301
|
Fenton OS, Olafson KN, Pillai PS, Mitchell MJ, Langer R. Advances in Biomaterials for Drug Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1705328. [PMID: 29736981 PMCID: PMC6261797 DOI: 10.1002/adma.201705328] [Citation(s) in RCA: 464] [Impact Index Per Article: 77.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 02/12/2018] [Indexed: 04/14/2023]
Abstract
Advances in biomaterials for drug delivery are enabling significant progress in biology and medicine. Multidisciplinary collaborations between physical scientists, engineers, biologists, and clinicians generate innovative strategies and materials to treat a range of diseases. Specifically, recent advances include major breakthroughs in materials for cancer immunotherapy, autoimmune diseases, and genome editing. Here, strategies for the design and implementation of biomaterials for drug delivery are reviewed. A brief history of the biomaterials field is first established, and then commentary on RNA delivery, responsive materials development, and immunomodulation are provided. Current challenges associated with these areas as well as opportunities to address long-standing problems in biology and medicine are discussed throughout.
Collapse
Affiliation(s)
- Owen S Fenton
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | - Katy N Olafson
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | - Padmini S Pillai
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, School of Engineering and Applied Science, Philadelphia, PA, 19104, USA
| | - Robert Langer
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| |
Collapse
|
302
|
Chen W, Zuo H, Li B, Duan C, Rolfe B, Zhang B, Mahony TJ, Xu ZP. Clay Nanoparticles Elicit Long-Term Immune Responses by Forming Biodegradable Depots for Sustained Antigen Stimulation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1704465. [PMID: 29655306 DOI: 10.1002/smll.201704465] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 01/29/2018] [Indexed: 05/21/2023]
Abstract
Nanomaterials have been widely tested as new generation vaccine adjuvants, but few evoke efficient immunoreactions. Clay nanoparticles, for example, layered double hydroxide (LDH) and hectorite (HEC) nanoparticles, have shown their potent adjuvanticity in generating effective and durable immune responses. However, the mechanism by which clay nanoadjuvants stimulate the immune system is not well understood. Here, it is demonstrated that LDH and HEC-antigen complexes form loose agglomerates in culture medium/serum. They also form nodules with loose structures in tissue after subcutaneous injection, where they act as a depot for up to 35 d. More importantly, clay nanoparticles actively and continuously recruit immune cells into the depot for up to one month, and stimulate stronger immune responses than FDA-approved adjuvants, Alum and QuilA. Sustained antigen release is also observed in clay nanoparticle depots, with 50-60% antigen released after 35 d. In contrast, Alum-antigen complexes show minimal antigen release from the depot. Importantly, LDH and HEC are more effective than QuilA and Alum in promoting memory T-cell proliferation. These findings suggest that both clay nanoadjuvants can serve as active vaccine platforms for sustained and potent immune responses.
Collapse
Affiliation(s)
- Weiyu Chen
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, QLD, 4072, Australia
| | - Huali Zuo
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, QLD, 4072, Australia
| | - Bei Li
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, QLD, 4072, Australia
| | - Chengcheng Duan
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, QLD, 4072, Australia
| | - Barbara Rolfe
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, QLD, 4072, Australia
| | - Bing Zhang
- Vaccine Delivery, Animal Science, Agri-Science Queensland, Department of Agriculture & Fisheries, Dutton Park, QLD, 4102, Australia
| | - Timothy J Mahony
- Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Zhi Ping Xu
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, QLD, 4072, Australia
| |
Collapse
|
303
|
Nasr SH, Tonson A, El-Dakdouki MH, Zhu DC, Agnew D, Wiseman R, Qian C, Huang X. Effects of Nanoprobe Morphology on Cellular Binding and Inflammatory Responses: Hyaluronan-Conjugated Magnetic Nanoworms for Magnetic Resonance Imaging of Atherosclerotic Plaques. ACS APPLIED MATERIALS & INTERFACES 2018; 10:11495-11507. [PMID: 29558108 PMCID: PMC5995107 DOI: 10.1021/acsami.7b19708] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Atherosclerosis is an inflammatory disease of arterial walls and the rupturing of atherosclerotic plaques is a major cause of heart attack and stroke. Imaging techniques that can enable the detection of atherosclerotic plaques before clinical manifestation are urgently needed. Magnetic resonance imaging (MRI) is a powerful technique to image the morphology of atherosclerotic plaques. In order to better analyze molecular processes in plaques, contrast agents that can selectively bind to plaque receptors will prove invaluable. CD44 is a cell surface protein overexpressed in plaque tissues, the level of which can be correlated with the risks of plaque rupture. Thus, targeting CD44 is an attractive strategy for detection of atherosclerotic plaques. Herein, we report the synthesis of hyaluronan-conjugated iron oxide nanoworms (HA-NWs). A new purification and gel electrophoresis protocol was developed to ensure the complete removal of free HA from HA-NWs. Compared to the more traditional spherical HA-bearing nanoparticles, HA-NWs had an elongated shape, which interacted much stronger with CD44-expressing cells in CD44- and HA-dependent manners. Furthermore, the HA-NWs did not induce much inflammatory response compared to the spherical HA nanoparticles. When assessed in vivo, HA-NWs enabled successful imaging of atherosclerotic plaques in a clinically relevant model of ApoE knockout transgenic mice for noninvasive plaque detection by MRI. Thus, nanoprobe shape engineering can be a useful strategy to significantly enhance their desired biological properties.
Collapse
Affiliation(s)
| | - Anne Tonson
- Department of Physiology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Mohammad H. El-Dakdouki
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
- Department of Chemistry, Beirut Arab University, P.O. Box 11-5020, Riad El Solh 11072809, Beirut, Lebanon
| | - David C. Zhu
- Department of Radiology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Dalen Agnew
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan 48824, United States
| | - Robert Wiseman
- Department of Physiology, Michigan State University, East Lansing, Michigan 48824, United States
- Department of Radiology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Chunqi Qian
- Department of Radiology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Xuefei Huang
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
- Department of Biomedical Engineering, Michigan State University, East Lansing, Michigan 48824, United States
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
304
|
Wang C, Zhu W, Luo Y, Wang BZ. Gold nanoparticles conjugating recombinant influenza hemagglutinin trimers and flagellin enhanced mucosal cellular immunity. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:1349-1360. [PMID: 29649593 DOI: 10.1016/j.nano.2018.03.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 03/09/2018] [Accepted: 03/31/2018] [Indexed: 01/13/2023]
Abstract
The immunogenicity of subunit vaccines can be augmented by formulating them into nanoparticles. We conjugated recombinant trimetric influenza A/Aichi/2/68(H3N2) hemagglutinin (HA) onto functionalized gold nanoparticle (AuNP) surfaces in a repetitive, oriented configuration. To further improve the immunogenicity, we generated Toll-like receptor 5 (TLR5) agonist flagellin (FliC)-coupled AuNPs as particulate adjuvants. Intranasal immunizations with an AuNP-HA and AuNP-FliC particle mixture elicited strong mucosal and systemic immune responses that protected hosts against lethal influenza challenges. Compared with the AuNP-HA alone group, the addition of AuNP-FliC improved mucosal B cell responses as characterized by elevated influenza specific IgA and IgG levels in nasal, tracheal, and lung washes. AuNP-HA/AuNP-FliC also stimulated antigen-specific interferon-γ (IFN-γ)-secreting CD4+ cell proliferation and induced strong effector CD8+ T cell activation. Our results indicate that intranasal co-delivery of antigen and adjuvant-displaying AuNPs enhanced vaccine efficacy by inducing potent cellular immune responses.
Collapse
Affiliation(s)
- Chao Wang
- Center for Inflammation, Immunity & infection, Georgia State University Institute for Biomedical Sciences, Atlanta, GA, USA
| | - Wandi Zhu
- Center for Inflammation, Immunity & infection, Georgia State University Institute for Biomedical Sciences, Atlanta, GA, USA
| | - Yuan Luo
- Center for Inflammation, Immunity & infection, Georgia State University Institute for Biomedical Sciences, Atlanta, GA, USA
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & infection, Georgia State University Institute for Biomedical Sciences, Atlanta, GA, USA.
| |
Collapse
|
305
|
Leng F, Liu F, Yang Y, Wu Y, Tian W. Strategies on Nanodiagnostics and Nanotherapies of the Three Common Cancers. NANOMATERIALS 2018; 8:nano8040202. [PMID: 29597315 PMCID: PMC5923532 DOI: 10.3390/nano8040202] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 03/18/2018] [Accepted: 03/23/2018] [Indexed: 02/07/2023]
Abstract
The emergence of nanomedicine has enriched the knowledge and strategies of treating diseases, and especially some incurable diseases, such as cancers, acquired immune deficiency syndrome (AIDS), and neurodegenerative diseases. The application of nanoparticles in medicine is in the core of nanomedicine. Nanoparticles can be used in drug delivery for improving the uptake of poorly soluble drugs, targeted delivery to a specific site, and drug bioavailability. Early diagnosis of and targeted therapies for cancers can significantly improve patients' quality of life and extend patients' lives. The advantages of nanoparticles have given them a progressively important role in the nanodiagnosis and nanotherapy of common cancers. To provide a reference for the further application of nanoparticles, this review focuses on the recent development and application of nanoparticles in the early diagnosis and treatment of the three common cancers (lung cancer, liver cancer, and breast cancer) by using quantum dots, magnetic nanoparticles, and gold nanoparticles.
Collapse
Affiliation(s)
- Fan Leng
- Department of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China.
| | - Fang Liu
- Department of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China.
| | - Yongtao Yang
- Department of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China.
| | - Yu Wu
- Department of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China.
| | - Weiqun Tian
- Department of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China.
| |
Collapse
|
306
|
Wu C, Chen H, Wu X, Cong X, Wang L, Wang Y, Yang Y, Li W, Sun T. The influence of tumor-induced immune dysfunction on the immune cell distribution of gold nanoparticles in vivo. Biomater Sci 2018; 5:1531-1536. [PMID: 28589972 DOI: 10.1039/c7bm00335h] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Gold nanoparticles (AuNPs) have been extensively explored as a drug carrier and have been widely used to provide advanced biomedical research tools in diagnostic imaging and therapy for cancer. Although the mononuclear phagocyte system and immune system are known to play the main roles in the clearance of AuNPs during the circulation, the particle distribution within the immune cells under the condition of immune dysfunction caused by tumor growth has not been thoroughly studied. Here, the cellular distribution of Cy5 labeled AuNPs with diameters of 5, 30 and 50 nm is characterized within the immune populations of the blood, spleen and bone marrow from tumor free and tumor bearing mice using flow cytometry. Tumor-associated immune dysfunction was observed in all immune organs and cell lineages, and it changed with tumor growth. Furthermore, the particle cellular distribution significantly changed in the tumor bearing mice compared with the tumor free mice. Finally, the particle distribution in the immune cells was also different at different stages of the tumor. Overall, these results can help inform and influence future AuNP design criteria including the future applications for nanoparticle-mediated cancer therapy.
Collapse
Affiliation(s)
- Chenxi Wu
- The First Hospital of Jilin University, Changchun, Jilin, 130021, China.
| | - Hongmei Chen
- The First Hospital of Jilin University, Changchun, Jilin, 130021, China.
| | - Xuan Wu
- The First Hospital of Jilin University, Changchun, Jilin, 130021, China. and Institute of Immunology, Jilin University, Changchun, Jilin, China
| | - Xiuxiu Cong
- The First Hospital of Jilin University, Changchun, Jilin, 130021, China. and Institute of Immunology, Jilin University, Changchun, Jilin, China
| | - Li Wang
- School of Life Science, University of Science & Technology of China, Hefei, Anhui, China
| | - Yucai Wang
- School of Life Science, University of Science & Technology of China, Hefei, Anhui, China
| | - Yongguang Yang
- The First Hospital of Jilin University, Changchun, Jilin, 130021, China. and Institute of Immunology, Jilin University, Changchun, Jilin, China and Columbia Center for Translational Immunology, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Wei Li
- The First Hospital of Jilin University, Changchun, Jilin, 130021, China.
| | - Tianmeng Sun
- The First Hospital of Jilin University, Changchun, Jilin, 130021, China. and Institute of Immunology, Jilin University, Changchun, Jilin, China
| |
Collapse
|
307
|
Tran TH, Tran TTP, Nguyen HT, Phung CD, Jeong JH, Stenzel MH, Jin SG, Yong CS, Truong DH, Kim JO. Nanoparticles for dendritic cell-based immunotherapy. Int J Pharm 2018; 542:253-265. [PMID: 29555438 DOI: 10.1016/j.ijpharm.2018.03.029] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 03/13/2018] [Accepted: 03/15/2018] [Indexed: 12/19/2022]
Abstract
Crosstalk among immune cells has attracted considerable attention with the advent of immunotherapy as a novel therapeutic approach for challenging diseases, especially cancer, which is the leading cause of mortality worldwide. Dendritic cells-the key antigen-presenting cells-play a pivotal role in immunological response by presenting exogenous epitopes to T cells, which induces the self-defense mechanisms of the body. Furthermore, nanotechnology has provided promising ways for diagnosing and treating cancer in the last decade. The progress in nanoparticle drug carrier development, combined with enhanced understanding of the immune system, has enabled harnessing of anti-tumor immunity. This review focuses on the recent advances in nanotechnology that have improved the therapeutic efficacy of immunotherapies, with emphasis on dendritic cell physiology and its role in presenting antigens and eliciting therapeutic T cell response.
Collapse
Affiliation(s)
- Tuan Hiep Tran
- Department for Management of Science and Technology Development, Ton Duc Thang University, Ho Chi Minh City, Vietnam; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, Vietnam.
| | - Thi Thu Phuong Tran
- The Institute of Molecular Genetics of Montpellier, CNRS, Montpellier, France
| | - Hanh Thuy Nguyen
- College of Pharmacy, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, Republic of Korea
| | - Cao Dai Phung
- College of Pharmacy, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, Republic of Korea
| | - Jee-Heon Jeong
- College of Pharmacy, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, Republic of Korea
| | - Martina H Stenzel
- Centre for Advanced Macromolecular Design, School of Chemistry, University of New South Wales, Kensington, NSW 2052, Australia
| | - Sung Giu Jin
- Department of Pharmaceutical Engineering, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
| | - Chul Soon Yong
- College of Pharmacy, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, Republic of Korea
| | - Duy Hieu Truong
- Institute of Research and Development, Duy Tan University, 03 Quang Trung, Da Nang, Vietnam.
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, Republic of Korea.
| |
Collapse
|
308
|
Pietroiusti A, Stockmann-Juvala H, Lucaroni F, Savolainen K. Nanomaterial exposure, toxicity, and impact on human health. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2018; 10:e1513. [PMID: 29473695 DOI: 10.1002/wnan.1513] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 01/03/2018] [Accepted: 01/16/2018] [Indexed: 12/17/2022]
Abstract
The use of engineered nanomaterials (ENM) has grown after the turn of the 21st century. Also, the production of ENM has globally grown, and exposure of workers especially via the lungs to ENM has increased. This review tackles with effects of ENM on workers' health because occupational environment is the main source of exposure to ENM. Assessment of exposure to ENM is demanding, and today there are no occupational exposure level (OEL) for ENM. This is partly due to challenges of such measurements, and in part to the unknown causality between ENM metrics and effects. There are also marked gaps in systematic knowledge on ENM hazards. Human health surveys of exposed workers, or human field studies have not identified specific effects of ENM linking them with a specific exposure. There is, however, a consensus that material characteristics such as size, and chemistry influence effects of ENM. Available data suggest that multiwalled carbon nanotubes (MWCNT) affect the immunological system and cause inflammation of the lungs, or signs of asthma whereas carbon nanofibers (CNF) may cause interstitial fibrosis. Metallic and metal oxide nanoparticles together with MWCNT induce genotoxicity, and a given type of MWCNT has been identified as a possible human carcinogen. Currently, lack of understanding of mechanisms of effects of ENM renders assessment of hazards and risks of ENM material-by-material a necessity. The so called "omics" approaches utilizing ENM-induced alterations in gene and protein expression may be useful in the development of a new paradigm for ENM hazard and risk assessment. This article is categorized under: Toxicology and Regulatory Issues in Nanomedicine > Toxicology of Nanomaterials.
Collapse
Affiliation(s)
- Antonio Pietroiusti
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | | | - Francesca Lucaroni
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Kai Savolainen
- Work Environment, Finnish Institute of Occupational Health, Helsinki, Finland
| |
Collapse
|
309
|
Assis NRG, Caires AJ, Figueiredo BC, Morais SB, Mambelli FS, Marinho FV, Ladeira LO, Oliveira SC. The use of gold nanorods as a new vaccine platform against schistosomiasis. J Control Release 2018; 275:40-52. [PMID: 29428201 DOI: 10.1016/j.jconrel.2018.02.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 01/24/2018] [Accepted: 02/02/2018] [Indexed: 12/20/2022]
Abstract
Schistosomiasis is an important parasitic disease affecting >207 million people in 76 countries around the world and causing approximately 250,000 deaths per year. At present, the main strategy adopted for the control of schistosomiasis is the use of safe chemotherapy, such as praziquantel. However, the high rates of reinfection after treatment restrict the use of this treatment approach and assume the need for other forms of control such as vaccination. Sm29 is a protein that is localized in the Schistosoma mansoni tegument of adult worms and schistosomula and is considered a powerful vaccine candidate. Because of the chemical, physical and immunological characteristics of nanoparticles, nanocarriers have received increasing attention. In the field of nanotechnology, gold nanorods are considered potential vaccine carriers. In this study, we bound S. mansoni rSm29 protein to gold nanorods either directly or by cysteamine functionalization. When the worm burden was evaluated, the AuNRs-NH2-rSm29 group of immunized mice showed the best protection level (34%). Following AuNRs-NH2-rSm29 immunization, we observed a Th1 immunological response in mice with higher production of IFN-γ, mainly by CD4+ and CD8+ T cells. Furthermore, AuNRs-NH2-rSm29 could activate dendritic cells in vitro, enhancing MHCII and MHCI expression and the production of IL-1β in a NLRP3-, ASC- and Caspase-1-dependent manner. In summary, our findings support the use of nanorods as an immunization strategy in vaccine development against infectious diseases.
Collapse
Affiliation(s)
- Natan R G Assis
- Laboratório de Imunologia de Doenças Infecciosas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), Conselho Nacional de Desenvolvimento Científico e Tecnológico, Ministério de Ciência Tecnologia e Inovação Salvador, Bahia, Brazil
| | - Anderson J Caires
- Center of Nanoscience, Nanotechnology and Innovation - CeNano2I, Department of Metallurgical and Materials Engineering, Federal University of Minas Gerais, Brazil
| | - Bárbara C Figueiredo
- Laboratório de Imunologia de Doenças Infecciosas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), Conselho Nacional de Desenvolvimento Científico e Tecnológico, Ministério de Ciência Tecnologia e Inovação Salvador, Bahia, Brazil
| | - Suellen B Morais
- Laboratório de Imunologia de Doenças Infecciosas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), Conselho Nacional de Desenvolvimento Científico e Tecnológico, Ministério de Ciência Tecnologia e Inovação Salvador, Bahia, Brazil
| | - Fábio S Mambelli
- Laboratório de Imunologia de Doenças Infecciosas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), Conselho Nacional de Desenvolvimento Científico e Tecnológico, Ministério de Ciência Tecnologia e Inovação Salvador, Bahia, Brazil
| | - Fábio V Marinho
- Laboratório de Imunologia de Doenças Infecciosas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), Conselho Nacional de Desenvolvimento Científico e Tecnológico, Ministério de Ciência Tecnologia e Inovação Salvador, Bahia, Brazil
| | - Luís O Ladeira
- Laboratório de Nanomateriais, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Sergio C Oliveira
- Laboratório de Imunologia de Doenças Infecciosas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), Conselho Nacional de Desenvolvimento Científico e Tecnológico, Ministério de Ciência Tecnologia e Inovação Salvador, Bahia, Brazil.
| |
Collapse
|
310
|
Ibrahim KE, Bakhiet AO, Awadalla ME, Khan HA. A priming dose protects against gold nanoparticles-induced proinflammatory cytokines mRNA expression in mice. Nanomedicine (Lond) 2018; 13:313-323. [PMID: 29231780 DOI: 10.2217/nnm-2017-0332] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
AIM To study the effect of priming doses of gold nanoparticles (GNPs) on proinflammatory cytokines in different organs of mice. MATERIALS & METHODS Mice were injected with a single or two doses (priming group) of GNPs (5, 20 and 50 nm) and sacrificed after 1 or 7 days. The mRNA expressions of IL-1β, IL-6 and TNF-α were determined in liver, kidney and spleen. RESULTS A single injection of 5 nm GNPs significantly increased the mRNA expressions of IL-1β and IL-6 in liver, which were normalized on day 7. In spleen, the GNPs of all sizes significantly increased IL-1β and IL-6 mRNA expressions on day 1 that persisted on day 7. The priming dose of GNPs protected the animals against the acute phase induction of IL-1β and IL-6 expressions in liver and spleen. CONCLUSION Primed animals showed protection against GNP-induced acute immune activation suggesting the importance of the priming dose in nanomedicine.
Collapse
Affiliation(s)
- Khalid Elfaki Ibrahim
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Amel Omer Bakhiet
- Deanship of Scientific Research, Sudan University of Science & Technology, Khartoum, Sudan
| | - Maaweya Elaeed Awadalla
- Department of Botany & Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Haseeb Ahmad Khan
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
311
|
Bookstaver ML, Tsai SJ, Bromberg JS, Jewell CM. Improving Vaccine and Immunotherapy Design Using Biomaterials. Trends Immunol 2018; 39:135-150. [PMID: 29249461 PMCID: PMC5914493 DOI: 10.1016/j.it.2017.10.002] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 09/29/2017] [Accepted: 10/03/2017] [Indexed: 12/13/2022]
Abstract
Polymers, lipids, scaffolds, microneedles, and other biomaterials are rapidly emerging as technologies to improve the efficacy of vaccines against infectious disease and immunotherapies for cancer, autoimmunity, and transplantation. New studies are also providing insight into the interactions between these materials and the immune system. This insight can be exploited for more efficient design of vaccines and immunotherapies. Here, we describe recent advances made possible through the unique features of biomaterials, as well as the important questions for further study.
Collapse
Affiliation(s)
- Michelle L Bookstaver
- Fischell Department of Bioengineering, University of Maryland, 8228 Paint Branch Drive, College Park, MD 20742, USA
| | - Shannon J Tsai
- Fischell Department of Bioengineering, University of Maryland, 8228 Paint Branch Drive, College Park, MD 20742, USA
| | - Jonathan S Bromberg
- Department of Surgery, University of Maryland School of Medicine, 29 South Greene Street, Baltimore, MD 21201, USA; Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, 800 West Baltimore Street, Baltimore, MD 21201, USA; Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201, USA; Marlene and Stewart Greenebaum Cancer Center, 22 South Greene Street, Baltimore, MD 21201, USA.
| | - Christopher M Jewell
- Fischell Department of Bioengineering, University of Maryland, 8228 Paint Branch Drive, College Park, MD 20742, USA; Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201, USA; Marlene and Stewart Greenebaum Cancer Center, 22 South Greene Street, Baltimore, MD 21201, USA; United States Department of Veteran Affairs, 10 North Greene Street, Baltimore, MD 21201, USA.
| |
Collapse
|
312
|
Shen Y, Hao T, Ou S, Hu C, Chen L. Applications and perspectives of nanomaterials in novel vaccine development. MEDCHEMCOMM 2018; 9:226-238. [PMID: 30108916 PMCID: PMC6083789 DOI: 10.1039/c7md00158d] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 10/17/2017] [Indexed: 01/22/2023]
Abstract
Vaccines show great potential for both prophylactic and therapeutic use in infections, cancer, and other diseases. With the rapid development of bio-technologies and materials sciences, nanomaterials are playing essential roles in novel vaccine formulations and can boost antigen effectiveness by operating as delivery systems to enhance antigen processing and/or as immune-potentiating adjuvants to induce or potentiate immune responses. The effect of nanoparticles in vaccinology showed enhanced antigen stability and immunogenicity as well as targeted delivery and slow release. However, obstacles remain due to the lack of fundamental knowledge on the detailed molecular working mechanism and in vivo bio-effects of nanoparticles. This review provides a broad overview of the current improvements in nanoparticles in vaccinology. Modern nanoparticle vaccines are classified by the nanoparticles' action based on either delivery system or immune potentiator approaches. The mechanisms of interaction of nanoparticles with the antigens and the immune system are discussed. Nanoparticle vaccines approved for use are also listed. A fundamental understanding of the in vivo bio-distribution and the fate of nanoparticles will accelerate the rational design of new nanoparticles comprising vaccines in the future.
Collapse
Affiliation(s)
- Yingbin Shen
- Department of Food Science and Engineering , School of Science and Engineering , Jinan University , Guangzhou 510632 , Guangdong , China . ; ; ; ; ; ; Tel: +86 138 801 32918
| | - Tianyao Hao
- Department of Food Science and Engineering , School of Science and Engineering , Jinan University , Guangzhou 510632 , Guangdong , China . ; ; ; ; ; ; Tel: +86 138 801 32918
| | - Shiyi Ou
- Department of Food Science and Engineering , School of Science and Engineering , Jinan University , Guangzhou 510632 , Guangdong , China . ; ; ; ; ; ; Tel: +86 138 801 32918
| | - Churan Hu
- Department of Food Science and Engineering , School of Science and Engineering , Jinan University , Guangzhou 510632 , Guangdong , China . ; ; ; ; ; ; Tel: +86 138 801 32918
| | - Long Chen
- Department of Food Science and Engineering , School of Science and Engineering , Jinan University , Guangzhou 510632 , Guangdong , China . ; ; ; ; ; ; Tel: +86 138 801 32918
| |
Collapse
|
313
|
Tazaki T, Tabata K, Ainai A, Ohara Y, Kobayashi S, Ninomiya T, Orba Y, Mitomo H, Nakano T, Hasegawa H, Ijiro K, Sawa H, Suzuki T, Niikura K. Shape-dependent adjuvanticity of nanoparticle-conjugated RNA adjuvants for intranasal inactivated influenza vaccines. RSC Adv 2018; 8:16527-16536. [PMID: 35540526 PMCID: PMC9080258 DOI: 10.1039/c8ra01690a] [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: 02/26/2018] [Accepted: 04/25/2018] [Indexed: 11/21/2022] Open
Abstract
Conjugation with gold nanorods enhanced the adjuvanticity of RNA adjuvant for intranasal inactivated influenza vaccines, providing efficient protection against infection in mice.
Collapse
|
314
|
Zhao J, Stenzel MH. Entry of nanoparticles into cells: the importance of nanoparticle properties. Polym Chem 2018. [DOI: 10.1039/c7py01603d] [Citation(s) in RCA: 228] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Knowledge of the interactions between nanoparticles (NPs) and cell membranes is of great importance for the design of safe and efficient nanomedicines.
Collapse
Affiliation(s)
- Jiacheng Zhao
- Centre for Advanced Macromolecular Design
- The University of New South Wales
- Sydney
- Australia
- School of Chemical Engineering
| | - Martina H. Stenzel
- Centre for Advanced Macromolecular Design
- The University of New South Wales
- Sydney
- Australia
- School of Chemistry
| |
Collapse
|
315
|
Abbaraju PL, Jambhrunkar M, Yang Y, Liu Y, Lu Y, Yu C. Asymmetric mesoporous silica nanoparticles as potent and safe immunoadjuvants provoke high immune responses. Chem Commun (Camb) 2018; 54:2020-2023. [DOI: 10.1039/c8cc00327k] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Asymmetric mesoporous silica nanoparticles with a head–tail structure are potent immunoadjuvants in delivering a peptide antigen, generating higher antibody immune response in mice compared to their symmetric counterparts.
Collapse
Affiliation(s)
- Prasanna Lakshmi Abbaraju
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland
- Brisbane
- Australia
| | - Manasi Jambhrunkar
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland
- Brisbane
- Australia
| | - Yannan Yang
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland
- Brisbane
- Australia
| | - Yang Liu
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland
- Brisbane
- Australia
| | - Yao Lu
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland
- Brisbane
- Australia
| | - Chengzhong Yu
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland
- Brisbane
- Australia
| |
Collapse
|
316
|
Biodegradable Polymeric Nanocarrier-Based Immunotherapy in Hepatitis Vaccination. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1078:303-320. [DOI: 10.1007/978-981-13-0950-2_16] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
317
|
Andrade LM, Cox L, Versiani AF, da Fonseca FG. A growing world of small things: a brief review on the nanostructured vaccines. Future Virol 2017. [DOI: 10.2217/fvl-2017-0086] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Vaccines are the most cost-effective intervention in the management of infectious disease. Much of what we perceive as quality of life is related to a good health status and disease absence, for which vaccines are substantially responsible. Nonetheless, there are many infectious diseases for which no vaccine solution is available. That could be due to limitations of the classic approaches to vaccine development, including inactivated, subunit and attenuated vaccines. Nanostructured immunogens belong to a class of nonclassic vaccines in which nanostructures are loaded with antigen-related molecules. Here, we briefly review important features of the nanostructured vaccines – mainly those based in carbon nanotubes and gold nanorods – and discuss their use to prevent infectious diseases, especially those caused by viruses.
Collapse
Affiliation(s)
- Lídia M Andrade
- Laboratório de Nanomateriais, Departamento de Física, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
- NanoBioMedical Research Group, Departamento de Física, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Laura Cox
- NanoBioMedical Research Group, Departamento de Física, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Campus Pampulha, CEP: 31270–901, Belo Horizonte, MG, Brasil
| | - Alice F Versiani
- NanoBioMedical Research Group, Departamento de Física, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Campus Pampulha, CEP: 31270–901, Belo Horizonte, MG, Brasil
| | - Flávio G da Fonseca
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Campus Pampulha, CEP: 31270–901, Belo Horizonte, MG, Brasil
| |
Collapse
|
318
|
Margulis K, Zhang X, Joubert L, Bruening K, Tassone CJ, Zare RN, Waymouth RM. Formation of Polymeric Nanocubes by Self‐Assembly and Crystallization of Dithiolane‐Containing Triblock Copolymers. Angew Chem Int Ed Engl 2017. [DOI: 10.1002/ange.201709564] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
| | - Xiangyi Zhang
- Department of Chemistry Stanford University Stanford CA 94305 USA
| | | | - Karsten Bruening
- Stanford Synchrotron Radiation Lightsource Stanford University 2575 Sand Hill Road, MS 69 Menlo Park CA 94025 USA
| | - Christopher J. Tassone
- Stanford Synchrotron Radiation Lightsource Stanford University 2575 Sand Hill Road, MS 69 Menlo Park CA 94025 USA
| | - Richard N. Zare
- Department of Chemistry Stanford University Stanford CA 94305 USA
| | | |
Collapse
|
319
|
Margulis K, Zhang X, Joubert L, Bruening K, Tassone CJ, Zare RN, Waymouth RM. Formation of Polymeric Nanocubes by Self‐Assembly and Crystallization of Dithiolane‐Containing Triblock Copolymers. Angew Chem Int Ed Engl 2017; 56:16357-16362. [DOI: 10.1002/anie.201709564] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Indexed: 12/12/2022]
Affiliation(s)
| | - Xiangyi Zhang
- Department of Chemistry Stanford University Stanford CA 94305 USA
| | | | - Karsten Bruening
- Stanford Synchrotron Radiation Lightsource Stanford University 2575 Sand Hill Road, MS 69 Menlo Park CA 94025 USA
| | - Christopher J. Tassone
- Stanford Synchrotron Radiation Lightsource Stanford University 2575 Sand Hill Road, MS 69 Menlo Park CA 94025 USA
| | - Richard N. Zare
- Department of Chemistry Stanford University Stanford CA 94305 USA
| | | |
Collapse
|
320
|
Neek M, Tucker JA, Kim TI, Molino NM, Nelson EL, Wang SW. Co-delivery of human cancer-testis antigens with adjuvant in protein nanoparticles induces higher cell-mediated immune responses. Biomaterials 2017; 156:194-203. [PMID: 29202325 DOI: 10.1016/j.biomaterials.2017.11.022] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 11/07/2017] [Accepted: 11/19/2017] [Indexed: 12/19/2022]
Abstract
Nanoparticles have attracted considerable interest as cancer vaccine delivery vehicles for inducing sufficient CD8+ T cell-mediated immune responses to overcome the low immunogenicity of the tumor microenvironment. Our studies described here are the first to examine the effects of clinically-tested human cancer-testis (CT) peptide epitopes within a synthetic nanoparticle. Specifically, we focused on two significant clinical CT targets, the HLA-A2 restricted epitopes of NY-ESO-1 and MAGE-A3, using a viral-mimetic packaging strategy. Our data shows that simultaneous delivery of a NY-ESO-1 epitope (SLLMWITQV) and CpG using the E2 subunit assembly of pyruvate dehydrogenase (E2 nanoparticle), resulted in a 25-fold increase in specific IFN-γ secretion in HLA-A2 transgenic mice. This translated to a 15-fold increase in lytic activity toward target cancer cells expressing the antigen. Immunization with a MAGE-A3 epitope (FLWGPRALV) delivered with CpG in E2 nanoparticles yielded an increase in specific IFN-γ secretion and cell lysis by 6-fold and 9-fold, respectively. Furthermore, combined delivery of NY-ESO-1 and MAGE-A3 antigens in E2 nanoparticles yielded an additive effect that increased lytic activity towards cells bearing NY-ESO-1+ and MAGE-A3+. Our investigations demonstrate that formulation of CT antigens within a nanoparticle can significantly enhance antigen-specific cell-mediated responses, and the combination of the two antigens in a vaccine can preserve the increased individual responses that are observed for each antigen alone.
Collapse
Affiliation(s)
- Medea Neek
- Department of Chemical Engineering and Materials Science, University of California, Irvine, CA 92697, USA
| | - Jo Anne Tucker
- Department of Medicine, University of California, Irvine, CA 92697, USA
| | - Tae Il Kim
- Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA
| | - Nicholas M Molino
- Department of Chemical Engineering and Materials Science, University of California, Irvine, CA 92697, USA
| | - Edward L Nelson
- Department of Medicine, University of California, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92697, USA; Institute for Immunology, University of California, Irvine, CA 92697, USA
| | - Szu-Wen Wang
- Department of Chemical Engineering and Materials Science, University of California, Irvine, CA 92697, USA; Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92697, USA.
| |
Collapse
|
321
|
Dykman LA, Staroverov SA, Fomin AS, Khanadeev VA, Khlebtsov BN, Bogatyrev VA. Gold nanoparticles as an adjuvant: Influence of size, shape, and technique of combination with CpG on antibody production. Int Immunopharmacol 2017; 54:163-168. [PMID: 29149704 DOI: 10.1016/j.intimp.2017.11.008] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 11/06/2017] [Accepted: 11/07/2017] [Indexed: 01/09/2023]
Abstract
Gold nanoparticles (GNPs) are advantageous as an adjuvant in the design of effective vaccines and in the preparation of high-affinity antibodies to haptens and complete antigens. Another method of activating immunocompetent cells with colloidal gold is to conjugate GNPs with CpG oligodeoxynucleotides (ODNs). We examined how the size and shape of GNPs and various combinations of GNPs and CpG ODNs 1826 affect the immune response. When animals were injected with a model antigen (BSA) coupled to gold nanospheres (diameters, 15 and 50nm), nanorods, nanoshells, and nanostars, the titers of the resultant antibodies differed substantially. The antibody titers decreased in the sequence GNPs-50nm>GNPs-15nm>nanoshells>nanostars>nanorods>native BSA. We conclude that 50 and 15nm gold nanospheres are the optimal antigen carrier and adjuvant for immunization. The highest titer of anti-BSA antibodies was detected in the blood serum of mice immunized simultaneously with BSA-GNP and CpG-GNP conjugates.
Collapse
Affiliation(s)
- Lev A Dykman
- Institute of Biochemistry and Physiology of Plants and Microorganisms, Russian Academy of Sciences, 13 Prospekt Entuziastov, Saratov 410049, Russia; Saratov Science Research Veterinary Institute, Russian Academy of Sciences, 6 Ulitsa 53-ei Strelkovoi Divizii, Saratov 410028, Russia.
| | - Sergey A Staroverov
- Institute of Biochemistry and Physiology of Plants and Microorganisms, Russian Academy of Sciences, 13 Prospekt Entuziastov, Saratov 410049, Russia; Saratov Science Research Veterinary Institute, Russian Academy of Sciences, 6 Ulitsa 53-ei Strelkovoi Divizii, Saratov 410028, Russia
| | - Alexander S Fomin
- Institute of Biochemistry and Physiology of Plants and Microorganisms, Russian Academy of Sciences, 13 Prospekt Entuziastov, Saratov 410049, Russia; Saratov Science Research Veterinary Institute, Russian Academy of Sciences, 6 Ulitsa 53-ei Strelkovoi Divizii, Saratov 410028, Russia
| | - Vitaly A Khanadeev
- Institute of Biochemistry and Physiology of Plants and Microorganisms, Russian Academy of Sciences, 13 Prospekt Entuziastov, Saratov 410049, Russia
| | - Boris N Khlebtsov
- Institute of Biochemistry and Physiology of Plants and Microorganisms, Russian Academy of Sciences, 13 Prospekt Entuziastov, Saratov 410049, Russia
| | - Vladimir A Bogatyrev
- Institute of Biochemistry and Physiology of Plants and Microorganisms, Russian Academy of Sciences, 13 Prospekt Entuziastov, Saratov 410049, Russia
| |
Collapse
|
322
|
Ahmad S, Zamry AA, Tan HTT, Wong KK, Lim J, Mohamud R. Targeting dendritic cells through gold nanoparticles: A review on the cellular uptake and subsequent immunological properties. Mol Immunol 2017; 91:123-133. [DOI: 10.1016/j.molimm.2017.09.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 06/15/2017] [Accepted: 09/01/2017] [Indexed: 02/07/2023]
|
323
|
Song C, Phuengkham H, Kim SY, Lee MS, Jeong JH, Shin SJ, Lim YT. Aminated nanomicelles as a designer vaccine adjuvant to trigger inflammasomes and multiple arms of the innate immune response in lymph nodes. Int J Nanomedicine 2017; 12:7501-7517. [PMID: 29066896 PMCID: PMC5644533 DOI: 10.2147/ijn.s144623] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
In this study, we suggest a designer vaccine adjuvant that can mimic the drainage of pathogens into lymph nodes and activate innate immune response in lymph nodes. By the amination of multivalent carboxyl groups in poly-(γ-glutamic acid) (γ-PGA) nanomicelles, the size was reduced for rapid entry into lymphatic vessels, and the immunologically inert nanomicelles were turned into potential activators of inflammasomes. Aminated γ-PGA nanomicelles (aPNMs) induced NLRP3 inflammasome activation and the subsequent release of proinflammatory IL-1β. The NLRP3-dependent inflammasome induction mechanism was confirmed through enzyme (cathepsin B and caspase-1) inhibitors and NLRP3 knockout mice model. After the aPNMs were combined with a clinically evaluated TLR3 agonist, polyinosinic-polycytidylic acid sodium salt (aPNM-IC), they triggered multiple arms of the innate immune response, including the secretion of pro-inflammatory cytokines by both inflammasomes and an inflammasome-independent pathway and the included type I interferons.
Collapse
Affiliation(s)
- Chanyoung Song
- SKKU Advanced Institute of Nanotechnology, School of Chemical Engineering
| | | | - Sun-Young Kim
- SKKU Advanced Institute of Nanotechnology, School of Chemical Engineering
| | - Min Sang Lee
- Department of Pharmacy, Sungkyunkwan University, Suwon
| | - Ji Hoon Jeong
- Department of Pharmacy, Sungkyunkwan University, Suwon
| | - Sung Jae Shin
- Department of Microbiology and Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yong Taik Lim
- SKKU Advanced Institute of Nanotechnology, School of Chemical Engineering
| |
Collapse
|
324
|
Li P, Song H, Zhang H, Yang P, Zhang C, Huang P, Kong D, Wang W. Engineering biodegradable guanidyl-decorated PEG-PCL nanoparticles as robust exogenous activators of DCs and antigen cross-presentation. NANOSCALE 2017; 9:13413-13418. [PMID: 28876002 DOI: 10.1039/c7nr04470d] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Nanoparticles (NPs)-based adjuvants are attracting much attention in the development of vaccines. Previously, we reported a type of guanidyl-decorated polymeric NPs used as antigen delivery carriers for the first time. However, its un-degradability may restrict potential clinical translation. More importantly, the specific cellular pathway by which dendritic cells (DCs) endocytosed these NPs and the relationship among guanidyl with the antigen cross-presentation, cytokine secretion, and lymph node targeting still remain unclear. Here, we show NPs assembled by biodegradable methoxyl poly(ethylene glycol)-block-poly(ε-caprolactone)-graft-poly(2-(guanidyl) ethyl methacrylate) (mPEG-b-PCL-g-PGEM, PECG) copolymers can robustly activate DCs and promote their maturation; additionally antigen cross-presentation was improved both in vitro and in vivo. Significantly, our results also demonstrate the increase of surface guanidyl on nanoparticles modulates the depot effect and lymph node drainage of PECG NPs-based adjuvants, as well as immune responses, by regulating the secretion of cytokines including IFN-γ and TNF-α. Our study provides insights into the action of guanidyl-decorated nanoscale adjuvants and new adjuvants for vaccines containing protein antigens. We anticipate the strategy of guanidyl decoration to be a starting point for the development of more exciting immunoadjuvants.
Collapse
Affiliation(s)
- Pan Li
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China.
| | | | | | | | | | | | | | | |
Collapse
|
325
|
Schubertová V, Martinez-Veracoechea FJ, Vácha R. Design of Multivalent Inhibitors for Preventing Cellular Uptake. Sci Rep 2017; 7:11689. [PMID: 28916832 PMCID: PMC5601900 DOI: 10.1038/s41598-017-11735-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 08/29/2017] [Indexed: 01/12/2023] Open
Abstract
Cellular entry, the first crucial step of viral infection, can be inhibited by molecules adsorbed on the virus surface. However, apart from using stronger affinity, little is known about the properties of such inhibitors that could increase their effectiveness. Our simulations showed that multivalent inhibitors can be designed to be much more efficient than their monovalent counterparts. For example, for our particular simulation model, a single multivalent inhibitor spanning 5 to 6 binding sites is enough to prevent the uptake compared to the required 1/3 of all the receptor binding sites needed to be blocked by monovalent inhibitors. Interestingly, multivalent inhibitors are more efficient in inhibiting the uptake not only due to their increased affinity but mainly due to the co-localization of the inhibited receptor binding sites at the virion's surface. Furthermore, we show that Janus-like inhibitors do not induce virus aggregation. Our findings may be generalized to other uptake processes including bacteria and drug-delivery.
Collapse
Affiliation(s)
- Veronika Schubertová
- Faculty of Science, Masaryk University, Brno, Czech Republic
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | | | - Robert Vácha
- Faculty of Science, Masaryk University, Brno, Czech Republic.
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic.
| |
Collapse
|
326
|
Jindal AB. The effect of particle shape on cellular interaction and drug delivery applications of micro- and nanoparticles. Int J Pharm 2017; 532:450-465. [PMID: 28917985 DOI: 10.1016/j.ijpharm.2017.09.028] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 09/08/2017] [Accepted: 09/12/2017] [Indexed: 01/04/2023]
Abstract
Encapsulation of therapeutic agents in nanoparticles offers several benefits including improved bioavailability, site specific delivery, reduced toxicity and in vivo stability of proteins and nucleotides over conventional delivery options. These benefits are consequence of distinct in vivo pharmacokinetic and biodistribution profile of nanoparticles, which is dictated by the complex interplay of size, surface charge and surface hydrophobicity. Recently, particle shape has been identified as a new physical parameter which has exerted tremendous impact on cellular uptake and biodistribution, thereby in vivo performance of nanoparticles. Improved therapeutic efficacy of anticancer agents using non-spherical particles is the recent development in the field. Additionally, immunological response of nanoparticles was also altered when antigens were loaded in non-spherical nanovehicles. The apparent impact of particle shape inspired the new research in the field of drug delivery. The present review therefore details the research in this field. The review focuses on methods of fabrication of particles of non-spherical geometries and impact of particle shape on cellular uptake, biodistribution, tumor targeting and production of immunological responses.
Collapse
Affiliation(s)
- Anil B Jindal
- Department of Pharmacy, Birla Institute of Technology and Science (BITS) Pilani,, Pilani Campus,, Rajasthan-333031, India.
| |
Collapse
|
327
|
Hu Y, Chi C, Wang S, Wang L, Liang P, Liu F, Shang W, Wang W, Zhang F, Li S, Shen H, Yu X, Liu H, Tian J. A Comparative Study of Clinical Intervention and Interventional Photothermal Therapy for Pancreatic Cancer. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2017; 29:1700448. [PMID: 28682465 DOI: 10.1002/adma.201700448] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 04/15/2017] [Indexed: 06/07/2023]
Abstract
Although nanoparticle-based photothermal therapy (PTT) has been intensively investigated recently, its comparative efficiency with any clinical cancer treatments has been rarely explored. Herein for the first time we report a systematic comparative study of clinical iodine-125 (125 I) interstitial brachytherapy (IBT-125-I) and interventional PTT (IPTT) in an orthotopic xenograft model of human pancreatic cancer. IPTT, based on the nanoparticles composing of anti-urokinase plasminogen activator receptor (uPAR) antibody, polyethylene glycol (PEG), and indocyanine green (ICG) modified gold nanoshells (hereinafter uIGNs), is directly applied to local pancreatic tumor deep in the abdomen. In comparison to IBT-125-I, a 25% higher median survival rate of IPTT with complete ablation by one-time intervention has been achieved. The IPTT could also inhibit pancreatic tumor metastasis which can be harnessed for effective cancer immunotherapy. All results show that this IPTT is a safe and radical treatment for eradicating tumor cells, and may benefit future clinical pancreatic cancer patients.
Collapse
Affiliation(s)
- Yanyan Hu
- Department of Interventional Ultrasound, General Hospital of People's Liberation Army, Beijing, 100853, China
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Chongwei Chi
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Shunhao Wang
- Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, State Key Laboratory of Chemical Resource Engineering, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lingxiong Wang
- Cancer Center lab, Division of Internal Medicine, General Hospital of People's Liberation Army, Beijing, 100853, China
| | - Ping Liang
- Department of Interventional Ultrasound, General Hospital of People's Liberation Army, Beijing, 100853, China
| | - Fangyi Liu
- Department of Interventional Ultrasound, General Hospital of People's Liberation Army, Beijing, 100853, China
| | - Wenting Shang
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Weiwei Wang
- Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, State Key Laboratory of Chemical Resource Engineering, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Fengrong Zhang
- Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, State Key Laboratory of Chemical Resource Engineering, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Shanshan Li
- Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, State Key Laboratory of Chemical Resource Engineering, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Heyun Shen
- Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, State Key Laboratory of Chemical Resource Engineering, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Xiaoling Yu
- Department of Interventional Ultrasound, General Hospital of People's Liberation Army, Beijing, 100853, China
| | - Huiyu Liu
- Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, State Key Laboratory of Chemical Resource Engineering, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Jie Tian
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
328
|
Yang Y, Jambhrunkar M, Abbaraju PL, Yu M, Zhang M, Yu C. Understanding the Effect of Surface Chemistry of Mesoporous Silica Nanorods on Their Vaccine Adjuvant Potency. Adv Healthc Mater 2017; 6. [PMID: 28557331 DOI: 10.1002/adhm.201700466] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Indexed: 12/17/2022]
Abstract
Mesoporous silica nanoparticles are reported as adjuvants in nanovaccines in generating robust antigen-specific immunity. However, the effect of surface chemistry in initiating and modulating the immune response remains largely unexplored. In this study, mesoporous silica nanorods (MSNRs) are modified with NH2 and C18 groups to investigate the influence of surface functional groups (OH, NH2 , and C18 ) on their adjuvant efficacy. It is found that compared to OH and NH2 groups, the hydrophobic C18 modification significantly enhances antigen uptake by antigen presenting cells and endosomal-lysosomal escape in vitro, dendritic cells, and macrophages maturation ex vivo, and elicits secretion of interferon-γ level and antibody response in immunized mice. Moreover, bare MSNR and MSNRNH2 exhibit T-helper 2 biased immune response, while MSNRC18 shows a T-helper 1 biased immune response. These findings suggest that the surface chemistry of nanostructured adjuvants has profound impact on the immune response, which provides useful guidance for the design of effective nanomaterial based vaccines.
Collapse
Affiliation(s)
- Yannan Yang
- Australian Institute for Bioengineering and Nanotechnology University of Queensland St Lucia QLD 4072 Australia
| | - Manasi Jambhrunkar
- Australian Institute for Bioengineering and Nanotechnology University of Queensland St Lucia QLD 4072 Australia
| | - Prasanna Lakshmi Abbaraju
- Australian Institute for Bioengineering and Nanotechnology University of Queensland St Lucia QLD 4072 Australia
| | - Meihua Yu
- Australian Institute for Bioengineering and Nanotechnology University of Queensland St Lucia QLD 4072 Australia
| | - Min Zhang
- Australian Institute for Bioengineering and Nanotechnology University of Queensland St Lucia QLD 4072 Australia
| | - Chengzhong Yu
- Australian Institute for Bioengineering and Nanotechnology University of Queensland St Lucia QLD 4072 Australia
| |
Collapse
|
329
|
Kinnear C, Moore TL, Rodriguez-Lorenzo L, Rothen-Rutishauser B, Petri-Fink A. Form Follows Function: Nanoparticle Shape and Its Implications for Nanomedicine. Chem Rev 2017; 117:11476-11521. [DOI: 10.1021/acs.chemrev.7b00194] [Citation(s) in RCA: 342] [Impact Index Per Article: 48.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Calum Kinnear
- Bio21 Institute & School of Chemistry, University of Melbourne, Parkville 3010, Australia
| | | | | | | | | |
Collapse
|
330
|
Nicolardi S, van der Burgt YEM, Codée JDC, Wuhrer M, Hokke CH, Chiodo F. Structural Characterization of Biofunctionalized Gold Nanoparticles by Ultrahigh-Resolution Mass Spectrometry. ACS NANO 2017; 11:8257-8264. [PMID: 28686409 PMCID: PMC5616101 DOI: 10.1021/acsnano.7b03402] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 07/07/2017] [Indexed: 05/24/2023]
Abstract
Biofunctionalized gold nanoparticles (AuNPs) enable innovative translational research and development in biomedicine. Biomolecules such as peptides, proteins, lipids, and carbohydrates can be assembled onto AuNPs to yield nanomaterials with unique properties for applications in imaging, photothermal therapy, vaccination strategies, and drug delivery. The characterization of functionalized AuNPs still remains an analytical challenge that normally requires the combination of multiple techniques. Laser desorption/ionization (LDI) and matrix-assisted LDI (MALDI) have been applied successfully in combination with time-of-flight (TOF) mass spectrometry (MS) for the analysis of the surface chemistry of AuNPs functionalized with synthetic ligands, however only for ligands with a molecular mass limited to 1000 Da. TOF-MS-based approaches in addition exhibit limited performance in terms of mass resolution and MS/MS possibilities. To overcome these limitations, we designed an approach for the analysis of AuNPs based on ultrahigh resolution Fourier transform ion cyclotron resonance (FTICR) MS and a combination of LDI and MALDI. To illustrate the performance of the method, we present a comprehensive characterization of the surface chemistry of AuNPs conjugated via a thiol-ending linker to either the ovalbumin peptide (OVA 323-339), the Lewis X antigen (Galβ1-4[Fucα1-3]GlcNAcβ1) trisaccharide, the tetramannoside Manα1-2Manα1-2Manα1-3Manα1, or a mixture of both carbohydrates. Collision-induced dissociation (CID) was used to characterize the structure of pseudomolecular ions generated by LDI/MALDI in-depth. These included [M + H]+ and [M + Na]+, and importantly also [M + Au]+ and [M + 2Au-H]+ ions. This first observation of gold-containing pseudomolecular ions provides direct evidence for the Au-conjugation of ligands. In addition, we show the applicability of the method to monitor proteolytic cleavage of peptides that are conjugated to the AuNP surface. The presented LDI/MALDI-FTICR-MS and MS/MS approach will be applicable to the characterization of a wide range of functionalized AuNPs.
Collapse
Affiliation(s)
- Simone Nicolardi
- Center
for Proteomics and Metabolomics and Department of Parasitology, Leiden University Medical Center, Leiden, 2333 ZA, The Netherlands
| | - Yuri E. M. van der Burgt
- Center
for Proteomics and Metabolomics and Department of Parasitology, Leiden University Medical Center, Leiden, 2333 ZA, The Netherlands
| | - Jeroen D. C. Codée
- Department
of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, 2333 CC, The Netherlands
| | - Manfred Wuhrer
- Center
for Proteomics and Metabolomics and Department of Parasitology, Leiden University Medical Center, Leiden, 2333 ZA, The Netherlands
| | - Cornelis H. Hokke
- Center
for Proteomics and Metabolomics and Department of Parasitology, Leiden University Medical Center, Leiden, 2333 ZA, The Netherlands
| | - Fabrizio Chiodo
- Center
for Proteomics and Metabolomics and Department of Parasitology, Leiden University Medical Center, Leiden, 2333 ZA, The Netherlands
- Department
of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, 2333 CC, The Netherlands
| |
Collapse
|
331
|
Wang C, Ye Y, Hu Q, Bellotti A, Gu Z. Tailoring Biomaterials for Cancer Immunotherapy: Emerging Trends and Future Outlook. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2017; 29. [PMID: 28556553 DOI: 10.1002/adma.201606036] [Citation(s) in RCA: 194] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 03/04/2017] [Indexed: 05/05/2023]
Abstract
Cancer immunotherapy, as a paradigm shift in cancer treatment, has recently received tremendous attention. The active cancer vaccination, immune checkpoint blockage (ICB) and chimeric antigen receptor (CAR) for T-cell-based adoptive cell transfer are among these developments that have achieved a significant increase in patient survival in clinical trials. Despite these advancements, emerging research at the interdisciplinary interface of cancer biology, immunology, bioengineering, and materials science is important to further enhance the therapeutic benefits and reduce side effects. Here, an overview of the latest studies on engineering biomaterials for the enhancement of anticancer immunity is given, including the perspectives of delivery of immunomodulatory therapeutics, engineering immune cells, and constructing immune-modulating scaffolds. The opportunities and challenges in this field are also discussed.
Collapse
Affiliation(s)
- Chao Wang
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27695, USA
- Division of Pharmacoengineering and Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Yanqi Ye
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27695, USA
- Division of Pharmacoengineering and Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Quanyin Hu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27695, USA
- Division of Pharmacoengineering and Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Adriano Bellotti
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27695, USA
- Department of Medicine University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Zhen Gu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27695, USA
- Division of Pharmacoengineering and Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Medicine University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| |
Collapse
|
332
|
Ding P, Zhang T, Li Y, Teng M, Sun Y, Liu X, Chai S, Zhou E, Jin Q, Zhang G. Nanoparticle orientationally displayed antigen epitopes improve neutralizing antibody level in a model of porcine circovirus type 2. Int J Nanomedicine 2017; 12:5239-5254. [PMID: 28769561 PMCID: PMC5533572 DOI: 10.2147/ijn.s140789] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Recent advancements in biotechnology have enabled the rapid identification and subsequent expression of pathogenic microbial major antigens that induce protective immune responses. However, subunit vaccines have not been successfully commercialized mainly due to the lack of sufficient levels of neutralizing antibodies (NAs). High levels of NA rely on the efficient recognition and cross-linking of multiple neutralizing epitopes with B-cell receptors (BCRs). Nanoparticles are able to display coupled antigenic arrays at high density and provide multiple binding molecular scenarios with BCRs. The high-resolution antigenic structure makes it possible to accurately display stable neutralizing epitopes. Therefore, the development of a nanovaccine that orientationally displays neutralizing epitopes is a feasible strategy. To address this hypothesis, the capsid (Cap) protein of porcine circovirus type 2 as model antigen was conjugated to gold nanoparticles (AuNPs) through direct reaction of the mercapto group of the unique cysteines with AuNPs, rendering Cap-AuNPs to have neutralizing epitopes on outer surface and an immunodominant epitope buried within the inner surface. In vitro studies showed that AuNPs promoted the phagocytosis of Cap protein and NA levels were significantly improved, meanwhile antibody levels against the immunodominant epitope was significantly reduced. In mouse studies, Cap-AuNP-immunized mice displayed a high production of interleukin (IL)-4, IL-10, and interferon-γ, suggesting that Cap-AuNPs can effectively activate CD4+ and CD8+ T cells and balance Th1 and Th2 cellular responses. This study presents a new vaccine design strategy based on antigen structure, where nanoparticles are coupled to antigens in well-ordered arrays and orientationally display neutralizing epitopes to enhance NA levels.
Collapse
Affiliation(s)
- Peiyang Ding
- College of Veterinary Medicine, Northwest A&F University, Yangling.,Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences
| | - Teng Zhang
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences.,College of Life Sciences, Henan Agricultural University
| | - Yafei Li
- College of Veterinary Medicine, Northwest A&F University, Yangling.,Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences
| | - Man Teng
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences
| | - Yaning Sun
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences
| | - Xiao Liu
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences.,College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou
| | - Shujun Chai
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences
| | - Enmin Zhou
- College of Veterinary Medicine, Northwest A&F University, Yangling
| | - Qianyue Jin
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, People's Republic of China
| | - Gaiping Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling.,Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences.,College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, People's Republic of China
| |
Collapse
|
333
|
Li X, Hufnagel S, Xu H, Valdes SA, Thakkar SG, Cui Z, Celio H. Aluminum (Oxy)Hydroxide Nanosticks Synthesized in Bicontinuous Reverse Microemulsion Have Potent Vaccine Adjuvant Activity. ACS APPLIED MATERIALS & INTERFACES 2017; 9:22893-22901. [PMID: 28621928 PMCID: PMC5556926 DOI: 10.1021/acsami.7b03965] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Insoluble aluminum salts such as aluminum (oxy)hydroxide are commonly used as vaccine adjuvants. Recently, there is evidence suggesting that the adjuvant activity of aluminum salt-based materials is tightly related to their physicochemical properties, including nanometer-scale size, shape with long aspect ratio, and low degree of crystallinity. Herein, for the first time, the bicontinuous reverse microemulsion (RM) technique was utilized to synthesize stick-like monodisperse aluminum (oxy)hydroxide nanoparticles with a long aspect ratio of ∼10, length of ∼80 nm, and low degree of crystallinity (denoted as Al-nanosticks). Moreover, the relationship between the physicochemical properties of Al-nanosticks and the bicontinuous RM was discussed. Compared to the commercial Alhydrogel, which contains micrometer-scale aluminum oxyhydroxide particular aggregates with moderate degree of crystallinity, the Al-nanosticks are more effective in adsorbing and delivering antigens (e.g., ovalbumin, OVA) into antigen-presenting cells, activating inflammasomes, and potentiating OVA-specific antibody responses in a mouse model. It is concluded that the aluminum (oxy)hydroxide nanosticks synthesized in the bicontinuous RM are promising new aluminum salt-based vaccine adjuvants.
Collapse
Affiliation(s)
- Xu Li
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, Texas, U.S.A
| | - Stephanie Hufnagel
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, Texas, U.S.A
| | - Haiyue Xu
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, Texas, U.S.A
| | - Solange A. Valdes
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, Texas, U.S.A
| | - Sachin G. Thakkar
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, Texas, U.S.A
| | - Zhengrong Cui
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, Texas, U.S.A
- Inner Mongolia Medical University, Inner Mongolia Key Laboratory of Molecular Biology, Hohhot, Inner Mongolia, China
| | - Hugo Celio
- The University of Texas at Austin, Texas Materials Institute, Austin, Texas, U.S.A
| |
Collapse
|
334
|
Wang C, Zhu W, Wang BZ. Dual-linker gold nanoparticles as adjuvanting carriers for multivalent display of recombinant influenza hemagglutinin trimers and flagellin improve the immunological responses in vivo and in vitro. Int J Nanomedicine 2017; 12:4747-4762. [PMID: 28740382 PMCID: PMC5503497 DOI: 10.2147/ijn.s137222] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Vaccination is the most cost-effective means of infectious disease control. Although current influenza vaccines are effective in battling closely matched strains, such vaccines have major limitations such as the requirement to produce new vaccines every season, an egg-dependent production system, long production periods, uncertainty in matching the vaccine to circulating strains, and the inability to react to new influenza pandemics resulting from genetic drift or shift. To overcome the intrinsic limitations of the conventional influenza vaccine, we have designed dual-linker gold nanoparticles (AuNPs) conjugated with both recombinant trimetric A/Aichi/2/68 (H3N2), hemagglutinin (HA) and TLR5 agonist flagellin (FliC) as a novel vaccine approach. Click chemistry and metal-chelating reactions were used to couple the two proteins. The conjugated proteins were found to possess high coupling specificity, high stability in harsh environments, high conjugation efficiency, and the ability to keep the appropriate protein conformations for immunogenicity and immunostimulation. Both AuNPs-HA/FliC and AuNPs-HA formulations induced higher levels of antibody responses than a mixture of soluble HA and FliC proteins when administered via a single intranasal immunization in mice. To further investigate the adjuvancy of these nanoparticles, in vitro experiments were conducted in both the JAWS II dendritic cell (DC) line and bone marrow-derived DC (BMDC) models. The results showed that dual-conjugated AuNPs were rapidly targeted and taken up by DCs. Consequently, DCs were induced toward maturation, as demonstrated by high levels of cytokine secretions and membrane costimulatory molecule expression. T cell proliferation was observed when splenic T cells were cocultured with AuNPs-HA/FliC-primed BMDCs. These results suggest that dual-conjugated AuNPs are effective at simultaneously displaying antigens and adjuvants in an oriented, multivalent format and can promote a strong immune response by activating DCs and T cells.
Collapse
Affiliation(s)
- Chao Wang
- Center for Inflammation, Immunity and Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, GA, USA
| | - Wandi Zhu
- Center for Inflammation, Immunity and Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, GA, USA
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity and Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, GA, USA
| |
Collapse
|
335
|
Ainai A, Suzuki T, Tamura SI, Hasegawa H. Intranasal Administration of Whole Inactivated Influenza Virus Vaccine as a Promising Influenza Vaccine Candidate. Viral Immunol 2017. [PMID: 28650274 DOI: 10.1089/vim.2017.0022] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The effect of the current influenza vaccine, an inactivated virus vaccine administered by subcutaneous/intramuscular injection, is limited to reducing the morbidity and mortality associated with seasonal influenza outbreaks. Intranasal vaccination, by contrast, mimics natural infection and induces not only systemic IgG antibodies but also local secretory IgA (S-IgA) antibodies found on the surface of the mucosal epithelium in the upper respiratory tract. S-IgA antibodies are highly effective at preventing virus infection. Although the live attenuated influenza vaccine (LAIV) administered intranasally can induce local antibodies, this vaccine is restricted to healthy populations aged 2-49 years because of safety concerns associated with using live viruses in a vaccine. Instead of LAIV, an intranasal vaccine made with inactivated virus could be applied to high-risk populations, including infants and elderly adults. Normally, a mucosal adjuvant would be required to enhance the effect of intranasal vaccination with an inactivated influenza vaccine. However, we found that intranasal administration of a concentrated, whole inactivated influenza virus vaccine without any mucosal adjuvant was enough to induce local neutralizing S-IgA antibodies in the nasal epithelium of healthy individuals with some immunological memory for seasonal influenza viruses. This intranasal vaccine is a novel candidate that could improve on the current injectable vaccine or the LAIV for the prevention of seasonal influenza epidemics.
Collapse
Affiliation(s)
- Akira Ainai
- Department of Pathology, National Institute of Infectious Diseases , Tokyo, Japan
| | - Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases , Tokyo, Japan
| | - Shin-Ichi Tamura
- Department of Pathology, National Institute of Infectious Diseases , Tokyo, Japan
| | - Hideki Hasegawa
- Department of Pathology, National Institute of Infectious Diseases , Tokyo, Japan
| |
Collapse
|
336
|
Chattopadhyay S, Chen JY, Chen HW, Hu CMJ. Nanoparticle Vaccines Adopting Virus-like Features for Enhanced Immune Potentiation. Nanotheranostics 2017; 1:244-260. [PMID: 29071191 PMCID: PMC5646730 DOI: 10.7150/ntno.19796] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 04/17/2017] [Indexed: 12/22/2022] Open
Abstract
Synthetic nanoparticles play an increasingly significant role in vaccine design and development as many nanoparticle vaccines show improved safety and efficacy over conventional formulations. These nanoformulations are structurally similar to viruses, which are nanoscale pathogenic organisms that have served as a key selective pressure driving the evolution of our immune system. As a result, mechanisms behind the benefits of nanoparticle vaccines can often find analogue to the interaction dynamics between the immune system and viruses. This review covers the advances in vaccine nanotechnology with a perspective on the advantages of virus mimicry towards immune potentiation. It provides an overview to the different types of nanomaterials utilized for nanoparticle vaccine development, including functionalization strategies that bestow nanoparticles with virus-like features. As understanding of human immunity and vaccine mechanisms continue to evolve, recognizing the fundamental semblance between synthetic nanoparticles and viruses may offer an explanation for the superiority of nanoparticle vaccines over conventional vaccines and may spur new design rationales for future vaccine research. These nanoformulations are poised to provide solutions towards pressing and emerging human diseases.
Collapse
Affiliation(s)
- Saborni Chattopadhyay
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan
| | - Jui-Yi Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hui-Wen Chen
- Department of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
- Research Center for Nanotechnology and Infectious Diseases, Taipei, Taiwan
| | - Che-Ming Jack Hu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Research Center for Nanotechnology and Infectious Diseases, Taipei, Taiwan
| |
Collapse
|
337
|
Nanoparticles for tumor immunotherapy. Eur J Pharm Biopharm 2017; 115:243-256. [DOI: 10.1016/j.ejpb.2017.03.013] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Revised: 03/01/2017] [Accepted: 03/17/2017] [Indexed: 12/21/2022]
|
338
|
Effect of halloysite nanotubes on the structure and function of important multiple blood components. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 75:72-78. [DOI: 10.1016/j.msec.2017.02.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 11/29/2016] [Accepted: 02/06/2017] [Indexed: 11/17/2022]
|
339
|
Andorko JI, Jewell CM. Designing biomaterials with immunomodulatory properties for tissue engineering and regenerative medicine. Bioeng Transl Med 2017; 2:139-155. [PMID: 28932817 PMCID: PMC5579731 DOI: 10.1002/btm2.10063] [Citation(s) in RCA: 137] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 04/14/2017] [Accepted: 04/24/2017] [Indexed: 12/29/2022] Open
Abstract
Recent research in the vaccine and immunotherapy fields has revealed that biomaterials have the ability to activate immune pathways, even in the absence of other immune-stimulating signals. Intriguingly, new studies reveal these responses are influenced by the physicochemical properties of the material. Nearly all of this work has been done in the vaccine and immunotherapy fields, but there is tremendous opportunity to apply this same knowledge to tissue engineering and regenerative medicine. This review discusses recent findings that reveal how material properties-size, shape, chemical functionality-impact immune response, and links these changes to emerging opportunities in tissue engineering and regenerative medicine. We begin by discussing what has been learned from studies conducted in the contexts of vaccines and immunotherapies. Next, research is highlighted that elucidates the properties of materials that polarize innate immune cells, including macrophages and dendritic cells, toward either inflammatory or wound healing phenotypes. We also discuss recent studies demonstrating that scaffolds used in tissue engineering applications can influence cells of the adaptive immune system-B and T cell lymphocytes-to promote regenerative tissue microenvironments. Through greater study of the intrinsic immunogenic features of implantable materials and scaffolds, new translational opportunities will arise to better control tissue engineering and regenerative medicine applications.
Collapse
Affiliation(s)
- James I. Andorko
- Fischell Department of BioengineeringUniversity of MarylandCollege ParkMD 20742
| | - Christopher M. Jewell
- Fischell Department of BioengineeringUniversity of MarylandCollege ParkMD 20742
- Department of Microbiology and ImmunologyUniversity of Maryland Medical SchoolBaltimoreMD 21201
- Marlene and Stewart Greenebaum Cancer CenterBaltimoreMD 21201
- United States Department of Veterans AffairsBaltimoreMD 21201
| |
Collapse
|
340
|
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: 12.4] [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]
|
341
|
Compostella F, Pitirollo O, Silvestri A, Polito L. Glyco-gold nanoparticles: synthesis and applications. Beilstein J Org Chem 2017; 13:1008-1021. [PMID: 28684980 PMCID: PMC5480336 DOI: 10.3762/bjoc.13.100] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 05/05/2017] [Indexed: 01/15/2023] Open
Abstract
Glyco-gold nanoparticles combine in a single entity the peculiar properties of gold nanoparticles with the biological activity of carbohydrates. The result is an exciting nanosystem, able to mimic the natural multivalent presentation of saccharide moieties and to exploit the peculiar optical properties of the metallic core. In this review, we present recent advances on glyco-gold nanoparticle applications in different biological fields, highlighting the key parameters which inspire the glyco nanoparticle design.
Collapse
Affiliation(s)
- Federica Compostella
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Saldini 50, 20133 Milan, Italy
| | - Olimpia Pitirollo
- Department of Chemistry, University of Milan, Via C. Golgi 19, 20133 Milan, Italy
| | - Alessandro Silvestri
- Department of Chemistry, University of Milan, Via C. Golgi 19, 20133 Milan, Italy
- CNR – ISTM, Nanotechnology Lab., Via G. Fantoli 16/15, 20138 Milan, Italy
| | - Laura Polito
- CNR – ISTM, Nanotechnology Lab., Via G. Fantoli 16/15, 20138 Milan, Italy
| |
Collapse
|
342
|
Carabineiro SAC. Applications of Gold Nanoparticles in Nanomedicine: Recent Advances in Vaccines. Molecules 2017; 22:E857. [PMID: 28531163 PMCID: PMC6154615 DOI: 10.3390/molecules22050857] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 05/17/2017] [Accepted: 05/19/2017] [Indexed: 11/20/2022] Open
Abstract
Nowadays, gold is used in (nano-)medicine, usually in the form of nanoparticles, due to the solid proofs given of its therapeutic effects on several diseases. Gold also plays an important role in the vaccine field as an adjuvant and a carrier, reducing toxicity, enhancing immunogenic activity, and providing stability in storage. An even brighter golden future is expected for gold applications in this area.
Collapse
Affiliation(s)
- Sónia Alexandra Correia Carabineiro
- Laboratório de Catálise e Materiais (LCM), Laboratório Associado LSRE-LCM, Faculdade de Engenharia, Universidade do Porto, 4200-465 Porto, Portugal.
| |
Collapse
|
343
|
Abbaraju PL, Meka AK, Song H, Yang Y, Jambhrunkar M, Zhang J, Xu C, Yu M, Yu C. Asymmetric Silica Nanoparticles with Tunable Head-Tail Structures Enhance Hemocompatibility and Maturation of Immune Cells. J Am Chem Soc 2017; 139:6321-6328. [PMID: 28440642 DOI: 10.1021/jacs.6b12622] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Asymmetric mesoporous silica nanoparticles (MSNs) with controllable head-tail structures have been successfully synthesized. The head particle type is tunable (solid or porous), and the tail has dendritic large pores. The tail length and tail coverage on head particles are adjustable. Compared to spherical silica nanoparticles with a solid structure (Stöber spheres) or large-pore symmetrical MSNs with fully covered tails, asymmetrical head-tail MSNs (HTMSNs) show superior hemocompatibility due to reduced membrane deformation of red blood cells and decreased level of reactive oxygen species. Moreover, compared to Stöber spheres, asymmetrical HTMSNs exhibit a higher level of uptake and in vitro maturation of immune cells including dendritic cells and macrophage. This study has provided a new family of nanocarriers with potential applications in vaccine development and immunotherapy.
Collapse
Affiliation(s)
- Prasanna Lakshmi Abbaraju
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland , Brisbane, QLD 4072, Australia
| | - Anand Kumar Meka
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland , Brisbane, QLD 4072, Australia
| | - Hao Song
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland , Brisbane, QLD 4072, Australia
| | - Yannan Yang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland , Brisbane, QLD 4072, Australia
| | - Manasi Jambhrunkar
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland , Brisbane, QLD 4072, Australia
| | - Jun Zhang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland , Brisbane, QLD 4072, Australia
| | - Chun Xu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland , Brisbane, QLD 4072, Australia
| | - Meihua Yu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland , Brisbane, QLD 4072, Australia
| | - Chengzhong Yu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland , Brisbane, QLD 4072, Australia
| |
Collapse
|
344
|
Hu X, Wu T, Bao Y, Zhang Z. Nanotechnology based therapeutic modality to boost anti-tumor immunity and collapse tumor defense. J Control Release 2017; 256:26-45. [PMID: 28434891 DOI: 10.1016/j.jconrel.2017.04.026] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 04/15/2017] [Accepted: 04/18/2017] [Indexed: 12/19/2022]
Abstract
Cancer is still the leading cause of death. While traditional treatments such as surgery, chemotherapy and radiotherapy play dominating roles, recent breakthroughs in cancer immunotherapy indicate that the influence of immune system on cancer development is virtually beyond our expectation. Manipulating the immune system to fight against cancer has been thriving in recent years. Further understanding of tumor anatomy provides opportunities to put a brake on immunosuppression by overcoming tumor intrinsic resistance or modulating tumor microenvironment. Nanotechnology which provides versatile engineered approaches to enhance therapeutic effects may potentially contribute to the development of future cancer treatment modality. In this review, we will focus on the application of nanotechnology both in boosting anti-tumor immunity and collapsing tumor defense.
Collapse
Affiliation(s)
| | | | - Yuling Bao
- Tongji School of Pharmacy, PR China; Department of Pharmacy, Tongji Hospital, PR China
| | - Zhiping Zhang
- Tongji School of Pharmacy, PR China; National Engineering Research Center for Nanomedicine, PR China; Hubei Engineering Research Center for Novel Drug Delivery System, HuaZhong University of Science and Technology, Wuhan 430030, PR China.
| |
Collapse
|
345
|
Ke S, Zhou T, Yang P, Wang Y, Zhang P, Chen K, Ren L, Ye S. Gold nanoparticles enhance TRAIL sensitivity through Drp1-mediated apoptotic and autophagic mitochondrial fission in NSCLC cells. Int J Nanomedicine 2017; 12:2531-2551. [PMID: 28408823 PMCID: PMC5383076 DOI: 10.2147/ijn.s129274] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Although tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and its agonistic receptors have been identified as highly promising antitumor agents preferentially eliminating cancer cells with minimal damage, the emergence of TRAIL resistance in most cancers may contribute to therapeutic failure. Thus, there is an urgent need for new approaches to overcome TRAIL resistance. Gold nanoparticles (AuNPs) are one of the most promising nanomaterials that show immense antitumor potential via targeting various cellular and molecular processes; however, the effects of AuNPs on TRAIL sensitivity in cancer cells remain unclear. In this study, we found that AuNPs combined with TRAIL exhibited a greater potency in promoting apoptosis in non-small-cell lung cancer (NSCLC) cells compared with TRAIL alone, suggesting that AuNPs sensitize cancer cells to TRAIL. Further experiments demonstrated that the combination of TRAIL and AuNPs was more effective in causing excessive mitochondrial fragmentation in cancer cells accompanied by a dramatic increase in mitochondrial recruitment of dynamin-related protein 1 (Drp1), mitochondrial dysfunctions, and enhancement of autophagy induction. Small interfering RNA (siRNA) silencing of Drp1 or inhibition of autophagy could effectively alleviate apoptosis in cells exposed to TRAIL combined with AuNPs. In vivo studies revealed that AuNPs augmented TRAIL sensitivity in tumor-bearing mice. Our data indicated that AuNPs potentiate apoptotic response to TRAIL in NSCLC cells through Drp1-dependent mitochondrial fission, and TRAIL combined with AuNPs can be a potential chemotherapeutic strategy for the treatment of NSCLC.
Collapse
Affiliation(s)
- Sunkui Ke
- Department of Thoracic Surgery, Zhongshan Hospital of Xiamen University
| | - Tong Zhou
- Department of Biomaterials, College of Materials, Xiamen University
| | - Peiyan Yang
- Department of Surgery, First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, People’s Republic of China
| | - Yange Wang
- Department of Biomaterials, College of Materials, Xiamen University
| | - Peng Zhang
- Department of Biomaterials, College of Materials, Xiamen University
| | - Keman Chen
- Department of Biomaterials, College of Materials, Xiamen University
| | - Lei Ren
- Department of Biomaterials, College of Materials, Xiamen University
| | - Shefang Ye
- Department of Biomaterials, College of Materials, Xiamen University
| |
Collapse
|
346
|
Marques Neto LM, Kipnis A, Junqueira-Kipnis AP. Role of Metallic Nanoparticles in Vaccinology: Implications for Infectious Disease Vaccine Development. Front Immunol 2017; 8:239. [PMID: 28337198 PMCID: PMC5340775 DOI: 10.3389/fimmu.2017.00239] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 02/20/2017] [Indexed: 02/04/2023] Open
Abstract
Subunit vaccines are safer but less immunogenic than live-attenuated vaccines or whole cell inactivated vaccines. Adjuvants are used to enhance and modulate antigen (Ag) immunogenicity, aiming to induce a protective and long-lasting immune response. Several molecules and formulations have been studied for their adjuvanticity, but only seven have been approved to formulate human vaccines. Metallic nanoparticles (MeNPs), particularly those containing gold and iron oxides, are widely used in medicine for diagnosis and therapy and have been used as carriers for drugs and vaccines. However, little is known about the immune response elicited by MeNPs or about their importance in the development of new vaccines. There is evidence that these particles display adjuvant characteristics, promoting cell recruitment, antigen-presenting cell activation, cytokine production, and inducing a humoral immune response. This review focuses on the characteristics of MeNPs that could facilitate the induction of a cellular immune response, particularly T-helper 1 and T-helper 17, and their potential functions as adjuvants for subunit vaccines.
Collapse
Affiliation(s)
- Lázaro Moreira Marques Neto
- Department of Microbiology, Immunology, Pathology and Parasitology, Institute of Tropical Pathology and Public Health, Federal University of Goiás , Goiânia, Goiás , Brazil
| | - André Kipnis
- Department of Microbiology, Immunology, Pathology and Parasitology, Institute of Tropical Pathology and Public Health, Federal University of Goiás , Goiânia, Goiás , Brazil
| | - Ana Paula Junqueira-Kipnis
- Department of Microbiology, Immunology, Pathology and Parasitology, Institute of Tropical Pathology and Public Health, Federal University of Goiás , Goiânia, Goiás , Brazil
| |
Collapse
|
347
|
Dykman LA, Khlebtsov NG. Immunological properties of gold nanoparticles. Chem Sci 2017; 8:1719-1735. [PMID: 28451297 PMCID: PMC5396510 DOI: 10.1039/c6sc03631g] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Accepted: 11/14/2016] [Indexed: 12/14/2022] Open
Abstract
In the past decade, gold nanoparticles have attracted strong interest from the nanobiotechnological community owing to the significant progress made in robust and easy-to-make synthesis technologies, in surface functionalization, and in promising biomedical applications. These include bioimaging, gene diagnostics, analytical sensing, photothermal treatment of tumors, and targeted delivery of various biomolecular and chemical cargos. For the last-named application, gold nanoparticles should be properly fabricated to deliver the cargo into the targeted cells through effective endocytosis. In this review, we discuss recent progress in understanding the selective penetration of gold nanoparticles into immune cells. The interaction of gold nanoparticles with immune cell receptors is discussed. As distinct from other published reviews, we present a summary of the immunological properties of gold nanoparticles. This review also summarizes what is known about the application of gold nanoparticles as an antigen carrier and adjuvant in immunization for the preparation of antibodies in vivo. For each of the above topics, the basic principles, recent advances, and current challenges are discussed. Thus, this review presents a detailed analysis of data on interaction of gold nanoparticles with immune cells. Emphasis is placed on the systematization of data over production of antibodies by using gold nanoparticles and adjuvant properties of gold nanoparticles. Specifically, we start our discussion with current data on interaction of various gold nanoparticles with immune cells. The next section describes existing technologies to improve production of antibodies in vivo by using gold nanoparticles conjugated with specific ligands. Finally, we describe what is known about adjuvant properties of bare gold or functionalized nanoparticles. In the Conclusion section, we present a short summary of reported data and some challenges and perspectives.
Collapse
Affiliation(s)
- Lev A Dykman
- Institute of Biochemistry and Physiology of Plants and Microorganisms , Russian Academy of Sciences , 13 Prospekt Entuziastov , Saratov 410049 , Russia . ;
| | - Nikolai G Khlebtsov
- Institute of Biochemistry and Physiology of Plants and Microorganisms , Russian Academy of Sciences , 13 Prospekt Entuziastov , Saratov 410049 , Russia . ;
- Saratov National Research State University , 83 Ulitsa Astrakhanskaya , Saratov 410012 , Russia
| |
Collapse
|
348
|
Gomes AC, Mohsen M, Bachmann MF. Harnessing Nanoparticles for Immunomodulation and Vaccines. Vaccines (Basel) 2017; 5:E6. [PMID: 28216554 PMCID: PMC5371742 DOI: 10.3390/vaccines5010006] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 01/23/2017] [Accepted: 01/25/2017] [Indexed: 12/13/2022] Open
Abstract
The first successful use of nanoparticles (NPs) for vaccination was reported almost 40 years ago with a virus-like particle-based vaccine against Hepatitis B. Since then, the term NP has been expanded to accommodate a large number of novel nano-sized particles engineered from a range of materials. The great interest in NPs is likely not only a result of the two successful vaccines against hepatitis B and Human Papilloma Virus (HPV) that use this technology, but also due to the versatility of those small-sized particles, as indicated by the wide range of applications reported so far, ranging from medicinal and cosmetics to purely technical applications. In this review, we will focus on the use of NPs, especially virus-like particles (VLPs), in the field of vaccines and will discuss their employment as vaccines, antigen display platforms, adjuvants and drug delivery systems.
Collapse
Affiliation(s)
- Ariane C Gomes
- The Jenner Institute, Oxford University, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK.
| | - Mona Mohsen
- The Jenner Institute, Oxford University, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK.
| | - Martin F Bachmann
- The Jenner Institute, Oxford University, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK.
- Inselspital, Universitatsspital, Sahlihaus 1, 3010 Bern, Switzerland.
| |
Collapse
|
349
|
Draz MS, Wang YJ, Chen FF, Xu Y, Shafiee H. Electrically Oscillating Plasmonic Nanoparticles for Enhanced DNA Vaccination against Hepatitis C Virus. ADVANCED FUNCTIONAL MATERIALS 2017; 27:1604139. [PMID: 29180949 PMCID: PMC5701658 DOI: 10.1002/adfm.201604139] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
The promise of DNA vaccines is far-reaching. However, the development of potent immunization methods remains a key challenge for its use in clinical applications. Here, an approach for in vivo DNA vaccination by electrically activated plasmonic Au nanoparticles is reported. The electrical excitation of plasmonic nanoparticles can drive vibrational and dipole-like oscillations that are able to disrupt nearby cell membranes. In combination with their intrinsic ability to focus and magnify the electric field on the surface of cells, Au nanoparticles allow enhanced cell poration and facilitate the uptake of DNA vaccine. Mice immunized with this approach showed up to 100-fold higher gene expression compared to control treatments (without nanoparticles) and exhibited significantly increased levels of both antibody and cellular immune responses against a model hepatitis C virus DNA vaccine. This approach can be tuned to establish controlled and targeted delivery of different types of therapeutic molecules into cells and live animals as well.
Collapse
Affiliation(s)
- Mohamed Shehata Draz
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA. Faculty of Science Tanta University Tanta 31527, Egypt. State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P. R. China
| | - Ying-Jie Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P. R. China
| | - Frank Fanqing Chen
- Life Sciences Division, Lawrence Berkeley National Laboratory, Mailstop 977, 1 Cyclotron Road, Berkeley, CA 94720, USA
| | - Yuhong Xu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
| | - Hadi Shafiee
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA. Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
350
|
Li P, Zhou J, Huang P, Zhang C, Wang W, Li C, Kong D. Self-assembled PEG- b-PDPA- b-PGEM copolymer nanoparticles as protein antigen delivery vehicles to dendritic cells: preparation, characterization and cellular uptake. Regen Biomater 2017; 4:11-20. [PMID: 28149525 PMCID: PMC5274708 DOI: 10.1093/rb/rbw044] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Revised: 10/22/2016] [Accepted: 10/27/2016] [Indexed: 12/13/2022] Open
Abstract
Antigen uptake by dendritic cells (DCs) is a key step for initiating antigen-specific T cell immunity. In the present study, novel synthetic polymeric nanoparticles were prepared as antigen delivery vehicles to improve the antigen uptake by DCs. Well-defined cationic and acid-responsive copolymers, monomethoxy poly(ethylene glycol)-block-poly(2-(diisopropyl amino) ethyl methacrylate)-block-poly(2-(guanidyl) ethyl methacrylate) (mPEG-b-PDPA-b-PGEM, PEDG) were synthesized by reversible addition-fragmentation chain transfer polymerization of 2-(diisopropylamino)ethyl methacrylate) and N-(tert-butoxycarbonyl) amino ethyl methacrylate monomers, followed by deprotection of tert-butyl protective groups and guanidinylation of obtained primary amines. 1H NMR, 13C NMR and GPC results indicated the successful synthesis of well-defined PEDG copolymers. PEDG copolymers could self-assemble into nanoparticles in aqueous solution, which were of cationic surface charges and showed acid-triggered disassembly contributed by PGEM and PDPA moieties, respectively. Significantly, PEDG nanoparticles could effectively condense with negatively charged model antigen ovalbumin (OVA) to form OVA/PEDG nanoparticle formulations with no influence on its secondary and tertiary structures demonstrating by far-UV circular dichroism and UV-vis spectra. In vitro antigen cellular uptake by bone marrow DCs (BMDCs) indicated using PEDG nanoparticles as antigen delivery vehicles could significantly improve the antigen uptake efficiency of OVA compared with free OVA or the commercialized Alum adjuvant. Moreover, as the surface cationic charges of OVA/PEDG nanoparticle formulations reduced, the uptake efficiency decreased correspondingly. Collectively, our work suggests that guanidinylated, cationic and acid-responsive PEDG nanoparticles represent a new kind of promising antigen delivery vehicle to DCs and hold great potential to serve as immunoadjuvants in the development of vaccines.
Collapse
Affiliation(s)
- Pan Li
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Junhui Zhou
- School of Chemical Engineering and Technology, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin University, Tianjin 300072, China
| | - Pingsheng Huang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Chuangnian Zhang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Weiwei Wang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Chen Li
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Deling Kong
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| |
Collapse
|