mice with RLP3-GEM with or without the adjuvant GEL01. The results showed that GEL01-adjuvanted RLP3-GEM increased the systemic humoral, cellular and local mucosal immune responses in the mouse model, especially in the intestinal tract. The above results indicate that the MERS-CoV BLP product has the potential to be developed into a promising mucosal candidate vaccine to protect against MERS-CoV infections.
Collapse
Affiliation(s)
- Entao Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130000, China
| | - Hang Chi
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130000, China.
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun 130000, China.
| | - Pei Huang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130000, China
- Animal Science and Technology College, Jilin Agricultural University, Changchun 130118, China
| | - Feihu Yan
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130000, China
| | - Ying Zhang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130000, China
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, China
| | - Chuanyu Liu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130000, China
- Animal Science and Technology College, Jilin Agricultural University, Changchun 130118, China
| | - Zhenshan Wang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130000, China
- Animal Science and Technology College, Jilin Agricultural University, Changchun 130118, China
| | - Guohua Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130000, China
- College of Animal Science and Technology, Shihezi University, Shihezi 832003, China
| | - Shengnan Zhang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130000, China
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, China
| | - Ruo Mo
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130000, China
- Animal Science and Technology College, Jilin Agricultural University, Changchun 130118, China
| | - Hongli Jin
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130000, China
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Hualei Wang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130000, China
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Na Feng
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130000, China
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun 130000, China
| | - Jianzhong Wang
- Animal Science and Technology College, Jilin Agricultural University, Changchun 130118, China
| | - Yuhai Bi
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Tiecheng Wang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130000, China
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun 130000, China
| | - Weiyang Sun
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130000, China
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun 130000, China
| | - Yuwei Gao
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130000, China
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun 130000, China
| | - Yongkun Zhao
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130000, China.
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun 130000, China.
| | - Songtao Yang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130000, China.
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun 130000, China.
| | - Xianzhu Xia
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130000, China.
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun 130000, China.
| |
Collapse
|
45
|
Polyethylenimine-coated PLGA nanoparticles-encapsulated Angelica sinensis polysaccharide as an adjuvant to enhance immune responses. Carbohydr Polym 2019; 223:115128. [PMID: 31427012 DOI: 10.1016/j.carbpol.2019.115128] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 06/24/2019] [Accepted: 07/24/2019] [Indexed: 01/28/2023]
Abstract
Nanoparticle delivery systems have been widely investigated as new vaccines strategy to enhance the immune responses to antigens against infectious diseases. The positively charged nanoparticles could efficiently improve the immune responses due to targeting and activating the antigen-presenting cells. In this study, the immunopotentiator Angelica sinensis polysaccharide (ASP) was encapsulated into Poly (lactic-co-glycolic acid) (PLGA) nanoparticles, and the polyethylenimine, one of the cationic polymers, was used to coat nanoparticles to develop a new nanoparticle delivery system (ASP-PLGA-PEI) with positively charged. The ASP-PLGA-PEI nanoparticles significantly activated macrophages, and promoted the expression of the MHCII and CD86 and the production of IL-1β and IL-12p70 cytokines of macrophages. Furthermore, the antigen adsorbed on the surface of the ASP-PLGA-PEI nanoparticles enhanced the antigen uptake by macrophages. Moreover, the mice immunized with PCV2 antigen adsorbed ASP-PLGA-PEI nanoparticles significantly enhanced PCV2-specific IgG immune response and the levels of cytokines, induced a mixed Th1/Th2 immune response with Th1 bias compared with other groups. These findings demonstrate that the positively charged nanoparticles (ASP-PLGA-PEI) have the potential to serve as an effective vaccine delivery and adjuvant system to induce vigorous and long-term immune responses.
Collapse
|
46
|
Zhang L, Wu S, Qin Y, Fan F, Zhang Z, Huang C, Ji W, Lu L, Wang C, Sun H, Leng X, Kong D, Zhu D. Targeted Codelivery of an Antigen and Dual Agonists by Hybrid Nanoparticles for Enhanced Cancer Immunotherapy. NANO LETTERS 2019; 19:4237-4249. [PMID: 30868883 DOI: 10.1021/acs.nanolett.9b00030] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Among approaches of current cancer immunotherapy, a dendritic cell (DC)-targeted vaccine based on nanotechnology could be a promising way to efficiently induce potent immune responses. To enhance DC targeting and vaccine efficiency, we included imiquimod (IMQ), a toll-like receptor 7/8 (TLR 7/8) agonist, and monophosphoryl lipid A (MPLA), a TLR4 agonist, to synthesize lipid-polymer hybrid nanoparticles using PCL-PEG-PCL and DOTAP (IMNPs) as well as DSPE-PEG-mannose (MAN-IMNPS). The spatiotemporal delivery of MPLA (within the outer lipid layer) to extracellular TLR4 and IMQ (in the hydrophobic core of NPs) to intracellular TLR7/8 can activate DCs synergistically to improve vaccine efficacy. Ovalbumin (OVA) as a model antigen was readily absorbed by positively charged DOTAP and showed a quick release in vitro. Our results demonstrated that this novel nanovaccine enhanced cellular uptake, cytokine production, and maturation of DCs. Compared with the quick metabolism of free OVA-agonists, the depot effect of OVA-IMNPs was observed, whereas MAN-OVA-IMNPs promoted trafficking to secondary lymphoid organs. After immunization with a subcutaneous injection, the nanovaccine, especially MAN-OVA-IMNPs, induced more antigen-specific CD8+ T cells, greater lymphocyte activation, stronger cross-presentation, and more generation of memory T cells, antibody, IFN-γ, and granzyme B. Prophylactic vaccination of MAN-OVA-IMNPs significantly delayed tumor development and prolonged the survival in mice. The therapeutic tumor challenge indicated that MAN-OVA-IMNPs prohibited tumor progression more efficiently than other formulations, and the combination with an immune checkpoint blockade further enhanced antitumor effects. Hence, the DC-targeted vaccine codelivery with IMQ and MPLA adjuvants by hybrid cationic nanoparticles in a spatiotemporal manner is a promising multifunctional antigen delivery system in cancer immunotherapy.
Collapse
Affiliation(s)
- Linhua Zhang
- Tianjin Key Laboratory of Biomedical Materials , Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300192 , China
| | - Shengjie Wu
- Tianjin Key Laboratory of Biomedical Materials , Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300192 , China
| | - Yu Qin
- Tianjin Key Laboratory of Biomedical Materials , Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300192 , China
| | - Fan Fan
- Tianjin Key Laboratory of Biomedical Materials , Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300192 , China
| | - Zhiming Zhang
- Tianjin Key Laboratory of Biomedical Materials , Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300192 , China
| | - Chenlu Huang
- Tianjin Key Laboratory of Biomedical Materials , Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300192 , China
| | - Weihang Ji
- Department of Biomedical Engineering , University of Minnesota , 7-116 Hasselmo Hall, 312 Church Street SE , Minneapolis , Minnesota 55455 , United States
| | - Lu Lu
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College , Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine , Tianjin 300192 , China
| | - Chun Wang
- Department of Biomedical Engineering , University of Minnesota , 7-116 Hasselmo Hall, 312 Church Street SE , Minneapolis , Minnesota 55455 , United States
| | - Hongfan Sun
- Tianjin Key Laboratory of Biomedical Materials , Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300192 , China
| | - Xigang Leng
- Tianjin Key Laboratory of Biomedical Materials , Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300192 , China
| | - Deling Kong
- The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences , Nankai University , Tianjin 300071 , China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute , Xuzhou Medical University , Xuzhou 221004 , Jiangsu , China
| | - Dunwan Zhu
- Tianjin Key Laboratory of Biomedical Materials , Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300192 , China
| |
Collapse
|
47
|
Hou C, Yi B, Jiang J, Chang YF, Yao X. Up-to-date vaccine delivery systems: robust immunity elicited by multifarious nanomaterials upon administration through diverse routes. Biomater Sci 2019; 7:822-835. [PMID: 30540292 DOI: 10.1039/c8bm01197d] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
In this review, we summarize the recent design strategies (2015-present) of nanomaterial-based vaccine delivery systems via multiple routes to induce robust protective immunity. The selected topics are focused on the novel design strategies of nanomaterial carriers for vaccine delivery. Inspired by recent advances, we also briefly introduce the emerging administration routes that may give rise to synergistic immune effects with advanced delivery systems. Ultimately, we present the existing challenges and survey the prospective development of various nanoparticle vaccine delivery systems.
Collapse
Affiliation(s)
- Changshun Hou
- Department of Biomedical Sciences, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P. R. China.
| | | | | | | | | |
Collapse
|
48
|
Wang N, Chen M, Wang T. Liposomes used as a vaccine adjuvant-delivery system: From basics to clinical immunization. J Control Release 2019; 303:130-150. [PMID: 31022431 PMCID: PMC7111479 DOI: 10.1016/j.jconrel.2019.04.025] [Citation(s) in RCA: 183] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 04/17/2019] [Accepted: 04/17/2019] [Indexed: 12/14/2022]
Abstract
Liposomes are widely utilized as a carrier to improve therapeutic efficacy of agents thanks to their merits of high loading capacity, targeting delivery, reliable protection of agents, good biocompatibility, versatile structure modification and adjustable characteristics, such as size, surface charge, membrane flexibility and the agent loading mode. In particular, in recent years, through modification with immunopotentiators and targeting molecules, and in combination with innovative immunization devices, liposomes are rapidly developed as a multifunctional vaccine adjuvant-delivery system (VADS) that has a high capability in inducing desired immunoresponses, as they can target immune cells and even cellular organelles, engender lysosome escape, and promote Ag cross-presentation, thus enormously enhancing vaccination efficacy. Moreover, after decades of development, several products developed on liposome VADS have already been authorized for clinical immunization and are showing great advantages over conventional vaccines. This article describes in depth some critical issues relevant to the development of liposomes as a VADS, including principles underlying immunization, physicochemical properties of liposomes as the immunity-influencing factors, functional material modification to enhance immunostimulatory functions, the state-of-the-art liposome VADSs, as well as the marketed vaccines based on a liposome VADS. Therefore, this article provides a comprehensive reference to the development of novel liposome vaccines.
Collapse
Affiliation(s)
- Ning Wang
- School of Food and Bioengineering, Hefei University of Technology, 193 Tun Brook Road, Hefei, Anhui Province 230009, China
| | - Minnan Chen
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China
| | - Ting Wang
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China.
| |
Collapse
|
49
|
Zhu D, Hu C, Fan F, Qin Y, Huang C, Zhang Z, Lu L, Wang H, Sun H, Leng X, Wang C, Kong D, Zhang L. Co-delivery of antigen and dual agonists by programmed mannose-targeted cationic lipid-hybrid polymersomes for enhanced vaccination. Biomaterials 2019; 206:25-40. [DOI: 10.1016/j.biomaterials.2019.03.012] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 03/01/2019] [Accepted: 03/11/2019] [Indexed: 02/06/2023]
|
50
|
Won JE, Byeon Y, Wi TI, Lee JM, Kang TH, Lee JW, Shin BC, Han HD, Park YM. Enhanced Antitumor Immunity Using a Tumor Cell Lysate-Encapsulated CO2-Generating Liposomal Carrier System and Photothermal Irradiation. ACS APPLIED BIO MATERIALS 2019; 2:2481-2489. [DOI: 10.1021/acsabm.9b00183] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Ji Eun Won
- Department of Immunology, School of Medicine, Konkuk University, 268 Chungwondaero, Chungju-Si, Chungcheongbuk-Do 380-701, South Korea
| | - Yeongseon Byeon
- Department of Immunology, School of Medicine, Konkuk University, 268 Chungwondaero, Chungju-Si, Chungcheongbuk-Do 380-701, South Korea
| | - Tae In Wi
- Department of Immunology, School of Medicine, Konkuk University, 268 Chungwondaero, Chungju-Si, Chungcheongbuk-Do 380-701, South Korea
| | - Jae Myeong Lee
- Department of Immunology, School of Medicine, Konkuk University, 268 Chungwondaero, Chungju-Si, Chungcheongbuk-Do 380-701, South Korea
| | - Tae Heung Kang
- Department of Immunology, School of Medicine, Konkuk University, 268 Chungwondaero, Chungju-Si, Chungcheongbuk-Do 380-701, South Korea
| | - Jeong Won Lee
- Department of Obstertrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 135-710, South Korea
| | - Byung Cheol Shin
- Bio/Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon 305-600, South Korea
| | - Hee Dong Han
- Department of Immunology, School of Medicine, Konkuk University, 268 Chungwondaero, Chungju-Si, Chungcheongbuk-Do 380-701, South Korea
| | - Yeong-Min Park
- Department of Immunology, School of Medicine, Konkuk University, 268 Chungwondaero, Chungju-Si, Chungcheongbuk-Do 380-701, South Korea
| |
Collapse
|