1
|
Wang J, Zhao Z, Wang Q, Shi J, Wong DWC, Cheung JCW. Advancements in Nanoparticle-Based Adjuvants for Enhanced Tuberculosis Vaccination: A Review. Vaccines (Basel) 2024; 12:1335. [PMID: 39771997 PMCID: PMC11680411 DOI: 10.3390/vaccines12121335] [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: 10/29/2024] [Revised: 11/21/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025] Open
Abstract
Tuberculosis (TB) remains a leading cause of morbidity and mortality worldwide, necessitating the development of more effective vaccines. Nanoparticle-based adjuvants represent a promising approach to enhancing tuberculosis vaccine efficacy. This review focuses on the advantages of nanoparticulate-loaded vaccines, emphasizing their ability to improve antigen delivery, safety, and immunogenicity. We discuss the various types of nanoparticles and their unique physicochemical properties that contribute to improved antigen delivery and sustained immune activation. Additionally, we highlight the advantages of nanoparticle-based adjuvants in inducing strong cellular and humoral immunity, enhancing vaccine stability, and reducing adverse effects. Finally, we address current challenges and future perspectives in the application of these novel adjuvants, emphasizing their potential to transform TB vaccine strategies and ultimately contribute to better global health outcomes.
Collapse
Affiliation(s)
- Jiao Wang
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
- Department of Clinical Laboratory, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan 430073, China
| | - Zian Zhao
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Quan Wang
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Jingyu Shi
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Duo Wai-Chi Wong
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - James Chung-Wai Cheung
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| |
Collapse
|
2
|
Qin S, He G, Yang J. Nanomaterial combined engineered bacteria for intelligent tumor immunotherapy. J Mater Chem B 2024; 12:9795-9820. [PMID: 39225508 DOI: 10.1039/d4tb00741g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Cancer remains the leading cause of human death worldwide. Compared to traditional therapies, tumor immunotherapy has received a lot of attention and research focus due to its potential to activate both innate and adaptive immunity, low toxicity to normal tissue, and long-term immune activity. However, its clinical effectiveness and large-scale application are limited due to the immunosuppression microenvironment, lack of spatiotemporal control, expensive cost, and long manufacturing time. Recently, nanomaterial combined engineered bacteria have emerged as a promising solution to the challenges of tumor immunotherapy, which offers spatiotemporal control, reversal of immunosuppression, and scalable production. Therefore, we summarize the latest research on nanomaterial-assisted engineered bacteria for precise tumor immunotherapies, including the cross-talk of nanomaterials and bacteria as well as their application in different immunotherapies. In addition, we further discuss the advantages and challenges of nanomaterial-engineered bacteria and their future prospects, inspiring more novel and intelligent tumor immunotherapy.
Collapse
Affiliation(s)
- Shurong Qin
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210023, China
| | - Guanzhong He
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210023, China
| | - Jingjing Yang
- Department of Biochemistry and Molecular Biology, School of Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
3
|
Croitoru GA, Pîrvulescu DC, Niculescu AG, Epistatu D, Rădulescu M, Grumezescu AM, Nicolae CL. Nanomaterials in Immunology: Bridging Innovative Approaches in Immune Modulation, Diagnostics, and Therapy. J Funct Biomater 2024; 15:225. [PMID: 39194663 DOI: 10.3390/jfb15080225] [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: 07/02/2024] [Revised: 07/30/2024] [Accepted: 08/12/2024] [Indexed: 08/29/2024] Open
Abstract
The intersection of immunology and nanotechnology has provided significant advancements in biomedical research and clinical applications over the years. Immunology aims to understand the immune system's defense mechanisms against pathogens. Nanotechnology has demonstrated its potential to manipulate immune responses, as nanomaterials' properties can be modified for the desired application. Research has shown that nanomaterials can be applied in diagnostics, therapy, and vaccine development. In diagnostics, nanomaterials can be used for biosensor development, accurately detecting biomarkers even at very low concentrations. Therapeutically, nanomaterials can act as efficient carriers for delivering drugs, antigens, or genetic material directly to targeted cells or tissues. This targeted delivery improves therapeutic efficacy and reduces the adverse effects on healthy cells and tissues. In vaccine development, nanoparticles can improve vaccine durability and extend immune responses by effectively delivering adjuvants and antigens to immune cells. Despite these advancements, challenges regarding the safety, biocompatibility, and scalability of nanomaterials for clinical applications are still present. This review will cover the fundamental interactions between nanomaterials and the immune system, their potential applications in immunology, and their safety and biocompatibility concerns.
Collapse
Affiliation(s)
- George-Alexandru Croitoru
- Faculty of Dental Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Street, 050474 Bucharest, Romania
| | - Diana-Cristina Pîrvulescu
- Faculty of Chemical Engineering and Biotechnology, National University of Science and Technology Politehnica Bucharest, 011061 Bucharest, Romania
| | - Adelina-Gabriela Niculescu
- Faculty of Chemical Engineering and Biotechnology, National University of Science and Technology Politehnica Bucharest, 011061 Bucharest, Romania
- Research Institute of the University of Bucharest-ICUB, University of Bucharest, 050657 Bucharest, Romania
| | - Dragoș Epistatu
- Faculty of Dental Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Street, 050474 Bucharest, Romania
| | - Marius Rădulescu
- Faculty of Chemical Engineering and Biotechnology, National University of Science and Technology Politehnica Bucharest, 011061 Bucharest, Romania
| | - Alexandru Mihai Grumezescu
- Faculty of Chemical Engineering and Biotechnology, National University of Science and Technology Politehnica Bucharest, 011061 Bucharest, Romania
- Research Institute of the University of Bucharest-ICUB, University of Bucharest, 050657 Bucharest, Romania
| | - Carmen-Larisa Nicolae
- Faculty of Dental Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Street, 050474 Bucharest, Romania
| |
Collapse
|
4
|
Lv T, Meng Y, Liu Y, Han Y, Xin H, Peng X, Huang J. RNA nanotechnology: A new chapter in targeted therapy. Colloids Surf B Biointerfaces 2023; 230:113533. [PMID: 37713955 DOI: 10.1016/j.colsurfb.2023.113533] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/14/2023] [Accepted: 09/04/2023] [Indexed: 09/17/2023]
Abstract
Nanoparticles have been widely studied in the fields of biotechnology, pharmacy, optics and medicine and have broad application prospects. Numerous studies have shown significant interest in utilizing nanoparticles for chemically coating or coupling drugs, aiming to address the challenges of drug delivery, including degradability and uncertainty. Furthermore, the utilization of lipid nanoparticles loaded with novel coronavirus antigen mRNA to control the COVID-19 pandemic has led to a notable surge in research on nanoparticle vaccines. Hence, nanoparticles have emerged as a crucial delivery system for disease prevention and treatment, bearing immense significance. Current research highlights that nanoparticles offer superior efficacy and potential compared to conventional drug treatment and prevention methods. Notably, for drug delivery applications, it is imperative to utilize biodegradable nanoparticles. This paper reviews the structures and characteristics of various biodegradable nanoparticles and their applications in biomedicine in order to inspire more researchers to further explore the functions of nanoparticles. RNA plays a pivotal role in regulating the occurrence and progression of diseases, but its inherent susceptibility to degradation poses a challenge. In light of this, we conducted a comprehensive review of the research advancements concerning RNA-containing biodegradable nanoparticles in the realm of disease prevention and treatment, focusing on cancer, inflammatory diseases, and viral infections.
Collapse
Affiliation(s)
- Tongtong Lv
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei, China; Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Yingying Meng
- Department of Gastroenterology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yifan Liu
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei, China; Department of Oncology, Jingzhou Hospital Affifiliated to Yangtze University, Jingzhou, Hubei, China
| | - Yukun Han
- Department of Medical Imaging, School of Medicine, and Positron Emission Computed Tomography (PET) Center of the First Affifiliated Hospital, Yangtze University, Jingzhou, Hubei, China
| | - Hongwu Xin
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Xiaochun Peng
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei, China; Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei, China.
| | - Jinbai Huang
- Department of Medical Imaging, School of Medicine, and Positron Emission Computed Tomography (PET) Center of the First Affifiliated Hospital, Yangtze University, Jingzhou, Hubei, China.
| |
Collapse
|
5
|
Xekouki K, Lagopati N, Demetzos C, Gazouli M, Pippa N. A mini review for lipid-based nanovaccines: from their design to their applications. J Liposome Res 2023; 33:214-233. [PMID: 36856671 DOI: 10.1080/08982104.2023.2170408] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/05/2022] [Accepted: 11/04/2022] [Indexed: 03/02/2023]
Abstract
Nanovaccines have shown to be effective, and this is the reason they are preferred than conventional vaccines. The scope of this review is to describe the role, mechanisms, and advantages of nano vaccines based on lipids, and present the most important types, their physicochemical characteristics, as well as their challenges. The most important categories of lipid nano-vaccines are liposomal nano vaccines and (virus-lipid nanoparticles (NPs)/virosomes. Examples of vaccine formulations from each category are presented and analyzed below, focusing on their structure and physicochemical characteristics. In all cases, a nanoscale platform is used, enriched with adjuvants, antigens, and other helping agents to trigger immune response process and achieve cell targeting, and eventually immunity against the desired disease. The exact mechanism of action of each vaccine is not always completely known or understood. Physicochemical characteristics, such as particle size, morphology/shape, and zeta potential are also mentioned as they seem to affect the properties and mechanism of action of the vaccine formulation.
Collapse
Affiliation(s)
- Katerina Xekouki
- Department of Pharmaceutical Technology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Nefeli Lagopati
- Laboratory of Biology, Department of Basic Medical Science, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Costas Demetzos
- Department of Pharmaceutical Technology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Gazouli
- Laboratory of Biology, Department of Basic Medical Science, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Natassa Pippa
- Department of Pharmaceutical Technology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
6
|
Jyoti K, Malik G, Chaudhary M, Madan J, Kamboj A. Hyaluronate decorated polyethylene glycol linked poly(lactide-co-glycolide) nanoparticles encapsulating MUC-1 peptide augmented mucosal immune response in Balb/c mice through inhalation route. Biochim Biophys Acta Gen Subj 2023; 1867:130317. [PMID: 36731729 DOI: 10.1016/j.bbagen.2023.130317] [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: 12/04/2022] [Revised: 01/16/2023] [Accepted: 01/27/2023] [Indexed: 02/01/2023]
Abstract
BACKGROUND AND OBJECTIVES NSCLC (Non-Small Cell Lung Cancer) clutches highest mortality rate in man and women globally. The present study was conducted to target MUC-1 peptide (M-1) into antigen presenting cells by cargo the peptide into hyaluronic acid decorated polyethylene glycol linked poly (D, l-lactide-co-glycolide) nanoparticles (M-1-PL-co-GA-PEG-sHA-NPs) for generating mucosal immunity through inhalation (i.h.) route. METHODOLOGY AND RESULTS The mean particle size and surface charge of M-1-PL-co-GA-PEG-sHA-NPs was measured to be 136.2 ± 18.38-nm and - 28.34 ± 6.77-mV, respectively, prepared by non-aggregated emulsion-diffusion evaporation method. The 28.42% percentage release of M-1 peptide from M-1-PL-co-GA-PEG-NPs was observed to be at 2 h and 95.29% at 8 h while the percentage release of M-1 peptide from M-1-PL-co-GA-PEG-sHA-NPs was observed to be 26.02% at 4 h and 97.95% at 24 h that proved the prolonged release of antigen. M-1-PL-co-GA-PEG-sHA-NPs demonstrated higher (P < 0.05) cellular uptake of 86.2% in RAW 264.7 cells in comparison to 27.6% of M-1-PL-co-GA-PEG-NPs. In addition, M-1-PL-co-GA-PEG-sHA-NPs induced remarkably (P < 0.05) elevated release of 80.6-pg/ml of TNF-α in comparison to 5-pg/ml by culture medium and 57.9-pg/ml of TNF-α by M-1-PL-co-GA-PEG-NPs. Similarly, M-1-PL-co-GA-PEG-sHA-NPs persuade remarkably (P < 0.05) elevated release of 225-pg/ml of IL-1β in comparison to 47-pg/ml by culture medium and 161.9-pg/ml of IL-1β by M-1-PL-co-GA-PEG-NPs. M-1-PL-co-GA-PEG-sHA-NPs might have been endocytosed through receptor mediated pathway owing to presence of sHA. Mice immunized through i.h. route with M-1-PL-co-GA-PEG-sHA-NPs induced strong (P < 0.05) IgA antibody titre as compared to M-1-PL-co-GA-PEG-NPs and M-1 peptide in dose-dosage regimen. CONCLUSION M-1-PL-co-GA-PEG-sHA-NPs nanovaccine warrants further analysis in xenograft model of NSCLC to showcase its antitumor capability.
Collapse
Affiliation(s)
- Kiran Jyoti
- IKG Punjab Technical University, Jalandhar, Punjab, India; Department of Pharmaceutics, Chandigarh College of Pharmacy, Mohali, Punjab, India.
| | - Garima Malik
- MM College of Pharmacy, Maharishi Markandeshwar University, Ambala, Haryana, India
| | | | - Jitender Madan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Anjoo Kamboj
- Department of Pharmaceutics, Chandigarh College of Pharmacy, Mohali, Punjab, India
| |
Collapse
|
7
|
Wang Y, Liu L, Zheng X, Liu X. Membrane-camouflaged biomimetic nanoparticles as potential immunomodulatory solutions for sepsis: An overview. Front Bioeng Biotechnol 2023; 11:1111963. [PMID: 36970623 PMCID: PMC10036601 DOI: 10.3389/fbioe.2023.1111963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 02/27/2023] [Indexed: 03/12/2023] Open
Abstract
Sepsis is a life-threatening organ dysfunction due to dysregulated host responses induced by infection. The presence of immune disturbance is key to the onset and development of sepsis but has remarkably limited therapeutic options. Advances in biomedical nanotechnology have provided innovative approaches to rebalancing the host immunity. In particular, the technique of membrane-coating has demonstrated remarkable improvements to therapeutic nanoparticles (NPs) in terms of tolerance and stability while also improving their biomimetic performance for immunomodulatory purposes. This development has led to the emergence of using cell-membrane-based biomimetic NPs in treating sepsis-associated immunologic derangements. In this minireview, we present an overview of the recent advances in membrane-camouflaged biomimetic NPs, highlighting their multifaceted immunomodulatory effects in sepsis such as anti-infection, vaccination, inflammation control, reversing of immunosuppression, and targeted delivery of immunomodulatory agents.
Collapse
Affiliation(s)
- Yanbei Wang
- School of Culture and Tourism, Chongqing City Management College, Chongqing, China
| | - Liping Liu
- School of Culture and Tourism, Chongqing City Management College, Chongqing, China
| | - Xinchuan Zheng
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, China
- *Correspondence: Xinchuan Zheng, ; Xin Liu,
| | - Xin Liu
- Medical Research Center, Southwest Hospital, Army Military Medical University, Chongqing, China
- *Correspondence: Xinchuan Zheng, ; Xin Liu,
| |
Collapse
|
8
|
Xu S, Wu Z, Cai G, Zhang Y, Peng S, Jiao L, Liu Z, Yang Y, Wang D. Astragalus polysaccharides combined with simvastatin as an immunostimulant enhances the immune adjuvanticity of oil-in-water emulsion and immune responses in mice. Vaccine 2023; 41:1684-1693. [PMID: 36754767 DOI: 10.1016/j.vaccine.2023.01.069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 12/12/2022] [Accepted: 01/30/2023] [Indexed: 02/08/2023]
Abstract
Oil-in-water emulsion-based adjuvants have demonstrated acceptable safety in many disease indications, while their adjuvant activities for vaccines still need to be improved. Recently, the strategy of combining adjuvants with multiple types of immunostimulants has been shown to enhance immune responses. In this study, astragalus polysaccharides were combined with simvastatin as an immunostimulant to construct a compound O/W emulsion adjuvant. The formulations were optimized according to the OVA-specific antibody responses induced in mice. For this reason, high (5 mg/mL), medium (2.5 mg/mL), and low (1.25 mg/mL) concentrations of astragalus polysaccharides and high (10 mg/mL), medium (1 mg/mL), and low (0.1 mg/mL) concentrations of simvastatin were selected. The final optimal formulation of the immunostimulant was a high concentration of astragalus polysaccharides combined with a medium concentration of simvastatin. The optimal compound O/W emulsion adjuvant could induce effective humoral and cellular immune responses that were stronger and more stable than those induced by aluminum adjuvant and Freund's adjuvant. The OVA/HAPS-MSim-OE induced dramatically strong and persistent IgG expressions and Th1-polarized immune responses. What's more, the highest CD4+/CD8+lymphocyte ratios were observed in OVA/HAPS-MSim-OE group. In addition, compound O/W emulsion adjuvant groups significantly promoted the secretion of IFN-γ and IL-6, which also indicated that the compound O/W emulsion adjuvants could induce both enhanced Th1 and Th2-mediated immune responses but prefer the Th1-mediated ones. This study would contribute to an interesting and promising direction in the development of emulsion-based adjuvants.
Collapse
Affiliation(s)
- Shuwen Xu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing, Jiangsu 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Zhiyong Wu
- Nanjing Traditional Chinese Veterinary Medicine Research Center, Nanjing, Jiangsu 210095, PR China
| | - Gaofeng Cai
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing, Jiangsu 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Yue Zhang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing, Jiangsu 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Song Peng
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing, Jiangsu 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Lina Jiao
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing, Jiangsu 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Zhenguang Liu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing, Jiangsu 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Yang Yang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing, Jiangsu 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Deyun Wang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing, Jiangsu 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China.
| |
Collapse
|
9
|
Baruah N, Ahamad N, Halder P, Koley H, Katti DS. Facile synthesis of multi-faceted, biomimetic and cross-protective nanoparticle-based vaccines for drug-resistant Shigella: a flexible platform technology. J Nanobiotechnology 2023; 21:34. [PMID: 36710326 PMCID: PMC9884485 DOI: 10.1186/s12951-023-01780-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 01/12/2023] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND No commercial vaccines are available against drug-resistant Shigella due to serotype-specific/narrow-range of protection. Nanoparticle-based biomimetic vaccines involving stable, conserved, immunogenic proteins fabricated using facile chemistries can help formulate a translatable cross-protective Shigella vaccine. Such systems can also negate cold-chain transportation/storage thus overcoming challenges prevalent in various settings. METHODS We explored facile development of biomimetic poly (lactide-co-glycolide)/PLGA 50:50 based nanovaccines (NVs), encapsulating conserved stabilized antigen(s)/immunostimulant of S. dysenteriae 1 origin surface-modified using simple chemistries. All encapsulants (IpaC/IpaB/LPS) and nanoparticles (NPs)-bare and modified (NV), were thoroughly characterized. Effect of IpaC on cellular uptake of NPs was assessed in-vitro. Immunogenicity of the NVs was assessed in-vivo in BALB/c mice by intranasal immunization. Cross-protective efficacy was assessed by intraperitoneally challenging the immunized groups with a high dose of heterologous S. flexneri 2a and observing for visible diarrhea, weight loss and survival. Passive-protective ability of the simplest NV was assessed in the 5-day old progeny of vaccinated mice. RESULTS All the antigens and immunostimulant to be encapsulated were successfully purified and found to be stable both before and after encapsulation into NPs. The ~ 300 nm sized NPs with a zeta potential of ~ - 25 mV released ~ 60% antigen by 14th day suggesting an appropriate delivery kinetics. The NPs could be successfully surface-modified with IpaC and/or CpG DNA. In vitro experiments revealed that the presence of IpaC can significantly increase cellular uptake of NPs. All NVs were found to be cytocompatible and highly immunogenic. Antibodies in sera of NV-immunized mice could recognize heterologous Shigella. Immunized sera also showed high antibody and cytokine response. The immunized groups were protected from diarrhea and weight loss with ~ 70-80% survival upon heterologous Shigella challenge. The simplest NV showed ~ 88% survival in neonates. CONCLUSIONS Facile formulation of biomimetic NVs can result in significant cross-protection. Further, passive protection in neonates suggest that parental immunization could protect infants, the most vulnerable group in context of Shigella infection. Non-invasive route of vaccination can also lead to greater patient compliance making it amenable for mass-immunization. Overall, our work contributes towards a yet to be reported platform technology for facile development of cross-protective Shigella vaccines.
Collapse
Affiliation(s)
- Namrata Baruah
- grid.417965.80000 0000 8702 0100Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016 Uttar Pradesh India ,grid.417965.80000 0000 8702 0100The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, 208016 Uttar Pradesh India
| | - Nadim Ahamad
- grid.417965.80000 0000 8702 0100Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016 Uttar Pradesh India
| | - Prolay Halder
- grid.419566.90000 0004 0507 4551Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, 700010 West Bengal India
| | - Hemanta Koley
- grid.419566.90000 0004 0507 4551Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, 700010 West Bengal India
| | - Dhirendra S. Katti
- grid.417965.80000 0000 8702 0100Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016 Uttar Pradesh India ,grid.417965.80000 0000 8702 0100The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, 208016 Uttar Pradesh India
| |
Collapse
|
10
|
Liu M, Thijssen S, Hennink WE, Garssen J, van Nostrum CF, Willemsen LM. Oral pretreatment with β-lactoglobulin derived peptide and CpG co-encapsulated in PLGA nanoparticles prior to sensitizations attenuates cow's milk allergy development in mice. Front Immunol 2023; 13:1053107. [PMID: 36703973 PMCID: PMC9872660 DOI: 10.3389/fimmu.2022.1053107] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 12/19/2022] [Indexed: 01/07/2023] Open
Abstract
Cow's milk allergy is a common food allergy among infants. Improved hygiene conditions and loss of microbial diversity are associated with increased risk of allergy development. The intestinal immune system is essential for oral tolerance induction. In this respect, bacterial CpG DNA is known to drive Th1 and regulatory T-cell (Treg) development via Toll-Like-Receptor 9 (TLR-9) signaling, skewing away from the allergic Th2 phenotype. We aimed to induce allergen specific tolerance via oral delivery of poly (lactic-co-glycolic acid) nanoparticles (NP) co-encapsulated with a selected β-lactoglobulin derived peptide (BLG-Pep) and TLR-9 ligand CpG oligodeoxynucleotide (CpG). In vivo, 3-4-week-old female C3H/HeOuJ mice housed in individually ventilated cages received 6-consecutive-daily gavages of either PBS, whey, BLG-Pep/NP, CpG/NP, a mixture of BLG-Pep/NP plus CpG/NP or co-encapsulated BLG-Pep+CpG/NP, before 5-weekly oral sensitizations with whey plus cholera toxin (CT) or only CT (sham) and were challenged with whey 5 days after the last sensitization. The co-encapsulated BLG-Pep+CpG/NP pretreatment, but not BLG-Pep/NP, CpG/NP or the mixture of BLG-Pep/NP plus CpG/NP, prevented the whey-induced allergic skin reactivity and prevented rise in serum BLG-specific IgE compared to whey-sensitized mice. Importantly, co-encapsulated BLG-Pep+CpG/NP pretreatment reduced dendritic cell (DC) activation and lowered the frequencies of PD-L1+ DC in the mesenteric lymph nodes compared to whey-sensitized mice. By contrast, co-encapsulated BLG-Pep+CpG/NP pretreatment increased the frequency of splenic PD-L1+ DC compared to the BLG-Pep/NP plus CpG/NP recipients, in association with lower Th2 development and increased Treg/Th2 and Th1/Th2 ratios in the spleen. Oral administration of PLGA NP co-encapsulated with BLG-Pep and CpG prevented rise in serum BLG-specific IgE and symptom development while lowering splenic Th2 cell frequency in these mice which were kept under strict hygienic conditions.
Collapse
Affiliation(s)
- Mengshan Liu
- Division of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands,Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Suzan Thijssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Wim E. Hennink
- Division of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands,Department of Immunology, Nutricia Research B.V., Utrecht, Netherlands
| | - Cornelus F. van Nostrum
- Division of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Linette E. M. Willemsen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands,*Correspondence: Linette E. M. Willemsen,
| |
Collapse
|
11
|
Jiang J, Huang Y, Zeng Z, Zhao C. Harnessing Engineered Immune Cells and Bacteria as Drug Carriers for Cancer Immunotherapy. ACS NANO 2023; 17:843-884. [PMID: 36598956 DOI: 10.1021/acsnano.2c07607] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Immunotherapy continues to be in the spotlight of oncology therapy research in the past few years and has been proven to be a promising option to modulate one's innate and adaptive immune systems for cancer treatment. However, the poor delivery efficiency of immune agents, potential off-target toxicity, and nonimmunogenic tumors significantly limit its effectiveness and extensive application. Recently, emerging biomaterial-based drug carriers, including but not limited to immune cells and bacteria, are expected to be potential candidates to break the dilemma of immunotherapy, with their excellent natures of intrinsic tumor tropism and immunomodulatory activity. More than that, the tiny vesicles and physiological components derived from them have similar functions with their source cells due to the inheritance of various surface signal molecules and proteins. Herein, we presented representative examples about the latest advances of biomaterial-based delivery systems employed in cancer immunotherapy, including immune cells, bacteria, and their derivatives. Simultaneously, opportunities and challenges of immune cells and bacteria-based carriers are discussed to provide reference for their future application in cancer immunotherapy.
Collapse
Affiliation(s)
- Jingwen Jiang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, People's Republic of China
| | - Yanjuan Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, People's Republic of China
| | - Zishan Zeng
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, People's Republic of China
| | - Chunshun Zhao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, People's Republic of China
| |
Collapse
|
12
|
Wang N, Zuo Y, Wu S, Huang C, Zhang L, Zhu D. Spatio-temporal delivery of both intra- and extracellular toll-like receptor agonists for enhancing antigen-specific immune responses. Acta Pharm Sin B 2022; 12:4486-4500. [PMID: 36561992 PMCID: PMC9764069 DOI: 10.1016/j.apsb.2022.05.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/25/2022] [Accepted: 05/10/2022] [Indexed: 12/25/2022] Open
Abstract
For cancer immunotherapy, triggering toll-like receptors (TLRs) in dendritic cells (DCs) can potentiate antigen-based immune responses. Nevertheless, to generate robust and long-lived immune responses, a well-designed nanovaccine should consider different locations of TLRs on DCs and co-deliver both antigens and TLR agonist combinations to synergistically induce optimal antitumor immunity. Herein, we fabricated lipid-polymer hybrid nanoparticles (LPNPs) to spatio-temporally deliver model antigen ovalbumin (OVA) on the surface of the lipid layer, TLR4 agonist monophosphoryl lipid A (MPLA) within the lipid layer, and TLR7 agonist imiquimod (IMQ) in the polymer core to synergistically activate DCs by both extra- and intra-cellular TLRs for enhancing adaptive immune responses. LPNPs-based nanovaccines exhibited a narrow size distribution at the mean diameter of 133.23 nm and zeta potential of -2.36 mV, showed a high OVA loading (around 70.83 μg/mg) and IMQ encapsulation efficiency (88.04%). Our data revealed that LPNPs-based nanovaccines showed great biocompatibility to immune cells and an excellent ability to enhance antigen internalization, thereby promoting DCs maturation and cytokines production. Compared to Free OVA, OVA-LPNPs promoted antigen uptake, lysosome escape, depot effect and migration to secondary lymphatic organs. In vivo immunization showed that IMQ-MPLA-OVA-LPNPs with dual agonists induced more powerful cellular and humoral immune responses. Moreover, prophylactic vaccination by IMQ-MPLA-OVA-LPNPs effectively suppressed tumor growth and increased survival efficacy. Hence, the nanovaccines we fabricated can effectively co-deliver antigens and different TLR agonists and realize coordinated stimulation of DCs in a spatio-temporal manner for enhanced immune responses, which provides a promising strategy for cancer immunotherapy.
Collapse
Affiliation(s)
- Nannan Wang
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Yueyue Zuo
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Shengjie Wu
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Chenlu Huang
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Linhua Zhang
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Dunwan Zhu
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| |
Collapse
|
13
|
pH-responsive Astragalus polysaccharide-loaded PLGA nanoparticles as an adjuvant system to improve immune responses. Int J Biol Macromol 2022; 222:1936-1947. [DOI: 10.1016/j.ijbiomac.2022.09.283] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/28/2022] [Accepted: 09/29/2022] [Indexed: 11/05/2022]
|
14
|
Zhang Y, Gu P, Jiao L, He J, Yu L, Liu Z, Yang Y, Hu Y, Liu J, Wang D. Chinese yam polysaccharides PLGA-stabilized Pickering emulsion as an adjuvant system for PCV- 2 vaccine to enhance immune response. Int J Biol Macromol 2022; 219:1034-1046. [PMID: 35963357 DOI: 10.1016/j.ijbiomac.2022.08.035] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/25/2022] [Accepted: 08/07/2022] [Indexed: 11/05/2022]
Abstract
Chinese yam polysaccharides (CYP) exhibit superior adjuvant activity and modulate the immune response, but the low bioavailability limits their clinical application. Pickering emulsions have been proven as an efficient vaccine delivery system to enhance the immune response. Here, we used the Chinese yam polysaccharides PLGA-stabilized Pickering emulsion adjuvant system (CYP-PPAS) loaded with Porcine circovirus 2 as a vaccine and focused on investigating its adjuvant activity on humoral and cellular immunity in mice. The CYP-PPAS increased PCV-2 antigen loading efficiency and showed a high antigen uptake efficiency by macrophages in vitro. In vivo, CYP-PPAS significantly facilitated DCs maturation in draining lymph nodes than CYP or PPAS alone group. The CYP-PPAS also induced an increased proliferation index and a CD4+/CD8+ ratio. Meanwhile, in contrast to the CYP and PPAS groups, CYP-PPAS elicited a stronger anti-PCV-2 IgG and mixed Th1/Th2 immune response. Specifically, the CYP-PPAS group displayed the high expression of CD107a, FasL, and Granzyme B secretion to augment a strong cytotoxic lymphocyte response. Overall, the CYP-PPAS was a successful adjuvant system for promoting humoral and cellular immune responses, which opens up an avenue for the development of effective adjuvants against infectious diseases.
Collapse
Affiliation(s)
- Yue Zhang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, 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, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Lina Jiao
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Jin He
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, 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, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Zhenguang Liu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yang Yang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, 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, 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, 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, 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
|
15
|
Abstract
A favorable outcome of the COVID-19 crisis might be achieved with massive vaccination. The proposed vaccines contain several different vaccine active principles (VAP), such as inactivated virus, antigen, mRNA, and DNA, which are associated with either standard adjuvants or nanomaterials (NM) such as liposomes in Moderna's and BioNTech/Pfizer's vaccines. COVID-19 vaccine adjuvants may be chosen among liposomes or other types of NM composed for example of graphene oxide, carbon nanotubes, micelles, exosomes, membrane vesicles, polymers, or metallic NM, taking inspiration from cancer nano-vaccines, whose adjuvants may share some of their properties with those of viral vaccines. The mechanisms of action of nano-adjuvants are based on the facilitation by NM of targeting certain regions of immune interest such as the mucus, lymph nodes, and zones of infection or blood irrigation, the possible modulation of the type of attachment of the VAP to NM, in particular VAP positioning on the NM external surface to favor VAP presentation to antigen presenting cells (APC) or VAP encapsulation within NM to prevent VAP degradation, and the possibility to adjust the nature of the immune response by tuning the physico-chemical properties of NM such as their size, surface charge, or composition. The use of NM as adjuvants or the presence of nano-dimensions in COVID-19 vaccines does not only have the potential to improve the vaccine benefit/risk ratio, but also to reduce the dose of vaccine necessary to reach full efficacy. It could therefore ease the overall spread of COVID-19 vaccines within a sufficiently large portion of the world population to exit the current crisis.
Collapse
Affiliation(s)
- Edouard Alphandéry
- Sorbonne Université, Muséum National d'Histoire Naturelle, UMR CNRS 7590, IRD, Institut de Minéralogie, de Physique des Matériaux et de Cosmochimie, IMPMC, 75005 Paris, France. .,Nanobacterie SARL, 36 Boulevard Flandrin, 75116, Paris, France.,Institute of Anatomy, UZH University of Zurich, Instiute of Anatomy, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| |
Collapse
|
16
|
Kaur A, Kanwar R, Kaushik D, Sakala IG, Honda-Okubo Y, Petrovsky N, Salunke DB, Mehta SK. Combined delivery of TLR2 and TLR7 agonists by Nanostructured lipid carriers induces potent vaccine adjuvant activity in mice. Int J Pharm 2021; 613:121378. [PMID: 34915144 DOI: 10.1016/j.ijpharm.2021.121378] [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: 07/02/2021] [Revised: 11/12/2021] [Accepted: 12/09/2021] [Indexed: 10/19/2022]
Abstract
Toll-like receptor (TLR) agonists are promising adjuvants and the combination of TLR agonists enhance immune responses by providing synergistic immune activity via triggering different signalling pathways. However, systematic cytotoxicity due to the immediate release of such immune potentiators from the site of injection hampers its clinical performance. Nanostructured lipid carriers (NLCs) offer a possibility to incorporate multiple TLR agonists with high encapsulation efficiency and slow drug release. Herein, we synthesized NLCs from didodecyldimethylammonium bromide (D12DAB) and oleic acid and used these to co-encapsulate a Pam2CS derivative (T-2, TLR2 agonist) with an imidazoquinoline derivative (T-7, TLR7 agonist) as a combination vaccine adjuvant. Hydrodynamic diameter and zeta potential of the prepared NLCs were found to be in the range of 200-500 nm and 23-27 mV, respectively. Spherical shape and size of prepared NLCs were also assessed through Field Emission Scanning Electron Microscopy (FE-SEM) and Transmission Electron Microscopy (TEM) analysis. In-vitro release studies of T-7 demonstrated sustained release and the addition of lipopeptide T-2 augmented encapsulation efficiency (from 84 to 92.9%) with a slight trigger in the release percentage. All NLC formulations were screened in TLR2/1, TLR2/6, TLR7 and TLR8 reporter cell lines and loaded NLC formulation showed high TLR2 and TLR7 agonistic activity. Adjuvant potency was evaluated through intramuscular immunization of female C57BL/6 mice with recombinant hepatitis B surface antigen and influenza hemagglutinin protein. T-2 and T-7 loaded NLCs induced good protective efficacy in mice challenged with a lethal dose of influenza virus.
Collapse
Affiliation(s)
- Arshpreet Kaur
- Department of Chemistry and Centre for Advanced Studies, Panjab University, Chandigarh, India
| | - Rohini Kanwar
- Department of Chemistry and Centre for Advanced Studies, Panjab University, Chandigarh, India; Mehr Chand Mahajan DAV College for Women, Chandigarh, India
| | - Deepender Kaushik
- Department of Chemistry and Centre for Advanced Studies, Panjab University, Chandigarh, India
| | - Isaac G Sakala
- Vaxine Pty Ltd, Warradale, Australia; College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Yoshikazu Honda-Okubo
- Vaxine Pty Ltd, Warradale, Australia; College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Nikolai Petrovsky
- Vaxine Pty Ltd, Warradale, Australia; College of Medicine and Public Health, Flinders University, Adelaide, Australia.
| | - Deepak B Salunke
- Department of Chemistry and Centre for Advanced Studies, Panjab University, Chandigarh, India; National Interdisciplinary Centre of Vaccines, Immunotherapeutics and Antimicrobials, Panjab University, Chandigarh, India.
| | - Surinder K Mehta
- Department of Chemistry and Centre for Advanced Studies, Panjab University, Chandigarh, India.
| |
Collapse
|
17
|
Lim JW, Ahn YR, Park G, Kim HO, Haam S. Application of Nanomaterials as an Advanced Strategy for the Diagnosis, Prevention, and Treatment of Viral Diseases. Pharmaceutics 2021; 13:1570. [PMID: 34683863 PMCID: PMC8540357 DOI: 10.3390/pharmaceutics13101570] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/19/2021] [Accepted: 09/23/2021] [Indexed: 12/23/2022] Open
Abstract
The coronavirus disease (COVID-19) pandemic poses serious global health concerns with the continued emergence of new variants. The periodic outbreak of novel emerging and re-emerging infectious pathogens has elevated concerns and challenges for the future. To develop mitigation strategies against infectious diseases, nano-based approaches are being increasingly applied in diagnostic systems, prophylactic vaccines, and therapeutics. This review presents the properties of various nanoplatforms and discusses their role in the development of sensors, vectors, delivery agents, intrinsic immunostimulants, and viral inhibitors. Advanced nanomedical applications for infectious diseases have been highlighted. Moreover, physicochemical properties that confer physiological advantages and contribute to the control and inhibition of infectious diseases have been discussed. Safety concerns limit the commercial production and clinical use of these technologies in humans; however, overcoming these limitations may enable the use of nanomaterials to resolve current infection control issues via application of nanomaterials as a platform for the diagnosis, prevention, and treatment of viral diseases.
Collapse
Affiliation(s)
- Jong-Woo Lim
- Department of Chemical and Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seoul 03722, Korea; (J.-W.L.); (G.P.)
| | - Yu-Rim Ahn
- Division of Chemical Engineering and Bioengineering, College of Art, Culture and Engineering, Kangwon National University, Chuncheon-si 24341, Gangwon-do, Korea;
- Biohealth-machinery Convergence Engineering, Kangwon National University, Chuncheon-si 24341, Gangwon-do, Korea
| | - Geunseon Park
- Department of Chemical and Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seoul 03722, Korea; (J.-W.L.); (G.P.)
| | - Hyun-Ouk Kim
- Division of Chemical Engineering and Bioengineering, College of Art, Culture and Engineering, Kangwon National University, Chuncheon-si 24341, Gangwon-do, Korea;
- Biohealth-machinery Convergence Engineering, Kangwon National University, Chuncheon-si 24341, Gangwon-do, Korea
| | - Seungjoo Haam
- Department of Chemical and Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seoul 03722, Korea; (J.-W.L.); (G.P.)
| |
Collapse
|
18
|
Wu S, Xia Y, Hu Y, Ma G. Bio-mimic particles for the enhanced vaccinations: Lessons learnt from the natural traits and pathogenic invasion. Adv Drug Deliv Rev 2021; 176:113871. [PMID: 34311014 DOI: 10.1016/j.addr.2021.113871] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/30/2021] [Accepted: 07/11/2021] [Indexed: 12/21/2022]
Abstract
In the combat against pathogens, the immune systems were evolved with the immune recognitions against the various danger signals, which responded vigorously upon the pathogen invasions and elicited potent antibodies or T cell engagement against the re-infections. Envisage with the prevailing pandemics and increasing demands for cancer vaccines, bio-mimic particles were developed to imitate the natural traits of the pathogens, which conferred the optimal strategies to stimulate the immune engagement and let to the increased vaccine efficacy. Here, the recent development in bio-mimic particles, as well as the natural cues from the pathogens were discussed. As such, the designing principles that adapted from the physiochemical properties of the pathogens were unfolded as the surface characteristics (hydrophobic, nano-pattern, antigen display, charge), properties (size, shape, softness) and the delivered components (peptide, protein, nuclear acids, toll-like receptor (TLR) agonist, antibody). Additionally, the strategies for the efficient delivery, regarding the biodistribution, internalization and presentation of the antigens were also illustrated. Through reviewing the state-of-art in biomimetic particles, the lesson learnt from the natural traits and pathogenic invasion may shed light on the rational design for the enhanced vaccinations.
Collapse
|
19
|
Yan Y, Yao D, Li X. Immunological Mechanism and Clinical Application of PAMP Adjuvants. Recent Pat Anticancer Drug Discov 2021; 16:30-43. [PMID: 33563182 DOI: 10.2174/1574892816666210201114712] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 11/20/2020] [Accepted: 11/29/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND The host innate immune system can recognize Pathogen-Associated Molecular Patterns (PAMPs) through Pattern Recognition Receptors (PRRs), thereby initiating innate immune responses and subsequent adaptive immune responses. PAMPs can be developed as a vaccine adjuvant for modulating and optimizing antigen-specific immune responses, especially in combating viral infections and tumor therapy. Although several PAMP adjuvants have been successfully developed they are still lacking in general, and many of them are in the preclinical exploration stage. OBJECTIVE This review summarizes the research progress and development direction of PAMP adjuvants, focusing on their immune mechanisms and clinical applications. METHODS PubMed, Scopus, and Google Scholar were screened for this information. We highlight the immune mechanisms and clinical applications of PAMP adjuvants. RESULTS Because of the differences in receptor positions, specific immune cells targets, and signaling pathways, the detailed molecular mechanism and pharmacokinetic properties of one agonist cannot be fully generalized to another agonist, and each PAMP should be studied separately. In addition, combination therapy and effective integration of different adjuvants can increase the additional efficacy of innate and adaptive immune responses. CONCLUSION The mechanisms by which PAMPs exert adjuvant functions are diverse. With continuous discovery in the future, constant adjustments should be made to build new understandings. At present, the goal of therapeutic vaccination is to induce T cells that can specifically recognize and eliminate tumor cells and establish long-term immune memory. Following immune checkpoint modulation therapy, cancer treatment vaccines may be an option worthy of clinical testing.
Collapse
Affiliation(s)
- Yu Yan
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266071, China
| | - Dan Yao
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266071, China
| | - Xiaoyu Li
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266071, China
| |
Collapse
|
20
|
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
|
21
|
Zhang M, Cheng S, Jin Y, Zhang N, Wang Y. Membrane engineering of cell membrane biomimetic nanoparticles for nanoscale therapeutics. Clin Transl Med 2021; 11:e292. [PMID: 33635002 PMCID: PMC7819108 DOI: 10.1002/ctm2.292] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/06/2021] [Accepted: 01/06/2021] [Indexed: 02/06/2023] Open
Abstract
In recent years, cell membrane camouflaging technology has emerged as an important strategy of nanomedicine, and the modification on the membranes is also a promising approach to enhance the properties of the nanoparticles, such as cancer targeting, immune evasion, and phototherapy sensitivity. Indeed, diversified approaches have been exploited to re-engineer the membranes of nanoparticles in several studies. In this review, first we discuss direct modification strategy of cell membrane camouflaged nanoparticles (CM-NP) via noncovalent, covalent, and enzyme-involved methods. Second, we explore how the membranes of CM-NPs can be re-engineered at the cellular level using strategies such as genetic engineering and membranes fusion. Due to the innate biological properties and excellent biocompatibility, the functionalized cell membrane-camouflaged nanoparticles have been widely applied in the fields of drug delivery, imaging, detoxification, detection, and photoactivatable therapy.
Collapse
Affiliation(s)
- Minghai Zhang
- Department of Obstetrics and Gynecology, Renji Hospital, School of MedicineShanghai Jiaotong UniversityShanghaiChina
| | - Shanshan Cheng
- Department of Obstetrics and Gynecology, Renji Hospital, School of MedicineShanghai Jiaotong UniversityShanghaiChina
| | - Yue Jin
- Department of Obstetrics and Gynecology, Renji Hospital, School of MedicineShanghai Jiaotong UniversityShanghaiChina
| | - Nan Zhang
- Department of Obstetrics and Gynecology, Renji Hospital, School of MedicineShanghai Jiaotong UniversityShanghaiChina
| | - Yu Wang
- Department of Obstetrics and Gynecology, Renji Hospital, School of MedicineShanghai Jiaotong UniversityShanghaiChina
- Shanghai Key Laboratory of Gynecologic OncologyShanghaiChina
| |
Collapse
|
22
|
Zaheer T, Pal K, Zaheer I. Topical review on nano-vaccinology: Biochemical promises and key challenges. Process Biochem 2021; 100:237-244. [PMID: 33013180 PMCID: PMC7521878 DOI: 10.1016/j.procbio.2020.09.028] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 09/16/2020] [Accepted: 09/23/2020] [Indexed: 12/14/2022]
Abstract
Nanomaterials have wide-ranging biomedical applications in prevention, treatment and control of diseases. Nanoparticle based vaccines have proven prodigious prophylaxis of various infectious and non-infectious diseases of human and animal concern. Nano-vaccines outnumber the conventional vaccines by virtue of plasticity in physio-chemical properties and ease of administration. The efficacy of nano-based vaccines may be attributed to the improved antigen stability, minimum immuno-toxicity, sustained release, enhanced immunogenicity and the flexibility of physical features of nanoparticles. Based on these, the nano-based vaccines have potential to evoke both cellular and humoral immune responses. Targeted and highly specific immunological pathways required for solid and long lasting immunity may be achieved with specially engineered nano-vaccines. This review presents an insight into the prevention of infectious diseases (of bacterial, viral and parasitic origin) and non-infectious diseases (cancer, auto-immune diseases) using nano-vaccinology. Additionally, key challenges to the effective utilization of nano-vaccines from bench to clinical settings have been highlighted as research domains for future.
Collapse
Key Words
- CAPN, calcium-phosphate nanoparticles
- CNT, carbon nanotube
- COVID-19, Corona virus disease-2019
- Chi-Alg, chitosan alginate
- HIV, human immune deficiency virus
- HPV, human papilloma virus
- ISCOMS, immune stimulating complexes
- IgA, immunoglobulin A
- Immunity
- MERS, Middle-East respiratory syndrome
- MRSA, methcillin resistant Staphylococcus aureus
- NMVs, nano multilamellar lipid vesicles
- Nanoparticles
- PLGA, poly(lactic-co-glycolic acid)
- PSNP, polystyrene nanoparticles
- Pathogens
- Prevention
- SAPN, Self-Assembling Protein Nanoparticle
- SARS-CoV-1, severe acute respiratory syndrome Coronavirus-1
- VLP, virus like particles
- Vaccine
Collapse
Affiliation(s)
- Tean Zaheer
- Department of Parasitology, University of Agriculture, Faisalabad, Faisalabad 38040, Pakistan
| | - Kaushik Pal
- Federal University of Rio de Janeiro, Cidade Universitária, Rio de Janeiro RJ, 21941-901, Brazil
- Wuhan University, 8 East Lake South Road, Wuchang 430072, Hubei Province, China
| | - Iqra Zaheer
- Department of Pathology, University of Agriculture, Faisalabad, Faisalabad 38040, Pakistan
| |
Collapse
|
23
|
Xia Y, Song T, Hu Y, Ma G. Synthetic Particles for Cancer Vaccines: Connecting the Inherent Supply Chain. Acc Chem Res 2020; 53:2068-2080. [PMID: 32945648 DOI: 10.1021/acs.accounts.0c00336] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cancer vaccines have opened a new paradigm for safe and effective antitumor therapy, but they still suffer from shortcomings such as insufficient immunogenicity and immune tolerance, which seldom makes them the first choice in clinic. In fact, similar to providing a high-end product, a robust antitumor effect depends on the inherent supply chain, which attains, processes, and presents tumor-associated antigens via antigen presenting cells to T cells, which then leads to lysis of the cancer cells to release more antigens to complete the supply chain. Under these circumstances, the failure of cancer vaccines can be treated as a blockade or chain rupture. Thus, for effective tumor treatment, the key is to rationally design logistic systems to restore the supply chain.Under these circumstances, this Account summarizes our recent attempts to exploit the immunogenic trait of synthetic particles to enhance the distribution, presentation, and immune activations of the whole priming process in cancer vaccines: (1) Raw material (tumor antigen/signals) procurement: We illustrated the efforts to deliver antigens to antigen presenting cells (APCs) and draining lymph nodes for potent internalizations, and put more emphasis on the structural effect of sizes, charges, shapes, and assembly strategies for the antigen depot, lymph node transfer, and APC endocytosis. (2) Manufacture of cytotoxic T lymphocytes (CTLs) via APC recognition and presentation: We centered on exploiting the softness of two-dimensional graphene and Pickering emulsions to dynamically potentiate the immune recognition, and demonstrating the recent advances in lysosome escape strategies for enhanced antigen cross-presentations. (3) Marketing the accumulations of CTLs and the reversal of an immunosuppressive microenvironment within the tumor: We demonstrated the previous attempts to inherently cultivate the tumor tropism of the T cells via the multiantigenic repertoire and discussed the advances and challenges of combinatory cancer vaccines with an immune checkpoint blockade to reinforce the antitumor efficacy. Collectively, this Account aims to illustrate the potential of the particulate cancer vaccines to recapitalize the inherent host immune responses for the maximum antitumor effect. And by integrating the antitumor supply chain, optimized synthetic particles may shed light on the development of safe and effective particulate cancer vaccines.
Collapse
Affiliation(s)
- Yufei Xia
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Science, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100190, P. R. China
- Innovation Academy for Green Manufacture, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Tiantian Song
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Science, Beijing 100190, P. R. China
- Beijing Institute of Technology, Beijing 100081, P.R. China
| | - Yuning Hu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Science, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100190, P. R. China
- Innovation Academy for Green Manufacture, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Guanghui Ma
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Science, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100190, P. R. China
- Innovation Academy for Green Manufacture, Chinese Academy of Sciences, Beijing 100190, P. R. China
| |
Collapse
|
24
|
Van Herck S, De Geest BG. Nanomedicine-mediated alteration of the pharmacokinetic profile of small molecule cancer immunotherapeutics. Acta Pharmacol Sin 2020; 41:881-894. [PMID: 32451411 PMCID: PMC7471422 DOI: 10.1038/s41401-020-0425-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/20/2020] [Indexed: 12/21/2022]
Abstract
The advent of immunotherapy is a game changer in cancer therapy with monoclonal antibody- and T cell-based therapeutics being the current flagships. Small molecule immunotherapeutics might offer advantages over the biological drugs in terms of complexity, tissue penetration, manufacturing cost, stability, and shelf life. However, small molecule drugs are prone to rapid systemic distribution, which might induce severe off-target side effects. Nanotechnology could aid in the formulation of the drug molecules to improve their delivery to specific immune cell subsets. In this review we summarize the current efforts in changing the pharmacokinetic profile of small molecule immunotherapeutics with a strong focus on Toll-like receptor agonists. In addition, we give our vision on limitations and future pathways in the route of nanomedicine to the clinical practice.
Collapse
Affiliation(s)
- Simon Van Herck
- Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000, Ghent, Belgium
| | - Bruno G De Geest
- Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000, Ghent, Belgium.
| |
Collapse
|
25
|
Wilson KL, Howard GP, Coatsworth H, Dinglasan RR, Mao HQ, Plebanski M. Biodegradable PLGA- b-PEG Nanoparticles Induce T Helper 2 (Th2) Immune Responses and Sustained Antibody Titers via TLR9 Stimulation. Vaccines (Basel) 2020; 8:E261. [PMID: 32485944 PMCID: PMC7349924 DOI: 10.3390/vaccines8020261] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 05/20/2020] [Accepted: 05/25/2020] [Indexed: 12/11/2022] Open
Abstract
Sustained immune responses, particularly antibody responses, are key for protection against many endemic infectious diseases. Antibody responses are often accompanied by T helper (Th) cell immunity. Herein we study small biodegradable poly (ethylene glycol)-b-poly (lactic-co-glycolic acid) nanoparticles (PEG-b-PLGA NPs, 25-50 nm) as antigen- or adjuvant-carriers. The antigen carrier function of PEG-b-PLGA NPs was compared against an experimental benchmark polystyrene nanoparticles (PS NPs, 40-50 nm), both conjugated with the model antigen ovalbumin (OVA-PS NPs, and OVA-PEG-b-PLGA NPs). The OVA-PEG-b-PLGA NPs induced sustained antibody responses to Day 120 after two immunizations. The OVA-PEG-b-PLGA NPs as a self-adjuvanting vaccine further induced IL-4 producing T-helper cells (Th2), but not IFN-γ producing T-cells (Th1). The PEG-b-PLGA NPs as a carrier for CpG adjuvant (CpG-PEG-b-PLGA NPs) were also tested as mix-in vaccine adjuvants comparatively for protein antigens, or for protein-conjugated to PS NPs or to PEG-b-PLGA NPs. While the addition of this adjuvant NP did not further increase T-cell responses, it improved the consistency of antibody responses across all immunization groups. Together these data support further development of PEG-b-PLGA NPs as a vaccine carrier, particularly where it is desired to induce Th2 immunity and achieve sustained antibody titers in the absence of affecting Th1 immunity.
Collapse
Affiliation(s)
- Kirsty L. Wilson
- School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology (RMIT) University, Melbourne, Victoria 3084, Australia;
- Department of Immunology and Pathology, Monash University, Melbourne, Victoria 3181, Australia
| | - Gregory P. Howard
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA;
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Heather Coatsworth
- Emerging Pathogens Institute, Department of Infectious Diseases & Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA;
| | - Rhoel R. Dinglasan
- Emerging Pathogens Institute, Department of Infectious Diseases & Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA;
| | - Hai-Quan Mao
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA;
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Magdalena Plebanski
- School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology (RMIT) University, Melbourne, Victoria 3084, Australia;
- Department of Immunology and Pathology, Monash University, Melbourne, Victoria 3181, Australia
| |
Collapse
|
26
|
Bhardwaj P, Bhatia E, Sharma S, Ahamad N, Banerjee R. Advancements in prophylactic and therapeutic nanovaccines. Acta Biomater 2020; 108:1-21. [PMID: 32268235 PMCID: PMC7163188 DOI: 10.1016/j.actbio.2020.03.020] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 03/13/2020] [Accepted: 03/17/2020] [Indexed: 02/07/2023]
Abstract
Vaccines activate suitable immune responses to fight against diseases but can possess limitations such as compromised efficacy and immunogenic responses, poor stability, and requirement of adherence to multiple doses. ‘Nanovaccines’ have been explored to elicit a strong immune response with the advantages of nano-sized range, high antigen loading, enhanced immunogenicity, controlled antigen presentation, more retention in lymph nodes and promote patient compliance by a lower frequency of dosing. Various types of nanoparticles with diverse pathogenic or foreign antigens can help to overcome immunotolerance and alleviate the need of booster doses as required with conventional vaccines. Nanovaccines have the potential to induce both cell-mediated and antibody-mediated immunity and can render long-lasting immunogenic memory. With such properties, nanovaccines have shown high potential for the prevention of infectious diseases such as acquired immunodeficiency syndrome (AIDS), malaria, tuberculosis, influenza, and cancer. Their therapeutic potential has also been explored in the treatment of cancer. The various kinds of nanomaterials used for vaccine development and their effects on immune system activation have been discussed with special relevance to their implications in various pathological conditions. Statement of Significance Interaction of nanoparticles with the immune system has opened multiple avenues to combat a variety of infectious and non-infectious pathological conditions. Limitations of conventional vaccines have paved the path for nanomedicine associated benefits with a hope of producing effective nanovaccines. This review highlights the role of different types of nanovaccines and the role of nanoparticles in modulating the immune response of vaccines. The applications of nanovaccines in infectious and non-infectious diseases like malaria, tuberculosis, AIDS, influenza, and cancers have been discussed. It will help the readers develop an understanding of mechanisms of immune activation by nanovaccines and design appropriate strategies for novel nanovaccines.
Collapse
|
27
|
Deak P, Kimani F, Cassaidy B, Esser-Kahn A. Determining Whether Agonist Density or Agonist Number Is More Important for Immune Activation via Micoparticle Based Assay. Front Immunol 2020; 11:642. [PMID: 32328073 PMCID: PMC7161694 DOI: 10.3389/fimmu.2020.00642] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 03/20/2020] [Indexed: 11/26/2022] Open
Abstract
It is unknown if surface bound toll-like-receptor (TLR) agonists activate cells via density or total molecular number. To answer this question, we developed a TLR agonist surface conjugated polystyrene microparticle (MP) system. Using a library of MPs with varying TLR agonist density and number, we simultaneously observed innate immune cell MP uptake and TNFα expression using ImageStream flow cytometry on a cell by cell basis. The data shows that total TLR number and not density drives cellular activation with a threshold of approximately 105-106 TLR agonists. We believe that this information will be crucial for the design of particulate vaccine formulations.
Collapse
Affiliation(s)
| | | | | | - Aaron Esser-Kahn
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, United States
| |
Collapse
|
28
|
Wu M, Mei T, Lin C, Wang Y, Chen J, Le W, Sun M, Xu J, Dai H, Zhang Y, Xue C, Liu Z, Chen B. Melanoma Cell Membrane Biomimetic Versatile CuS Nanoprobes for Homologous Targeting Photoacoustic Imaging and Photothermal Chemotherapy. ACS APPLIED MATERIALS & INTERFACES 2020; 12:16031-16039. [PMID: 32186357 DOI: 10.1021/acsami.9b23177] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Modulating the surface properties of nanoparticles (NPs) is an important approach to accomplish immune escape, prolonged the blood retention time, and enhance the ability of targeted drug delivery. The camouflage of cancer cell membrane onto nanoparticles has been proved to be an ideal approach to enhance active targeting ability of NPs. Herein, we isolated the membrane of melanoma cells to coat doxorubicin (DOX) and indocyanine green (ICG)-loaded hollow copper sulfide NPs (ID-HCuSNP@B16F10) for targeted photothermal therapy, photoacoustic imaging, and chemotherapy. A remarkable in vitro anticancer effect after irradiation and homologous targeting can be observed in B16F10 cells after the treatment of ID-HCuSNP@B16F10. Moreover, ID-HCuSNP@B16F10 exhibits excellent photothermal effect in melanoma animal models and achieves a high tumor ablation rate. This biomimetic system can realize high drug loading efficiency, enhanced targeting ability, and ideal antitumor efficiency.
Collapse
Affiliation(s)
- Minliang Wu
- Department of Plastic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Tianxiao Mei
- Institute for Regenerative Medicine and Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Tongji University School of Medicine, Shanghai 200092, China
| | - Chenyu Lin
- Institute for Regenerative Medicine and Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Tongji University School of Medicine, Shanghai 200092, China
| | - Yuchong Wang
- Department of Plastic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Jingyao Chen
- Institute for Regenerative Medicine and Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Tongji University School of Medicine, Shanghai 200092, China
| | - Wenjun Le
- Institute for Regenerative Medicine and Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Tongji University School of Medicine, Shanghai 200092, China
| | - Mengyan Sun
- Department of Plastic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Jianguo Xu
- Department of Plastic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Haiying Dai
- Department of Plastic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Yifan Zhang
- Institute for Regenerative Medicine and Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Tongji University School of Medicine, Shanghai 200092, China
| | - Chunyu Xue
- Department of Plastic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Zhongmin Liu
- Institute for Regenerative Medicine and Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Tongji University School of Medicine, Shanghai 200092, China
| | - Bingdi Chen
- Institute for Regenerative Medicine and Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Tongji University School of Medicine, Shanghai 200092, China
| |
Collapse
|
29
|
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]
|
30
|
Vijayan V, Mohapatra A, Uthaman S, Park IK. Recent Advances in Nanovaccines Using Biomimetic Immunomodulatory Materials. Pharmaceutics 2019; 11:E534. [PMID: 31615112 PMCID: PMC6835828 DOI: 10.3390/pharmaceutics11100534] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/09/2019] [Accepted: 10/11/2019] [Indexed: 12/28/2022] Open
Abstract
The development of vaccines plays a vital role in the effective control of several fatal diseases. However, effective prophylactic and therapeutic vaccines have yet to be developed for completely curing deadly diseases, such as cancer, malaria, HIV, and serious microbial infections. Thus, suitable vaccine candidates need to be designed to elicit appropriate immune responses. Nanotechnology has been found to play a unique role in the design of vaccines, providing them with enhanced specificity and potency. Nano-scaled materials, such as virus-like particles, liposomes, polymeric nanoparticles (NPs), and protein NPs, have received considerable attention over the past decade as potential carriers for the delivery of vaccine antigens and adjuvants, due to their beneficial advantages, like improved antigen stability, targeted delivery, and long-time release, for which antigens/adjuvants are either encapsulated within, or decorated on, the NP surface. Flexibility in the design of nanomedicine allows for the programming of immune responses, thereby addressing the many challenges encountered in vaccine development. Biomimetic NPs have emerged as innovative natural mimicking biosystems that can be used for a wide range of biomedical applications. In this review, we discuss the recent advances in biomimetic nanovaccines, and their use in anti-bacterial therapy, anti-HIV therapy, anti-malarial therapy, anti-melittin therapy, and anti-tumor immunity.
Collapse
Affiliation(s)
- Veena Vijayan
- Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju 58128, Korea.
| | - Adityanarayan Mohapatra
- Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju 58128, Korea.
| | - Saji Uthaman
- Department of Polymer Science and Engineering, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Korea.
| | - In-Kyu Park
- Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju 58128, Korea.
| |
Collapse
|
31
|
Baindara P, Mandal SM. Antimicrobial Peptides and Vaccine Development to Control Multi-drug Resistant Bacteria. Protein Pept Lett 2019; 26:324-331. [PMID: 31237198 DOI: 10.2174/0929866526666190228162751] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 02/10/2019] [Accepted: 02/12/2019] [Indexed: 12/20/2022]
Abstract
Antimicrobial resistance (AMR) reported to increase globally at alarming levels in the recent past. A number of potential alternative solutions discussed and implemented to control AMR in bacterial pathogens. Stringent control over the clinical application of antibiotics for a reduction in uses is a special consideration along with alternative solutions to fight against AMR. Although alternatives to conventional antibiotics like antimicrobial peptides (AMP) might warrant serious consideration to fight against AMR, there is a thriving recognition for vaccines in encountering the problem of AMR. Vaccines can reduce the prevalence of AMR by reducing the number of specific pathogens, which result in cutting down the antimicrobial need and uses. However, conventional vaccines produced using live or attenuated microorganisms while the presence of immunologically redundant biological components or impurities might cause major side effects and health related problems. Here we discussed AMPs based vaccination strategies as an emerging concept to overcome the disadvantages of traditional vaccines while boosting the AMPs to control multidrug resistant bacteria or AMR. Nevertheless, the poor immune response is a major challenge in the case of peptide vaccines as minimal antigenic epitopes used for immunization in peptide vaccines.
Collapse
Affiliation(s)
- Piyush Baindara
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Santi M Mandal
- Central Research Facility, Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur 721302, WB, India
| |
Collapse
|
32
|
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: 92] [Impact Index Per Article: 15.3] [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
|
33
|
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
|
34
|
Wu M, Le W, Mei T, Wang Y, Chen B, Liu Z, Xue C. Cell membrane camouflaged nanoparticles: a new biomimetic platform for cancer photothermal therapy. Int J Nanomedicine 2019; 14:4431-4448. [PMID: 31354269 PMCID: PMC6588714 DOI: 10.2147/ijn.s200284] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 04/03/2019] [Indexed: 12/17/2022] Open
Abstract
Targeted drug delivery by nanoparticles (NPs) is an essential technique to achieve the ideal therapeutic effect for cancer. However, it requires large amounts of work to imitate the biomarkers on the surface of the cell membrane and cannot fully retain the bio-function and interactions among cells. Cell membranes have been studied to form biomimetic NPs to achieve functions like immune escape, targeted drug delivery, and immune modulation, which inherit the ability to interact with the in vivo environments. Currently, erythrocyte, leukocyte, mesenchymal stem cell, cancer cell and platelet have been applied in coating photothermal agents and anti-cancer drugs to achieve increased photothermal conversion efficiency and decreased side effects in cancer ablation. In this review, we discuss the recent development of cell membrane-coated NPs in the application of photothermal therapy and cancer targeting. The underlying biomarkers of cell membrane-coated nanoparticles (CMNPs) are discussed, and future research directions are suggested.
Collapse
Affiliation(s)
- Minliang Wu
- Department of Plastic Surgery,Changhai Hospital, Second Military Medical University, Shanghai200433, People’s Republic of China
| | - Wenjun Le
- Institute for Regenerative Medicine and Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai200092, People’s Republic of China
| | - Tianxiao Mei
- Institute for Regenerative Medicine and Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai200092, People’s Republic of China
| | - Yuchong Wang
- Department of Plastic Surgery,Changhai Hospital, Second Military Medical University, Shanghai200433, People’s Republic of China
| | - Bingdi Chen
- Institute for Regenerative Medicine and Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai200092, People’s Republic of China
| | - Zhongmin Liu
- Institute for Regenerative Medicine and Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai200092, People’s Republic of China
| | - Chunyu Xue
- Department of Plastic Surgery,Changhai Hospital, Second Military Medical University, Shanghai200433, People’s Republic of China
| |
Collapse
|
35
|
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: 75] [Impact Index Per Article: 12.5] [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]
|
36
|
Nanomaterial Exposure Induced Neutrophil Extracellular Traps: A New Target in Inflammation and Innate Immunity. J Immunol Res 2019; 2019:3560180. [PMID: 30944832 PMCID: PMC6421747 DOI: 10.1155/2019/3560180] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 01/04/2019] [Accepted: 01/21/2019] [Indexed: 02/05/2023] Open
Abstract
Nanotechnology has become a novel subject with impact in many research and technology areas. Nanoparticles (NPs), as a key component in nanotechnology, are widely used in many areas such as optical, magnetic, electrical, and mechanical engineering. The biomedical and pharmaceutical industries have embraced NPs as a viable drug delivery modality. As such, the potential for NP-induced cytotoxicity has emerged as a major concern for NP drug delivery systems. Thus, it is important to understand how NPs affect the innate immune system. As the most abundant myeloid cell type in innate immune responses, neutrophils are critical for concerns about potentially toxic side effects of NPs. When activated by innate immune stimuli, neutrophils may initiate NETosis to release neutrophil extracellular traps (NETs). Herein, we have reviewed the relationship between NPs and the induction of NETosis and release of NETs.
Collapse
|
37
|
Gu P, Liu Z, Sun Y, Ou N, Hu Y, Liu J, Wu Y, Wang D. Angelica sinensis polysaccharide encapsulated into PLGA nanoparticles as a vaccine delivery and adjuvant system for ovalbumin to promote immune responses. Int J Pharm 2018; 554:72-80. [PMID: 30399435 DOI: 10.1016/j.ijpharm.2018.11.008] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 09/20/2018] [Accepted: 11/03/2018] [Indexed: 01/27/2023]
Abstract
Nanoparticles (NPs)-based vaccine delivery systems are widely used for their ability to control the release of antigens and promote immune responses against cancer or infectious diseases. In this study, the immunopotentiator Angelica sinensis polysaccharide (ASP) and model protein antigen ovalbumin (OVA) were encapsulated into Poly(lactic-co-glycolic acid) (PLGA) to formulate the novel NPs-based vaccine delivery system (ASP-PLGA/OVA). These formulations were subcutaneously administered to mice, then the magnitude and kinetics of antibody and cellular immune responses were assessed. The ASP-PLGA/OVA NPs were pherical in shape with smooth surfaces, approximately 225.2 nm in average size, negatively charged (around -11.27 mV), and the encapsulation efficiency of OVA at around 66.28%, respectively. Furthermore, ASP-PLGA/OVA NPs could keep stable at 4 °C over 30 days and provide a sustained and controlled release of OVA from the NPs. The results demonstrated that mice immunized with ASP-PLGA/OVA NPs could significantly enhance lymphocyte proliferation and improve the ratio of CD4+ to CD8+ T cells, thereby ASP-PLGA/OVA NPs could induce a strong cellular immune response. Moreover, the ASP-PLGA/OVA NPs could induce vigorous and long-term IgG immune responses with a mixed Th1 and Th2 responses and up-regulate the levels of Th-associated cytokines. These results suggested that ASP-PLGA/OVA NPs, which stimulated strong and continuous antibody responses and induced cellular immune responses, could potentially serve as an efficient and safe vaccine delivery and adjuvant system against infections and diseases.
Collapse
Affiliation(s)
- Pengfei Gu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Zhenguang Liu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yaqin Sun
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Ning Ou
- Institute of Traditional Chinese Veterinary Medicine, 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
| | - Jiaguo Liu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yi Wu
- Institute of Traditional Chinese Veterinary Medicine, 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.
| |
Collapse
|
38
|
Collier MA, Junkins RD, Gallovic MD, Johnson BM, Johnson MM, Macintyre AN, Sempowski GD, Bachelder EM, Ting JPY, Ainslie KM. Acetalated Dextran Microparticles for Codelivery of STING and TLR7/8 Agonists. Mol Pharm 2018; 15:4933-4946. [PMID: 30281314 DOI: 10.1021/acs.molpharmaceut.8b00579] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Vaccines are the most effective tool for preventing infectious diseases; however, subunit vaccines, considered the safest type, suffer from poor immunogenicity and require adjuvants to create a strong and sustained immune response. As adjuvants, pathogen-associated molecular patterns (PAMPs) offer potent immunostimulatory properties and defined mechanisms of action through their cognate pattern recognition receptors (PRRs). Their activity can be further enhanced through combining two or more PAMPs, particularly those that activate multiple immune signaling pathways. However, the cytosolic localization of many PRRs requires intracellular delivery of PAMPs for optimal biological activity, which is particularly true of the stimulator of interferon genes (STING) PRR. Using acetalated dextran (Ace-DEX) microparticles (MPs) encapsulating STING agonist 3'3'-cyclic GMP-AMP (cGAMP) combined with soluble PAMPS, we screened the effect of codelivery of adjuvants using primary mouse bone marrow derived dendritic cells (BMDCs). We identified that codelivery of cGAMP MPs and soluble Toll-like receptor 7/8 (TLR7/8) agonist resiquimod (R848) elicited the broadest cytokine response. cGAMP and R848 were then coencapsulated within Ace-DEX MPs via electrospray. Using the model antigen ovalbumin, we observed that Ace-DEX MPs coencapsulating cGAMP and R848 (cGAMP/R848 Ace-DEX MPs) induced antigen-specific cellular immunity, and a balanced Th1/Th2 humoral response that was greater than cGAMP Ace-DEX MPs alone and PAMPs delivered in separate MPs. These data indicate that polymeric Ace-DEX MPs loaded with STING and TLR7/8 agonists represent a potent cellular and humoral vaccine adjuvant.
Collapse
Affiliation(s)
| | | | | | | | | | - Andrew N Macintyre
- Duke Human Vaccine Institute , Duke University Medical Center , Durham , North Carolina 27710 , United States
| | - Gregory D Sempowski
- Duke Human Vaccine Institute , Duke University Medical Center , Durham , North Carolina 27710 , United States
| | | | | | | |
Collapse
|
39
|
Jin JW, Tang SQ, Rong MZ, Zhang MQ. Synergistic effect of dual targeting vaccine adjuvant with aminated β-glucan and CpG-oligodeoxynucleotides for both humoral and cellular immune responses. Acta Biomater 2018; 78:211-223. [PMID: 30098441 DOI: 10.1016/j.actbio.2018.08.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 07/10/2018] [Accepted: 08/03/2018] [Indexed: 12/13/2022]
Abstract
Presently, clinically approved adjuvants (such as aluminum salts) fail to induce cellular immune responses, which is crucial to defend against intracellular pathogens (including HIV, malaria, tuberculosis and Ebola) and cancer. However, Freund's complete adjuvant potently stimulates both humoral and cellular immune responses, accompanying by high toxicity and severe side reactions. Here in this work, a CpG-oligodeoxynucleotides (CpG-OND) crosslinked aminated β-glucan-Ovalbumin dual targeting nanoparticle (CpG-OND-AG-OVA) is prepared through a simple and mild ionic complexation method. The aminated β-glucan plays dual roles as antigen presenting cells (APCs) targeted carrier and immunopotentiator (targeting and activating dectin-1 on APCs). Meanwhile, CpG-OND also plays dual roles as ionic crosslinker and immunopotentiator (targeting and activating Toll-like receptor 9 in APCs). The adjuvant activity of the particles is evaluated through in vitro and in vivo experiments. The particles significantly enhance uptake and sustained proteolytic processing of antigens, and result in APCs maturation, inducing robust Th1 and Th2-type immune responses comparable to Freund's adjuvant without obvious toxicity. The potent adjuvant activity of the nanoparticles may originate from dual targeting synergistic effects between aminated β-glucan and CpG-OND. Accordingly, the dual targeting nanoparticles may be a promising vaccine adjuvant for inducing robust humoral and cellular immune responses against infectious diseases and cancers. STATEMENT OF SIGNIFICANCE An ideal adjuvant for subunit vaccine should act as both a carrier to enhance the uptake, sustained processing and cytosolic delivery of antigens, and an immunopotentiator to stimulate antigen presenting cells (APCs) for activation of naive T cells. Additionally, it should be easy to obtain and safe with negligible toxicity. Unfortunately, both synthetic and natural polymers that have been developed into antigen delivery system cannot completely fulfill the requirements. In the present study, the authors design nanoparticles with aminated β-glucan and CpG-oligodeoxynucleotides (CpG-OND) through a simple and mild method. β-Glucan (a dectin-1 and TLR2 targeted PAMP) and CpG-OND (a TLR9 targeted PAMP) are readily accessible. Aminated β-glucan plays dual roles in the nanoparticle as APCs targeted carrier and immunopotentiator. Meanwhile, CpG-OND also plays dual roles as crosslinker and APCs targeted immunopotentiator. By making use of synergistic effect of the dual targeting vaccine adjuvant with aminated β-glucan and CpG-OND, the nanoparticles induce robust antigen specific immune responses comparable to Freund's adjuvant without obvious toxicity.
Collapse
Affiliation(s)
- Jing Wei Jin
- Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, GD HPPC Lab, School of Chemistry, Sun Yat-sen University, Guangzhou 510275, China
| | - Shun Qing Tang
- Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Min Zhi Rong
- Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, GD HPPC Lab, School of Chemistry, Sun Yat-sen University, Guangzhou 510275, China.
| | - Ming Qiu Zhang
- Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, GD HPPC Lab, School of Chemistry, Sun Yat-sen University, Guangzhou 510275, China.
| |
Collapse
|
40
|
Jin JW, Peng WL, Tang SQ, Rong MZ, Zhang MQ. Antigen uptake and immunoadjuvant activity of pathogen-mimetic hollow silica particles conjugated with β-glucan. J Mater Chem B 2018; 6:6288-6301. [PMID: 32254619 DOI: 10.1039/c8tb02129e] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The aim of vaccines is to imitate the immune responses induced by pathogen infection without causing disease. Therefore, strategies of designing vaccine delivery systems by mimicking key features of pathogens are often used. For this purpose, the present study prepares pathogen-mimicking β-glucan-conjugated hollow silica particles by using polystyrene or bacteria particles as templates. The particles perfectly duplicate the structure and morphology of pathogens and possess excellent properties of hollow silica particles, including large opening pore channels, large interior cavities, high loading of OVA (ovalbumin) and controlled release capability, biocompatibility, tunability of surface functionality and immunopotentiating activity. In addition, the particles are antigen presenting cells (APCs) targeted by specific interaction with β-glucan specific receptors on the surface of APCs, which can enhance the uptake and sustained proteolytic processing of antigens and induce APC maturation. Eventually, potent Th1 and Th2-type immune responses are aroused. The size and shape of the particles have a significant impact on the antigen uptake and immunoadjuvant activity. The degree of antigen uptake enhancement is ranked in the following order: PS HSP@glucan (nanoscale spherical) > E. coli HSP@glucan (micron-sized rod-like) > S. aureus HSP@glucan (micron-sized spherical). The PS HSP@glucan is more apt to induce a Th1-type immune response, while the E. coli HSP@glucan is more apt to induce a Th2-type immune response. The particles may thus provide a promising strategy for development of novel vaccine delivery systems for inducing robust humoral and cellular immune responses against infectious diseases and cancers.
Collapse
Affiliation(s)
- Jing Wei Jin
- Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, GD HPPC Lab, School of Chemistry, Sun Yat-sen University, Guangzhou 510275, China.
| | | | | | | | | |
Collapse
|
41
|
Kuai R, Sun X, Yuan W, Ochyl LJ, Xu Y, Hassani Najafabadi A, Scheetz L, Yu MZ, Balwani I, Schwendeman A, Moon JJ. Dual TLR agonist nanodiscs as a strong adjuvant system for vaccines and immunotherapy. J Control Release 2018; 282:131-139. [PMID: 29702142 PMCID: PMC6056764 DOI: 10.1016/j.jconrel.2018.04.041] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 04/23/2018] [Accepted: 04/23/2018] [Indexed: 12/17/2022]
Abstract
Recent studies have shown that certain combinations of Toll-like receptor (TLR) agonists can induce synergistic immune activation. However, it remains challenging to achieve such robust responses in vivo in a manner that is effective, facile, and amenable for clinical translation. Here, we show that MPLA, a TLR4 agonist, and CpG, a TLR9 agonist, can be efficiently co-loaded into synthetic high-density lipoprotein nanodiscs, forming a potent adjuvant system (ND-MPLA/CpG) that can be readily combined with a variety of subunit antigens, including proteins and peptides. ND-MPLA/CpG significantly enhanced activation of dendritic cells, compared with free dual adjuvants or nanodiscs delivering a single TLR agonist. Importantly, mice immunized with physical mixtures of protein antigens ND-MPLA/CpG generated strong humoral responses, including induction of IgG responses against protein convertase subtilisin/kexin 9 (PCSK9), leading to 17-30% reduction of the total plasma cholesterol levels. Moreover, ND-MPLA/CpG exerted strong anti-tumor efficacy in multiple murine tumor models. Compared with free adjuvants, ND-MPLA/CpG admixed with ovalbumin markedly improved antigen-specific CD8+ T cell responses by 8-fold and promoted regression of B16F10-OVA melanoma (P < 0.0001). Furthermore, ND-MPLA/CpG admixed with E7 peptide antigen elicited ~20% E7-specific CD8+ T cell responses and achieved complete regression of established TC-1 tumors in all treated animals. Taken together, our work highlights the simplicity, versatility, and potency of dual TLR agonist nanodiscs for applications in vaccines and cancer immunotherapy.
Collapse
Affiliation(s)
- Rui Kuai
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Xiaoqi Sun
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Wenmin Yuan
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lukasz J Ochyl
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yao Xu
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Alireza Hassani Najafabadi
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lindsay Scheetz
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Min-Zhi Yu
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ishina Balwani
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA.
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
42
|
Lin LCW, Chattopadhyay S, Lin JC, Hu CMJ. Advances and Opportunities in Nanoparticle- and Nanomaterial-Based Vaccines against Bacterial Infections. Adv Healthc Mater 2018; 7:e1701395. [PMID: 29508547 DOI: 10.1002/adhm.201701395] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 01/22/2018] [Indexed: 02/06/2023]
Abstract
As the dawn of the postantibiotic era we approach, antibacterial vaccines are becoming increasingly important for managing bacterial infection and reducing the need for antibiotics. Despite the success of vaccination, vaccines remain unavailable for many pressing microbial diseases, including tuberculosis, chlamydia, and staphylococcus infections. Amid continuing research efforts in antibacterial vaccine development, the advancement of nanomaterial engineering has brought forth new opportunities in vaccine designs. With increasing knowledge in antibacterial immunity and immunologic adjuvants, innovative nanoparticles are designed to elicit the appropriate immune responses for effective antimicrobial defense. Rationally designed nanoparticles are demonstrated to overcome delivery barriers to shape the adaptive immunity. This article reviews the advances in nanoparticle- and nanomaterial-based antibacterial vaccines and summarizes the development of nanoparticulate adjuvants for immune potentiation against microbial pathogens. In addition, challenges and progress in ongoing antibacterial vaccine development are discussed to highlight the opportunities for future vaccine designs.
Collapse
Affiliation(s)
- Leon Chien-Wei Lin
- Institute of Biomedical Sciences; Academia Sinica; 128, Sec. 2, Academia Road Nangang District Taipei 11529 Taiwan
| | - Saborni Chattopadhyay
- Institute of Biomedical Sciences; Academia Sinica; 128, Sec. 2, Academia Road Nangang District Taipei 11529 Taiwan
| | - Jung-Chen Lin
- Institute of Biomedical Sciences; Academia Sinica; 128, Sec. 2, Academia Road Nangang District Taipei 11529 Taiwan
| | - Che-Ming Jack Hu
- Institute of Biomedical Sciences; Academia Sinica; 128, Sec. 2, Academia Road Nangang District Taipei 11529 Taiwan
| |
Collapse
|
43
|
Ai X, Hu M, Wang Z, Zhang W, Li J, Yang H, Lin J, Xing B. Recent Advances of Membrane-Cloaked Nanoplatforms for Biomedical Applications. Bioconjug Chem 2018; 29:838-851. [PMID: 29509403 DOI: 10.1021/acs.bioconjchem.8b00103] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In terms of the extremely small size and large specific surface area, nanomaterials often exhibit unusual physical and chemical properties, which have recently attracted considerable attention in bionanotechnology and nanomedicine. Currently, the extensive usage of nanotechnology in medicine holds great potential for precise diagnosis and effective therapeutics of various human diseases in clinical practice. However, a detailed understanding regarding how nanomedicine interacts with the intricate environment in complex living systems remains a pressing and challenging goal. Inspired by the diversified membrane structures and functions of natural prototypes, research activities on biomimetic and bioinspired membranes, especially for those cloaking nanosized platforms, have increased exponentially. By taking advantage of the flexible synthesis and multiple functionality of nanomaterials, a variety of unique nanostructures including inorganic nanocrystals and organic polymers have been widely devised to substantially integrate with intrinsic biomoieties such as lipids, glycans, and even cell and bacteria membrane components, which endow these abiotic nanomaterials with specific biological functionalities for the purpose of detailed investigation of the complicated interactions and activities of nanomedicine in living bodies, including their immune response activation, phagocytosis escape, and subsequent clearance from vascular system. In this review, we summarize the strategies established recently for the development of biomimetic membrane-cloaked nanoplatforms derived from inherent host cells (e.g., erythrocytes, leukocytes, platelets, and exosomes) and invasive pathogens (e.g., bacteria and viruses), mainly attributed to their versatile membrane properties in biological fluids. Meanwhile, the promising biomedical applications based on nanoplatforms inspired by diverse moieties, such as selective drug delivery in targeted sites and effective vaccine development for disease prevention, have also been outlined. Finally, the potential challenges and future prospects of the biomimetic membrane-cloaked nanoplatforms are also discussed.
Collapse
Affiliation(s)
- Xiangzhao Ai
- Division of Chemistry and Biological Chemistry, School of Physical & Mathematical Sciences , Nanyang Technological University , Singapore , 637371
| | - Ming Hu
- Division of Chemistry and Biological Chemistry, School of Physical & Mathematical Sciences , Nanyang Technological University , Singapore , 637371
| | - Zhimin Wang
- Division of Chemistry and Biological Chemistry, School of Physical & Mathematical Sciences , Nanyang Technological University , Singapore , 637371
| | - Wenmin Zhang
- Division of Chemistry and Biological Chemistry, School of Physical & Mathematical Sciences , Nanyang Technological University , Singapore , 637371.,College of Chemistry , Fuzhou University , Fuzhou , Fujian 350116 , China
| | - Juan Li
- College of Chemistry , Fuzhou University , Fuzhou , Fujian 350116 , China
| | - Huanghao Yang
- College of Chemistry , Fuzhou University , Fuzhou , Fujian 350116 , China
| | - Jun Lin
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry , Chinese Academy of Sciences , Changchun , 130022 , China
| | - Bengang Xing
- Division of Chemistry and Biological Chemistry, School of Physical & Mathematical Sciences , Nanyang Technological University , Singapore , 637371.,College of Chemistry , Fuzhou University , Fuzhou , Fujian 350116 , China
| |
Collapse
|
44
|
Optimization of angelica sinensis polysaccharide-loaded Poly (lactic-co-glycolicacid) nanoparticles by RSM and its immunological activity in vitro. Int J Biol Macromol 2018; 107:222-229. [DOI: 10.1016/j.ijbiomac.2017.08.176] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 07/12/2017] [Accepted: 08/30/2017] [Indexed: 12/17/2022]
|
45
|
Liu Y, Hardie J, Zhang X, Rotello VM. Effects of engineered nanoparticles on the innate immune system. Semin Immunol 2017; 34:25-32. [PMID: 28985993 PMCID: PMC5705289 DOI: 10.1016/j.smim.2017.09.011] [Citation(s) in RCA: 193] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 09/26/2017] [Accepted: 09/27/2017] [Indexed: 02/04/2023]
Abstract
Engineered nanoparticles (NPs) have broad applications in industry and nanomedicine. When NPs enter the body, interactions with the immune system are unavoidable. The innate immune system, a non-specific first line of defense against potential threats to the host, immediately interacts with introduced NPs and generates complicated immune responses. Depending on their physicochemical properties, NPs can interact with cells and proteins to stimulate or suppress the innate immune response, and similarly activate or avoid the complement system. NPs size, shape, hydrophobicity and surface modification are the main factors that influence the interactions between NPs and the innate immune system. In this review, we will focus on recent reports about the relationship between the physicochemical properties of NPs and their innate immune response, and their applications in immunotherapy.
Collapse
Affiliation(s)
- Yuanchang Liu
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA, 01003, USA
| | - Joseph Hardie
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA, 01003, USA
| | - Xianzhi Zhang
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA, 01003, USA
| | - Vincent M Rotello
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA, 01003, USA.
| |
Collapse
|
46
|
Riitho V, Walters AA, Somavarapu S, Lamp B, Rümenapf T, Krey T, Rey FA, Oviedo-Orta E, Stewart GR, Locker N, Steinbach F, Graham SP. Design and evaluation of the immunogenicity and efficacy of a biomimetic particulate formulation of viral antigens. Sci Rep 2017; 7:13743. [PMID: 29062078 PMCID: PMC5653838 DOI: 10.1038/s41598-017-13915-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 09/19/2017] [Indexed: 11/17/2022] Open
Abstract
Subunit viral vaccines are typically not as efficient as live attenuated or inactivated vaccines at inducing protective immune responses. This paper describes an alternative ‘biomimetic’ technology; whereby viral antigens were formulated around a polymeric shell in a rationally arranged fashion with a surface glycoprotein coated on to the surface and non-structural antigen and adjuvant encapsulated. We evaluated this model using BVDV E2 and NS3 proteins formulated in poly-(D, L-lactic-co-glycolic acid) (PLGA) nanoparticles adjuvanted with polyinosinic:polycytidylic acid (poly(I:C) as an adjuvant (Vaccine-NP). This Vaccine-NP was compared to ovalbumin and poly(I:C) formulated in a similar manner (Control-NP) and a commercial adjuvanted inactivated BVDV vaccine (IAV), all inoculated subcutaneously and boosted prior to BVDV-1 challenge. Significant virus-neutralizing activity, and E2 and NS3 specific antibodies were observed in both Vaccine-NP and IAV groups following the booster immunisation. IFN-γ responses were observed in ex vivo PBMC stimulated with E2 and NS3 proteins in both vaccinated groups. We observed that the protection afforded by the particulate vaccine was comparable to the licenced IAV formulation. In conclusion, the biomimetic particulates showed a promising immunogenicity and efficacy profile that may be improved by virtue of being a customisable mode of delivery.
Collapse
Affiliation(s)
- Victor Riitho
- Virology Department, Animal and Plant Health Agency, Woodham Lane, Addlestone, KT15 3NB, United Kingdom.,Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, United Kingdom.,International Livestock Research Institute, P.O. Box 30709, Nairobi, 00100, Kenya
| | - Adam A Walters
- Virology Department, Animal and Plant Health Agency, Woodham Lane, Addlestone, KT15 3NB, United Kingdom.,Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, United Kingdom.,The Jenner Institute, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, United Kingdom
| | | | - Benjamin Lamp
- Institute for Virology, University of Veterinary Medicine Vienna, Veterinaerplatz 1, 1210, Vienna, Austria
| | - Till Rümenapf
- Institute for Virology, University of Veterinary Medicine Vienna, Veterinaerplatz 1, 1210, Vienna, Austria
| | - Thomas Krey
- Institut Pasteur, Unité de Virologie Structurale, Department Virologie, Paris CNRS UMR, 3569, Paris, France.,Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.,German Center for Infection Research (DZIF), 30625, Hannover, Germany
| | - Felix A Rey
- Institut Pasteur, Unité de Virologie Structurale, Department Virologie, Paris CNRS UMR, 3569, Paris, France
| | - Ernesto Oviedo-Orta
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, United Kingdom.,Sanofi Pasteur, 1541, Avenue Marcel Merieux - Campus Merieux, 69280, Marcy, L'Etoile, France
| | - Graham R Stewart
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, United Kingdom
| | - Nicolas Locker
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, United Kingdom
| | - Falko Steinbach
- Virology Department, Animal and Plant Health Agency, Woodham Lane, Addlestone, KT15 3NB, United Kingdom.,Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, United Kingdom
| | - Simon P Graham
- Virology Department, Animal and Plant Health Agency, Woodham Lane, Addlestone, KT15 3NB, United Kingdom. .,Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, United Kingdom. .,The Pirbright Institute, Ash Road, Pirbright, Woking, GU24 0NF, United Kingdom.
| |
Collapse
|
47
|
Ebrahimian M, Hashemi M, Maleki M, Hashemitabar G, Abnous K, Ramezani M, Haghparast A. Co-delivery of Dual Toll-Like Receptor Agonists and Antigen in Poly(Lactic-Co-Glycolic) Acid/Polyethylenimine Cationic Hybrid Nanoparticles Promote Efficient In Vivo Immune Responses. Front Immunol 2017; 8:1077. [PMID: 28955328 PMCID: PMC5601407 DOI: 10.3389/fimmu.2017.01077] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Accepted: 08/18/2017] [Indexed: 11/16/2022] Open
Abstract
Strategies to design delivery vehicles are critical in modern vaccine-adjuvant development. Nanoparticles (NPs) encapsulating antigen(s) and adjuvant(s) are promising vehicles to deliver antigen(s) and adjuvant(s) to antigen-presenting cells (APCs), allowing optimal immune responses against a specific pathogen. In this study, we developed a novel adjuvant delivery approach for induction of efficient in vivo immune responses. Polyethylenimine (PEI) was physically conjugated to poly(lactic-co-glycolic) acid (PLGA) to form PLGA/PEI NPs. This complex was encapsulated with resiquimod (R848) as toll-like receptor (TLR) 7/8 agonist, or monophosphoryl lipid A (MPLA) as TLR4 agonist and co-assembled with cytosine-phosphorothioate-guanine oligodeoxynucleotide (CpG ODN) as TLR9 agonist to form a tripartite formulation [two TLR agonists (inside and outside NPs) and PLGA/PEI NPs as delivery system]. The physicochemical characteristics, cytotoxicity and cellular uptake of these synthesized delivery vehicles were investigated. Cellular viability test revealed no pronounced cytotoxicity as well as increased cellular uptake compared to control groups in murine macrophage cells (J774 cell line). In the next step, PLGA (MPLA or R848)/PEI (CpG ODN) were co-delivered with ovalbumin (OVA) encapsulated into PLGA NPs to enhance the induction of immune responses. The immunogenicity properties of these co-delivery formulations were examined in vivo by evaluating the cytokine (IFN-γ, IL-4, and IL-1β) secretion and antibody (IgG1, IgG2a) production. Robust and efficient immune responses were achieved after in vivo administration of PLGA (MPLA or R848)/PEI (CpG ODN) co-delivered with OVA encapsulated in PLGA NPs in BALB/c mice. Our results demonstrate a rational design of using dual TLR agonists in a context-dependent manner for efficient nanoparticulate adjuvant-vaccine development.
Collapse
Affiliation(s)
- Mahboubeh Ebrahimian
- Division of Biotechnology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
- Immunology Section, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Maryam Hashemi
- Nanotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohsen Maleki
- Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Gholamreza Hashemitabar
- Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Haghparast
- Division of Biotechnology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
- Immunology Section, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
48
|
Effects of gold nanoparticle-based vaccine size on lymph node delivery and cytotoxic T-lymphocyte responses. J Control Release 2017; 256:56-67. [DOI: 10.1016/j.jconrel.2017.04.024] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 03/19/2017] [Accepted: 04/17/2017] [Indexed: 01/05/2023]
|
49
|
Combinatorial drug delivery approaches for immunomodulation. Adv Drug Deliv Rev 2017; 114:161-174. [PMID: 28532690 DOI: 10.1016/j.addr.2017.05.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 05/05/2017] [Accepted: 05/17/2017] [Indexed: 12/19/2022]
Abstract
Immunotherapy has been widely explored for applications to both augment and suppress intrinsic host immunity. Clinical achievements have seen a number of immunotherapeutic drugs displace established strategies like chemotherapy in treating immune-associated diseases. However, single drug approaches modulating an individual arm of the immune system are often incompletely effective. Imperfect mechanistic understanding and heterogeneity within disease pathology have seen monotherapies inadequately equipped to mediate complete disease remission. Recent success in applications of combinatorial immunotherapy has suggested that targeting multiple biological pathways simultaneously may be critical in treating complex immune pathologies. Drug delivery approaches through engineered biomaterials offer the potential to augment desired immune responses while mitigating toxic side-effects by localizing immunotherapy. This review discusses recent advances in immunotherapy and highlights newly explored combinatorial drug delivery approaches. Furthermore, prospective future directions for immunomodulatory drug delivery to exploit are provided.
Collapse
|
50
|
Zhang X, Zhao X, Luckanagul JA, Yan J, Nie Y, Lee LA, Wang Q. Polymer-Protein Core-Shell Nanoparticles for Enhanced Antigen Immunogenicity. ACS Macro Lett 2017; 6:442-446. [PMID: 35610867 DOI: 10.1021/acsmacrolett.7b00049] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Nanoengineered vaccine platforms can be modeled after viruses and other pathogens with highly organized and repetitive structures that trigger the host immune system. Here we demonstrated a pyridine-grafted poly(ε-caprolactone)-based polymer-protein core-shell nanoparticles (PPCS-NPs) platform can effectively trigger the host immune system and lead to significantly higher antibody titers.
Collapse
Affiliation(s)
- Xiaolei Zhang
- Department
of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
- MicroSep Biological
Science Co. Ltd., Wuxi, Jiangsu 214400, People’s Republic of China
| | - Xia Zhao
- State
Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, People’s Republic of China
| | - Jittima Amie Luckanagul
- Department
of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
- Department
of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, 254 Phayathai Road, Bangkok, 10330, Thailand
| | - Jing Yan
- Department
of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Yuzhe Nie
- Department
of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
- Department
of Life Science, Northeast Forestry University, Harbin, Heilongjiang 150040, People’s Republic of China
| | - L. Andrew Lee
- Department
of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Qian Wang
- Department
of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
| |
Collapse
|