1
|
Mohammadzadeh V, Rahiman N, Cabral H, Quader S, Zirak MR, Taghavizadeh Yazdi ME, Jaafari MR, Alavizadeh SH. Poly-γ-glutamic acid nanoparticles as adjuvant and antigen carrier system for cancer vaccination. J Control Release 2023; 362:278-296. [PMID: 37640110 DOI: 10.1016/j.jconrel.2023.08.049] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 08/20/2023] [Accepted: 08/23/2023] [Indexed: 08/31/2023]
Abstract
Vaccination is an innovative strategy for cancer treatment by leveraging various components of the patients' immunity to boost an anti-tumor immune response. Rationally designed nanoparticles are well suited to maximize cancer vaccination by the inclusion of immune stimulatory adjuvants. Also, nanoparticles might control the pharmacokinetics and destination of the immune potentiating compounds. Poly-γ-glutamic acid (γ-PGA) based nanoparticles (NPs), which have a natural origin, can be easily taken up by dendritic cells (DCs), which leads to the secretion of cytokines which ameliorates the stimulation capacity of T cells. The intrinsic adjuvant properties and antigen carrier properties of γ-PGA NPs have been the focus of recent investigations as they can modulate the tumor microenvironment, can contribute to systemic anti-tumor immunity and subsequently inhibit tumor growth. This review provides a comprehensive overview on the potential of γ-PGA NPs as antigen carriers and/or adjuvants for anti-cancer vaccination.
Collapse
Affiliation(s)
- Vahideh Mohammadzadeh
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Niloufar Rahiman
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo 113-0033, Japan
| | - Sabina Quader
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, Kawasaki 210-0821, Japan
| | - Mohammad Reza Zirak
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyedeh Hoda Alavizadeh
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Liao Z, Huang J, Lo PC, Lovell JF, Jin H, Yang K. Self-adjuvanting cancer nanovaccines. J Nanobiotechnology 2022; 20:345. [PMID: 35883176 PMCID: PMC9316869 DOI: 10.1186/s12951-022-01545-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 07/04/2022] [Indexed: 11/12/2022] Open
Abstract
Nanovaccines, a new generation of vaccines that use nanoparticles as carriers and/or adjuvants, have been widely used in the prevention and treatment of various diseases, including cancer. Nanovaccines have sparked considerable interest in cancer therapy due to a variety of advantages, including improved access to lymph nodes (LN), optimal packing and presentation of antigens, and induction of a persistent anti-tumor immune response. As a delivery system for cancer vaccines, various types of nanoparticles have been designed to facilitate the delivery of antigens and adjuvants to lymphoid organs and antigen-presenting cells (APCs). Particularly, some types of nanoparticles are able to confer an immune-enhancing capability and can themselves be utilized for adjuvant-like effect for vaccines, suggesting a direction for a better use of nanomaterials and the optimization of cancer vaccines. However, this role of nanoparticles in vaccines has not been well studied. To further elucidate the role of self-adjuvanting nanovaccines in cancer therapy, we review the mechanisms of antitumor vaccine adjuvants with respect to nanovaccines with self-adjuvanting properties, including enhancing cross-presentation, targeting signaling pathways, biomimicking of the natural invasion process of pathogens, and further unknown mechanisms. We surveyed self-adjuvanting cancer nanovaccines in clinical research and discussed their advantages and challenges. In this review, we classified self-adjuvanting cancer nanovaccines according to the underlying immunomodulatory mechanism, which may provide mechanistic insights into the design of nanovaccines in the future.
Collapse
Affiliation(s)
- Zhiyun Liao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jing Huang
- College of Biomedicine and Health and College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Pui-Chi Lo
- Department of Biomedical Sciences, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Jonathan F Lovell
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY, 14260, USA
| | - Honglin Jin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China. .,College of Biomedicine and Health and College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Kunyu Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
3
|
Firdaus FZ, Skwarczynski M, Toth I. Developments in Vaccine Adjuvants. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2412:145-178. [PMID: 34918245 DOI: 10.1007/978-1-0716-1892-9_8] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Vaccines, including subunit, recombinant, and conjugate vaccines, require the use of an immunostimulator/adjuvant for maximum efficacy. Adjuvants not only enhance the strength and longevity of immune responses but may also influence the type of response. In this chapter, we review the adjuvants that are available for use in human vaccines, such as alum, MF59, AS03, and AS01. We extensively discuss their composition, characteristics, mechanism of action, and effects on the immune system. Additionally, we summarize recent trends in adjuvant discovery, providing a brief overview of saponins, TLRs agonists, polysaccharides, nanoparticles, cytokines, and mucosal adjuvants.
Collapse
Affiliation(s)
- Farrhana Ziana Firdaus
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Mariusz Skwarczynski
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia. .,Institute of Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia. .,School of Pharmacy, The University of Queensland, Woolloongabba, QLD, Australia.
| |
Collapse
|
4
|
Research progress on the utilisation of embedding technology and suitable delivery systems for improving the bioavailability of nattokinase: A review. FOOD STRUCTURE 2021. [DOI: 10.1016/j.foostr.2021.100219] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
5
|
Wang S. pH-Responsive Amphiphilic Carboxylate Polymers: Design and Potential for Endosomal Escape. Front Chem 2021; 9:645297. [PMID: 33834015 PMCID: PMC8021698 DOI: 10.3389/fchem.2021.645297] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 02/22/2021] [Indexed: 12/19/2022] Open
Abstract
The intracellular delivery of emerging biomacromolecular therapeutics, such as genes, peptides, and proteins, remains a great challenge. Unlike small hydrophobic drugs, these biotherapeutics are impermeable to the cell membrane, thus relying on the endocytic pathways for cell entry. After endocytosis, they are entrapped in the endosomes and finally degraded in lysosomes. To overcome these barriers, many carriers have been developed to facilitate the endosomal escape of these biomacromolecules. This mini-review focuses on the development of anionic pH-responsive amphiphilic carboxylate polymers for endosomal escape applications, including the design and synthesis of these polymers, the mechanistic insights of their endosomal escape capability, the challenges in the field, and future opportunities.
Collapse
Affiliation(s)
- Shiqi Wang
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| |
Collapse
|
6
|
Lv F, Jin Y, Feng X, Fan M, Ren C, Dai X, Zhang J, Li Z, Jin Y, Liu H. Traceable metallic antigen release for enhanced cancer immunotherapy. JOURNAL OF NANOPARTICLE RESEARCH : AN INTERDISCIPLINARY FORUM FOR NANOSCALE SCIENCE AND TECHNOLOGY 2021; 23:130. [PMID: 34149308 PMCID: PMC8202220 DOI: 10.1007/s11051-021-05256-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 06/01/2021] [Indexed: 05/04/2023]
Abstract
Tumor vaccine has shown outstanding advantages and good therapeutic effects in tumor immunotherapy. However, antigens in tumor vaccines can be easily cleared by the reticuloendothelium system in advance, which leads to poor therapeutic effect of tumor vaccines. Moreover, it was still hard to monitor the fate and distribution of antigens. To address these limitations, we synthesized a traceable nanovaccine based on gold nanocluster-labeled antigens and upconversion nanoparticles (UCNPs) for the treatment of melanoma in this study. PH-sensitive Schiff base bond is introduced between UCNPs and gold nanocluster-labeled ovalbumin antigens for monitoring antigens release. Our studies demonstrated that UCNPs conjugated metallic antigen showed excellent biocompatibility, pH-sensitive and therapeutic effect.
Collapse
Affiliation(s)
- Fangfang Lv
- College of Pharmaceutical Science, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Institute of Life Science and Green Development, Hebei University, Baoding, 071002 China
| | - Yan Jin
- College of Chemistry & Environmental Science, Institute of Life Science and Green Development, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002 China
| | - Xiaochen Feng
- College of Chemistry & Environmental Science, Institute of Life Science and Green Development, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002 China
| | - Miao Fan
- College of Chemistry & Environmental Science, Institute of Life Science and Green Development, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002 China
| | - Cui Ren
- College of Pharmaceutical Science, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Institute of Life Science and Green Development, Hebei University, Baoding, 071002 China
| | - Xinyue Dai
- College of Chemistry & Environmental Science, Institute of Life Science and Green Development, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002 China
| | - Jinchao Zhang
- College of Chemistry & Environmental Science, Institute of Life Science and Green Development, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002 China
| | - Zhenhua Li
- College of Chemistry & Environmental Science, Institute of Life Science and Green Development, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002 China
| | - Yi Jin
- College of Basic Medical Science, Hebei University, Baoding, 071000 China
| | - Huifang Liu
- College of Pharmaceutical Science, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Institute of Life Science and Green Development, Hebei University, Baoding, 071002 China
| |
Collapse
|
7
|
Vu MN, Kelly HG, Wheatley AK, Peng S, Pilkington EH, Veldhuis NA, Davis TP, Kent SJ, Truong NP. Cellular Interactions of Liposomes and PISA Nanoparticles during Human Blood Flow in a Microvascular Network. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2002861. [PMID: 32583981 PMCID: PMC7361276 DOI: 10.1002/smll.202002861] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/04/2020] [Indexed: 05/21/2023]
Abstract
A key concept in nanomedicine is encapsulating therapeutic or diagnostic agents inside nanoparticles to prolong blood circulation time and to enhance interactions with targeted cells. During circulation and depending on the selected application (e.g., cancer drug delivery or immune modulators), nanoparticles are required to possess low or high interactions with cells in human blood and blood vessels to minimize side effects or maximize delivery efficiency. However, analysis of cellular interactions in blood vessels is challenging and is not yet realized due to the diverse components of human blood and hemodynamic flow in blood vessels. Here, the first comprehensive method to analyze cellular interactions of both synthetic and commercially available nanoparticles under human blood flow conditions in a microvascular network is developed. Importantly, this method allows to unravel the complex interplay of size, charge, and type of nanoparticles on their cellular associations under the dynamic flow of human blood. This method offers a unique platform to study complex interactions of any type of nanoparticles in human blood flow conditions and serves as a useful guideline for the rational design of liposomes and polymer nanoparticles for diverse applications in nanomedicine.
Collapse
Affiliation(s)
- Mai N. Vu
- Australian Research Council Centre of Excellence in Convergent Bio‐Nano Science and TechnologyParkvilleVIC3052Australia
- Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVIC3052Australia
- Peter Doherty Institute for Infection and ImmunityDepartment of Microbiology and ImmunologyUniversity of MelbourneMelbourneVIC3000Australia
- Department of PharmaceuticsHanoi University of PharmacyHanoi10000Vietnam
| | - Hannah G. Kelly
- Australian Research Council Centre of Excellence in Convergent Bio‐Nano Science and TechnologyParkvilleVIC3052Australia
- Peter Doherty Institute for Infection and ImmunityDepartment of Microbiology and ImmunologyUniversity of MelbourneMelbourneVIC3000Australia
| | - Adam K. Wheatley
- Australian Research Council Centre of Excellence in Convergent Bio‐Nano Science and TechnologyParkvilleVIC3052Australia
- Peter Doherty Institute for Infection and ImmunityDepartment of Microbiology and ImmunologyUniversity of MelbourneMelbourneVIC3000Australia
| | - Scott Peng
- Australian Research Council Centre of Excellence in Convergent Bio‐Nano Science and TechnologyParkvilleVIC3052Australia
- Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVIC3052Australia
| | - Emily H. Pilkington
- Australian Research Council Centre of Excellence in Convergent Bio‐Nano Science and TechnologyParkvilleVIC3052Australia
- Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVIC3052Australia
- Peter Doherty Institute for Infection and ImmunityDepartment of Microbiology and ImmunologyUniversity of MelbourneMelbourneVIC3000Australia
| | - Nicholas A. Veldhuis
- Australian Research Council Centre of Excellence in Convergent Bio‐Nano Science and TechnologyParkvilleVIC3052Australia
- Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVIC3052Australia
| | - Thomas P. Davis
- Australian Research Council Centre of Excellence in Convergent Bio‐Nano Science and TechnologyParkvilleVIC3052Australia
- Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVIC3052Australia
- Australia Institute of Bioengineering & NanotechnologyUniversity of QueenslandBrisbaneQLD4072Australia
| | - Stephen J. Kent
- Australian Research Council Centre of Excellence in Convergent Bio‐Nano Science and TechnologyParkvilleVIC3052Australia
- Peter Doherty Institute for Infection and ImmunityDepartment of Microbiology and ImmunologyUniversity of MelbourneMelbourneVIC3000Australia
- Melbourne Sexual Health Centre and Department of Infectious DiseasesAlfred Hospital and Central Clinical SchoolMonash UniversityMelbourneVIC3004Australia
| | - Nghia P. Truong
- Australian Research Council Centre of Excellence in Convergent Bio‐Nano Science and TechnologyParkvilleVIC3052Australia
- Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVIC3052Australia
| |
Collapse
|
8
|
Hashemi V, Farhadi S, Ghasemi Chaleshtari M, Seashore-Ludlow B, Masjedi A, Hojjat-Farsangi M, Namdar A, Ajjoolabady A, Mohammadi H, Ghalamfarsa G, Jadidi-Niaragh F. Nanomedicine for improvement of dendritic cell-based cancer immunotherapy. Int Immunopharmacol 2020; 83:106446. [PMID: 32244048 DOI: 10.1016/j.intimp.2020.106446] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/24/2020] [Accepted: 03/24/2020] [Indexed: 02/08/2023]
Abstract
Dendritic cell (DC)-based cancer immunotherapy has shown impressive outcomes, including the development of the first FDA-approved anti-cancer vaccine. However, the clinical application of DC-based cancer immunotherapy is associated with various challenges. Promising novel tools for the administration of cancer vaccines has emerged from recent developments in nanoscale biomaterials. One current strategy to enhance targeted drug delivery, while minimizing drug-related toxicities, is the use of nanoparticles (NPs). These can be utilized for antigen delivery into DCs, which have been shown to provide potent T cell-stimulating effects. Therefore, NP delivery represents one promising approach for creating an effective and stable immune response without toxic side effects. The current review surveys cancer immunotherapy with particular attention toward NP-based delivery methods that target DCs.
Collapse
Affiliation(s)
- Vida Hashemi
- Department of Basic Science, Faculty of Medicine, Maragheh University of Medical Sciences, Maragheh, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shohreh Farhadi
- Student Research Committee, Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Brinton Seashore-Ludlow
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institute, Stockholm, Sweden
| | - Ali Masjedi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Hojjat-Farsangi
- Bioclinicum, Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden; The Persian Gulf Marine Biotechnology Medicine Research Center, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Afshin Namdar
- Department of Oncology, Cross Cancer Institute, The University of Alberta, Edmonton, Alberta, Canada
| | - Amir Ajjoolabady
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Mohammadi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Ghasem Ghalamfarsa
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran.
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
9
|
Ikeda M, Akagi T, Nagao M, Akashi M. Formulation Stability of Amphiphilic Poly(γ-Glutamic Acid) Nanoparticle and Evaluation of Cardiotoxicity of NPs With Human iPSC-Derived 3D-Cardiomyocyte Tissues. J Pharm Sci 2020; 109:2969-2974. [PMID: 32070703 DOI: 10.1016/j.xphs.2020.01.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 12/18/2019] [Accepted: 01/23/2020] [Indexed: 11/25/2022]
Abstract
We conducted a stability study of biodegradable and amphiphilic nanoparticles (NPs) consisting of phenylalanine-attached poly(γ-glutamic acid) for drug delivery to find the optimal formulation and define the optimal storage conditions using novel quantitative analytical methods. The stability of NP suspension and lyophilized NP powder manufactured by a dimethyl sulfoxide-based and an ethanol-based process was assessed under 5°C, 25°C/60% relative humidity and 40°C/75% relative humidity. The content of phenylalanine-attached poly(γ-glutamic acid), impurities, absolute molecular weight, appearance, clarity of solution, particle size, zeta potential, particle matter, osmolality, water content, and pH were evaluated as parameters of NP stability. Lyophilized NPs with trehalose showed better stability. The lyophilized NP formulation could therefore provide a stable and high-quality product for clinical studies and shows promise as an effective drug delivery system carrier. The cardiotoxicity of prospective impurities contained in NPs and reagents used in the manufacturing process with human-induced pluripotent stem cell-derived 3-dimensional cardiomyocyte tissues by centrifugation layer-by-layer technique was also evaluated. As a result, cardiotoxicity for NPs and reagents was not observed, and it was clarified that the potential risk to human safety from NPs is low. The applicability of the cardiotoxicity evaluation approaches with human-induced pluripotent stem cell-derived 3-dimensional cardiomyocyte tissues will be evaluated by centrifugation layer-by-layer technique.
Collapse
Affiliation(s)
- Mayumi Ikeda
- Analytical Development, Pharmaceutical Sciences, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan; Building Block Science Joint Research Chair, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita 565-0871, Japan
| | - Takami Akagi
- Building Block Science Joint Research Chair, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita 565-0871, Japan
| | - Masao Nagao
- Analytical Development, Pharmaceutical Sciences, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Mitsuru Akashi
- Building Block Science Joint Research Chair, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita 565-0871, Japan.
| |
Collapse
|
10
|
Castro F, Pinto ML, Almeida R, Pereira F, Silva AM, Pereira CL, Santos SG, Barbosa MA, Gonçalves RM, Oliveira MJ. Chitosan/poly(γ-glutamic acid) nanoparticles incorporating IFN-γ for immune response modulation in the context of colorectal cancer. Biomater Sci 2019; 7:3386-3403. [PMID: 31233057 DOI: 10.1039/c9bm00393b] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
IFN-γ therapy has been approved by the Food and Drug Administration (FDA) for the treatment of chronic granulomatous disease and severe malignant osteopetrosis. Despite the promising IFN-γ-based therapeutic applications, its limited success in clinical trials is related with limitations inherent to its molecular properties and with the difficulties to deliver it locally or with adequate periodicity to achieve a therapeutic effect. We have previously shown that chitosan (Ch)/poly(γ-glutamic acid) (γ-PGA) nanoparticles (NPs) are immunostimulatory, impairing colorectal cancer cell invasion. Ch is a biocompatible cationic polysaccharide extensively studied and already approved for biomedical applications while γ-PGA is a poly(amino acid), biodegradable and negatively charged. Here, we evaluated the potential of Ch/γ-PGA NPs as vehicles for IFN-γ and their ability to modulate immune cells' phenotype. In this study, Ch/IFN-γ/γ-PGA nanoparticles (IFN-γ-NPs) prepared by a co-acervation method, presenting a size of approximately 180 nm and a low polydispersity index, were tested for their immunomodulatory activity. These IFN-γ-NPs induced an immunostimulatory profile on dendritic cells (DCs) with increased cell surface costimulatory molecules and secretion of pro-inflammatory cytokines, including IL-6, IL-12p40 and TNF-α. IFN-γ-NPs also modulated the IL-10-stimulated macrophage profile, increasing their ability to secrete the pro-inflammatory cytokines IL-6, IL-12p40 and TNF-α. Concomitantly, these phenotypic alterations enhanced T cell proliferation. In addition, the ability of DCs and macrophages to induce colorectal cancer cell invasion was hampered in the presence of IFN-γ-NPs. Although the major observations were mediated by Ch/γ-PGA NPs, the incorporation of IFN-γ into NPs potentiated the expression of CD40 and CD86, and the impairment of colorectal cancer cell invasion. This work bridges the previously reported immunostimulatory capacity of Ch/γ-PGA NPs with their potential as carriers for immunomodulatory molecules, like IFN-γ, opening new avenues for their use in clinical settings.
Collapse
Affiliation(s)
- Flávia Castro
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal. and ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Marta L Pinto
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal. and CNC - Centro de Neurociências e Biologia Celular, Universidade de Coimbra, Portugal
| | - Rui Almeida
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.
| | - Flávia Pereira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal. and CBMA - Centro de Biologia Molecular e Ambiental, Universidade do Minho, Braga, Portugal
| | - Andreia M Silva
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal. and ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Catarina L Pereira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal. and ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Susana G Santos
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal. and ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Mário A Barbosa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal. and ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Raquel M Gonçalves
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.
| | - Maria J Oliveira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal. and Departamento de Patologia e Oncologia, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| |
Collapse
|
11
|
Musielak M, Piotrowski I, Suchorska WM. Superparamagnetic iron oxide nanoparticles (SPIONs) as a multifunctional tool in various cancer therapies. Rep Pract Oncol Radiother 2019; 24:307-314. [PMID: 31193459 DOI: 10.1016/j.rpor.2019.04.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 01/25/2019] [Accepted: 04/20/2019] [Indexed: 12/19/2022] Open
Abstract
Over the past two decades nanotechnology has become an important part of novel medical research. Researchers have made great progress in developing nanotechnology applications used for detecting and treating oncological diseases. Recently, many research groups have focused on the use of superparamagnetic iron oxide nanoparticles (SPIONs) in cancer treatment. Due to the range of therapeutic properties and possibilities of various modifications, SPIONs are a promising and multifunctional tool in various cancer therapies and may help to overcome the limitations of conventional therapies. Moreover, it is still necessary to develop new methods of treatment with expected properties, such as lower toxicity, long-lasting effectiveness and higher selectivity. Analyzing the literature data, we found that currently SPIONs are used in the transport of drugs, immunotherapy and hyperthermia. The main aim of this review is to present various cancer treatment therapies utilizing SPIONs, the importance of the experiments carried out by research groups and further perspectives in the nanotechnological use of SPIONs.
Collapse
Affiliation(s)
- Marika Musielak
- Radiobiology Laboratory, Greater Poland Cancer Centre, 61-866 Poznań, Poland
| | - Igor Piotrowski
- Radiobiology Laboratory, Greater Poland Cancer Centre, 61-866 Poznań, Poland.,Department of Electroradiology, Poznań University of Medical Sciences, 61-701 Poznań, Poland
| | - Wiktoria M Suchorska
- Radiobiology Laboratory, Greater Poland Cancer Centre, 61-866 Poznań, Poland.,Department of Electroradiology, Poznań University of Medical Sciences, 61-701 Poznań, Poland
| |
Collapse
|
12
|
|
13
|
Affiliation(s)
- Jenny Lou
- Department of Medical BiophysicsUniversity of Toronto Toronto M5G 1L7 Canada
- Princess Margaret Cancer CenterUniversity Health Network Toronto M5G 2C1 Canada
- Centre for Pharmaceutical OncologyUniversity of Toronto Toronto M5S 3M2 Canada
| | - Li Zhang
- Toronto General Hospital Research InstituteUniversity Health Network Toronto M5G 2C4 Canada
- Department of ImmunologyUniversity of Toronto Toronto M5S 1A8 Canada
- Department of Laboratory Medicine and PathobiologyUniversity of Toronto Toronto M5S 1A8 Canada
| | - Gang Zheng
- Department of Medical BiophysicsUniversity of Toronto Toronto M5G 1L7 Canada
- Princess Margaret Cancer CenterUniversity Health Network Toronto M5G 2C1 Canada
- Centre for Pharmaceutical OncologyUniversity of Toronto Toronto M5S 3M2 Canada
| |
Collapse
|
14
|
Jia J, Zhang Y, Xin Y, Jiang C, Yan B, Zhai S. Interactions Between Nanoparticles and Dendritic Cells: From the Perspective of Cancer Immunotherapy. Front Oncol 2018; 8:404. [PMID: 30319969 PMCID: PMC6167641 DOI: 10.3389/fonc.2018.00404] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 09/04/2018] [Indexed: 02/02/2023] Open
Abstract
Dendritic cells (DCs) are the primary antigen-presenting cells and play key roles in the orchestration of the innate and adaptive immune system. Targeting DCs by nanotechnology stands as a promising strategy for cancer immunotherapy. The physicochemical properties of nanoparticles (NPs) influence their interactions with DCs, thus altering the immune outcome of DCs by changing their functions in the processes of maturation, homing, antigen processing and antigen presentation. In this review, we summarize the recent progress in targeting DCs using NPs as a drug delivery carrier in cancer immunotherapy, the recognition of NPs by DCs, and the ways the physicochemical properties of NPs affect DCs' functions. Finally, the molecular pathways in DCs that are affected by NPs are also discussed.
Collapse
Affiliation(s)
- Jianbo Jia
- Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Institute of Environmental Research at Greater Bay, Guangzhou University, Guangzhou, China
| | - Yi Zhang
- School of Chemistry and Chemical Engineering, Shandong University, Jinan, China
| | - Yan Xin
- School of Chemistry and Chemical Engineering, Shandong University, Jinan, China
| | - Cuijuan Jiang
- School of Environmental Science and Engineering, Shandong University, Jinan, China
| | - Bing Yan
- Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Institute of Environmental Research at Greater Bay, Guangzhou University, Guangzhou, China.,School of Environmental Science and Engineering, Shandong University, Jinan, China
| | - Shumei Zhai
- School of Chemistry and Chemical Engineering, Shandong University, Jinan, China
| |
Collapse
|
15
|
Zhang L, Chang J, Zhao Y, Xu H, Wang T, Li Q, Xing L, Huang J, Wang Y, Liang Q. Fabrication of a triptolide-loaded and poly-γ-glutamic acid-based amphiphilic nanoparticle for the treatment of rheumatoid arthritis. Int J Nanomedicine 2018; 13:2051-2064. [PMID: 29670349 PMCID: PMC5894725 DOI: 10.2147/ijn.s151233] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Triptolide (TP) exhibits immunosuppressive, cartilage-protective and anti-inflammatory effects in rheumatoid arthritis. However, the toxicity of TP limits its widespread use. To decrease the toxic effects, we developed a novel nano-drug carrier system containing TP using poly-γ-glutamic acid-grafted di-tert-butyl L-aspartate hydrochloride (PAT). PAT had an average diameter of 79±18 nm, a narrow polydispersity index (0.18), a strong zeta potential (−32 mV) and a high drug encapsulation efficiency (EE1=48.6%) and loading capacity (EE2=19.2%), and exhibited controlled release (t1/2=29 h). The MTT assay and flow cytometry results indicated that PAT could decrease toxicity and apoptosis induced by free TP on RAW264.7 cells. PAT decreased lipopolysaccharides/interferon γ-induced cytokines expression of macrophage (P<0.05). In vivo, PAT accumulated at inflammatory joints, improved the survival rate and had fewer side effects on tumor necrosis factor α transgenic mice, compared to TP. The blood biochemical indexes revealed that PAT did not cause much damage to the kidney (urea nitrogen and creatinine) and liver (alanine aminotransferase and aspartate aminotransferase). In addition, PAT reduced inflammatory synovial tissue area (P<0.05), cartilage loss (P<0.05), tartrate-resistant acid phosphatase-positive osteoclast area (P<0.05) and bone erosion (P<0.05) in both knee and ankle joints, and showed similar beneficial effect as free TP. In summary, our newly formed nanoparticle, PAT, can reduce the toxicity and guarantee the efficacy of TP, which represents an effective drug candidate for RA with low adverse side effect.
Collapse
Affiliation(s)
- Li Zhang
- Department of Orthopaedics, Longhua Hospital.,Institute of Spine.,Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Junli Chang
- Department of Orthopaedics, Longhua Hospital.,Institute of Spine.,Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Yongjian Zhao
- Department of Orthopaedics, Longhua Hospital.,Institute of Spine.,Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Hao Xu
- Department of Orthopaedics, Longhua Hospital.,Institute of Spine.,Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Tengteng Wang
- Department of Orthopaedics, Longhua Hospital.,Institute of Spine.,Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Qiang Li
- Department of Orthopaedics, Longhua Hospital.,Institute of Spine.,Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Lianping Xing
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Jing Huang
- School of Life Science, East China Normal University
| | - Yongjun Wang
- Department of Orthopaedics, Longhua Hospital.,Institute of Spine.,Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China.,School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Qianqian Liang
- Department of Orthopaedics, Longhua Hospital.,Institute of Spine.,Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| |
Collapse
|
16
|
Castro F, Pinto ML, Silva AM, Pereira CL, Teixeira GQ, Gomez-Lazaro M, Santos SG, Barbosa MA, Gonçalves RM, Oliveira MJ. Pro-inflammatory chitosan/poly(γ-glutamic acid) nanoparticles modulate human antigen-presenting cells phenotype and revert their pro-invasive capacity. Acta Biomater 2017; 63:96-109. [PMID: 28919508 DOI: 10.1016/j.actbio.2017.09.016] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 09/06/2017] [Accepted: 09/13/2017] [Indexed: 02/06/2023]
Abstract
Anticancer immune responses depend on efficient presentation of tumor antigens and co-stimulatory signals provided by antigen-presenting cells (APCs). However, it is described that immature dendritic cells (DCs) and macrophages at the tumor site may have an immunosuppressive profile, which limits the activity of effector T cells and supports tumor progression. Therapeutic targeting of these innate immune cells, either aiming at their elimination or re-polarization towards an immunostimulatory profile, has been pointed as an attractive approach to control tumor progression. In the present work, we assessed the potential of Chitosan (Ch)/Poly(γ-glutamic acid) (γ-PGA) nanoparticles (NPs) to modulate macrophages and DCs inflammatory profile and to impair their ability to promote cancer cell invasion. Interestingly, Ch/γ-PGA NPs, prepared by co-acervation method, induced an immunostimulatory DCs phenotype, enhancing the expression of the co-stimulatory molecules CD86, CD40 and HLA-DR, and the secretion of the pro-inflammatory cytokines TNF-α, IL-12p40 and IL-6. Furthermore, Ch/γ-PGA NPs re-educated IL-10-stimulated macrophages towards a pro-inflammatory profile, decreasing the expression of CD163 and promoting the secretion of IL-12p40 and TNF-α. These alterations in the immune cells phenotype promoted CD4+ and CD8+ T cell activation/proliferation and partially inhibited APCs' ability to induce colorectal cancer cell invasion. Overall, our findings open new perspectives on the use of Ch/γ-PGA NPs as an immunomodulatory therapy for antigen-presenting cells reprogramming, providing a new tool for anticancer therapies. STATEMENT OF SIGNIFICANCE The immune system is responsible to detect and destroy abnormal cells preventing the development of cancer. However, the immunosuppressive tumor microenvironment can compromise the immune response favoring tumor progression. Thus, immune system modulation towards an immunostimulatory profile can improve anticancer therapies. This research focus on the development of chitosan/poly(γ-glutamic acid) nanoparticles (NPs) to modulate human antigen-presenting cells (APCs) phenotype and to counteract their pro-invasive capacity. Interestingly, Ch/γ-PGA NPs had a prominent effect in inducing macrophages and dendritic cells immunostimulatory phenotype, thus favoring T cell proliferation and inhibiting colorectal cancer cell invasion. We propose that their combination with other immunomodulatory drugs or conventional anticancer therapies can improve patients' outcome.
Collapse
|
17
|
Khalil IR, Burns ATH, Radecka I, Kowalczuk M, Khalaf T, Adamus G, Johnston B, Khechara MP. Bacterial-Derived Polymer Poly-y-Glutamic Acid (y-PGA)-Based Micro/Nanoparticles as a Delivery System for Antimicrobials and Other Biomedical Applications. Int J Mol Sci 2017; 18:ijms18020313. [PMID: 28157175 PMCID: PMC5343849 DOI: 10.3390/ijms18020313] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Accepted: 01/18/2017] [Indexed: 12/12/2022] Open
Abstract
In the past decade, poly-γ-glutamic acid (γ-PGA)-based micro/nanoparticles have garnered remarkable attention as antimicrobial agents and for drug delivery, owing to their controlled and sustained-release properties, low toxicity, as well as biocompatibility with tissue and cells. γ-PGA is a naturally occurring biopolymer produced by several gram-positive bacteria that, due to its biodegradable, non-toxic and non-immunogenic properties, has been used successfully in the medical, food and wastewater industries. Moreover, its carboxylic group on the side chains can offer an attachment point to conjugate antimicrobial and various therapeutic agents, or to chemically modify the solubility of the biopolymer. The unique characteristics of γ-PGA have a promising future for medical and pharmaceutical applications. In the present review, the structure, properties and micro/nanoparticle preparation methods of γ-PGA and its derivatives are covered. Also, we have highlighted the impact of micro/nanoencapsulation or immobilisation of antimicrobial agents and various disease-related drugs on biodegradable γ-PGA micro/nanoparticles.
Collapse
Affiliation(s)
- Ibrahim R Khalil
- Faculty of Science and Engineering, University of Wolverhampton, Wulfruna Street, Wolverhampton WV1 1LY, UK.
- Polish Academy of Sciences, Centre of Polymer and Carbon Materials, Zabrze 41-819, Poland.
| | - Alan T H Burns
- Faculty of Science and Engineering, University of Wolverhampton, Wulfruna Street, Wolverhampton WV1 1LY, UK.
| | - Iza Radecka
- Faculty of Science and Engineering, University of Wolverhampton, Wulfruna Street, Wolverhampton WV1 1LY, UK.
| | - Marek Kowalczuk
- Faculty of Science and Engineering, University of Wolverhampton, Wulfruna Street, Wolverhampton WV1 1LY, UK.
- Polish Academy of Sciences, Centre of Polymer and Carbon Materials, Zabrze 41-819, Poland.
| | - Tamara Khalaf
- Faculty of Science and Engineering, University of Wolverhampton, Wulfruna Street, Wolverhampton WV1 1LY, UK.
| | - Grazyna Adamus
- Polish Academy of Sciences, Centre of Polymer and Carbon Materials, Zabrze 41-819, Poland.
| | - Brian Johnston
- Faculty of Science and Engineering, University of Wolverhampton, Wulfruna Street, Wolverhampton WV1 1LY, UK.
| | - Martin P Khechara
- Faculty of Science and Engineering, University of Wolverhampton, Wulfruna Street, Wolverhampton WV1 1LY, UK.
| |
Collapse
|
18
|
Ma X, Gong N, Zhong L, Sun J, Liang XJ. Future of nanotherapeutics: Targeting the cellular sub-organelles. Biomaterials 2016; 97:10-21. [DOI: 10.1016/j.biomaterials.2016.04.026] [Citation(s) in RCA: 123] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 04/02/2016] [Accepted: 04/20/2016] [Indexed: 11/25/2022]
|
19
|
Fang RH, Kroll AV, Zhang L. Nanoparticle-Based Manipulation of Antigen-Presenting Cells for Cancer Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2015; 11:5483-96. [PMID: 26331993 PMCID: PMC4641138 DOI: 10.1002/smll.201501284] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 06/20/2015] [Indexed: 05/18/2023]
Abstract
Immunotherapeutic approaches for treating cancer overall have been receiving a considerable amount of interest due to the recent approval of several clinical formulations. Among the different modalities, anticancer vaccination acts by training the body to endogenously generate a response against tumor cells. However, despite the large amount of work that has gone into the development of such vaccines, the near absence of clinically approved formulations highlights the many challenges facing those working in the field. The generation of potent endogenous anticancer responses poses unique challenges due to the similarity between cancer cells and normal, healthy cells. As researchers continue to tackle the limited efficacy of vaccine formulations, fresh and novel approaches are being sought after to address many of the underlying problems. Here the application of nanoparticle technology towards the development of anticancer vaccines is discussed. Specifically, there is a focus on the benefits of using such strategies to manipulate antigen presenting cells (APCs), which are essential to the vaccination process, and how nanoparticle-based platforms can be rationally engineered to elicit appropriate downstream immune responses.
Collapse
Affiliation(s)
- Ronnie H. Fang
- Department of NanoEngineering and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ashley V. Kroll
- Department of NanoEngineering and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Liangfang Zhang
- Department of NanoEngineering and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
20
|
Amoozgar Z, Goldberg MS. Targeting myeloid cells using nanoparticles to improve cancer immunotherapy. Adv Drug Deliv Rev 2015; 91:38-51. [PMID: 25280471 DOI: 10.1016/j.addr.2014.09.007] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 09/15/2014] [Accepted: 09/16/2014] [Indexed: 12/23/2022]
Abstract
While nanoparticles have traditionally been used to deliver cytotoxic drugs directly to tumors to induce cancer cell death, emerging data suggest that nanoparticles are likely to generate a larger impact on oncology through the delivery of agents that can stimulate antitumor immunity. Tumor-targeted nanocarriers have generally been used to localize chemotherapeutics to tumors and thus decrease off-target toxicity while enhancing efficacy. Challengingly, tumor heterogeneity and evolution render tumor-intrinsic approaches likely to succumb to relapse. The immune system offers exquisite specificity, cytocidal potency, and long-term activity that leverage an adaptive memory response. For this reason, the ability to manipulate immune cell specificity and function would be desirable, and nanoparticles represent an exciting means by which to perform such manipulation. Dendritic cells and tumor-associated macrophages are cells of the myeloid lineage that function as natural phagocytes, so they naturally take up nanoparticles. Dendritic cells direct the specificity and potency of cellular immune responses that can be targeted for cancer vaccines. Herein, we discuss the specific criteria needed for efficient vaccine design, including but not limited to the route of administration, size, morphology, surface charge, targeting ligands, and nanoparticle composition. In contrast, tumor-associated macrophages are critical mediators of immunosuppression whose trans-migratory abilities can be exploited to localize therapeutics to the tumor core and which can be directly targeted for elimination or for repolarization to a tumor suppressive phenotype. It is likely that a combination of targeting dendritic cells to stimulate antitumor immunity and tumor-associated macrophages to reduce immune suppression will impart significant benefits and result in durable antitumor responses.
Collapse
|
21
|
Li W, Wei H, Li H, Gao J, Feng SS, Guo Y. Cancer nanoimmunotherapy using advanced pharmaceutical nanotechnology. Nanomedicine (Lond) 2015; 9:2587-605. [PMID: 25490427 DOI: 10.2217/nnm.14.127] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Immunotherapy is a promising option for cancer treatment that might cure cancer with fewer side effects by primarily activating the host's immune system. However, the effect of traditional immunotherapy is modest, frequently due to tumor escape and resistance of multiple mechanisms. Pharmaceutical nanotechnology, which is also called cancer nanotechnology or nanomedicine, has provided a practical solution to solve the limitations of traditional immunotherapy. This article reviews the latest developments in immunotherapy and nanomedicine, and illustrates how nanocarriers (including micelles, liposomes, polymer-drug conjugates, solid lipid nanoparticles and biodegradable nanoparticles) could be used for the cellular transfer of immune effectors for active and passive nanoimmunotherapy. The fine engineering of nanocarriers based on the unique features of the tumor microenvironment and extra-/intra-cellular conditions of tumor cells can greatly tip the triangle immunobalance among host, tumor and nanoparticulates in favor of antitumor responses, which shows a promising prospect for nanoimmunotherapy.
Collapse
Affiliation(s)
- Wei Li
- International Joint Cancer Institute, The Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, PR China
| | | | | | | | | | | |
Collapse
|
22
|
Nanoengineering of therapeutics for retinal vascular disease. Eur J Pharm Biopharm 2015; 95:323-30. [PMID: 26022642 DOI: 10.1016/j.ejpb.2015.05.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Revised: 04/29/2015] [Accepted: 05/05/2015] [Indexed: 01/07/2023]
Abstract
Retinal vascular diseases, including diabetic retinopathy, neovascular age related macular degeneration, and retinal vein occlusion, are leading causes of blindness in the Western world. These diseases share several common disease mechanisms, including vascular endothelial growth factor (VEGF) signaling, hypoxia, and inflammation, which provide opportunities for common therapeutic strategies. Treatment of these diseases using laser therapy, anti-VEGF injections, and/or steroids has significantly improved clinical outcomes. However, these strategies do not address the underlying root causes of pathology, and may have deleterious side effects. Furthermore, many patients continue to progress toward legal blindness despite receiving regular therapy. Nanomedicine, the engineering of therapeutics at the 1-100 nm scale, is a promising approach for improving clinical management of retinal vascular diseases. Nanomedicine-based technologies have the potential to revolutionize the treatment of ophthalmology, through enabling sustained release of drugs over several months, reducing side effects due to specific targeting of dysfunctional cells, and interfacing with currently "undruggable" targets. We will discuss emerging nanomedicine-based applications for the treatment of complications associated with retinal vascular diseases, including angiogenesis and inflammation.
Collapse
|
23
|
Ko WC, Chang CK, Wang HJ, Wang SJ, Hsieh CW. Process optimization of microencapsulation of curcumin in γ-polyglutamic acid using response surface methodology. Food Chem 2015; 172:497-503. [DOI: 10.1016/j.foodchem.2014.09.100] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 08/04/2014] [Accepted: 09/17/2014] [Indexed: 11/28/2022]
|
24
|
The Antiviral Effect of High-Molecular Weight Poly-Gamma-Glutamate against Newcastle Disease Virus on Murine Macrophage Cells. Adv Virol 2014; 2014:301386. [PMID: 25610463 PMCID: PMC4293867 DOI: 10.1155/2014/301386] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 11/29/2014] [Accepted: 11/30/2014] [Indexed: 12/28/2022] Open
Abstract
This study demonstrates the capacity of HM-γ-PGA treatment to significantly protect murine macrophage cells (RAW 264.7 cells) against NDV infection. Such protection can be explained by the induction of antiviral state of HM-γ-PGA in RAW 264.7 cells via TLR4-mediated IRF-3, IRF-7, IFN-β, and IFN-related gene induction as shown in time-dependent changes in mRNA expression confirmed by polymerase chain reaction (PCR). Moreover, the present research also showed that HM-γ-PGA can induce proinflammatory cytokine secretion in RAW 264.7 as measured by enzyme-linked immunosorbent assay (ELISA). Therefore, our findings suggest that HM-γ-PGA can be a potential antiviral substance that can inhibit NDV infection through its stimulation of antiviral state on RAW 264.7 cells. These results have been consistent with the previous studies showing that HM-γ-PGA can protect RAW 264.7 cells and mice against influenza infection. However, it should be noted that although murine macrophage cells are susceptible to NDV, they are not the natural host cells of the virus; thus further in vivo and in vitro studies involving chicken and chicken immune cells are needed to fully assess the efficacy and applicability of HM-γ-PGA in the poultry industry.
Collapse
|
25
|
Conniot J, Silva JM, Fernandes JG, Silva LC, Gaspar R, Brocchini S, Florindo HF, Barata TS. Cancer immunotherapy: nanodelivery approaches for immune cell targeting and tracking. Front Chem 2014; 2:105. [PMID: 25505783 PMCID: PMC4244808 DOI: 10.3389/fchem.2014.00105] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 10/31/2014] [Indexed: 12/14/2022] Open
Abstract
Cancer is one of the most common diseases afflicting people globally. New therapeutic approaches are needed due to the complexity of cancer as a disease. Many current treatments are very toxic and have modest efficacy at best. Increased understanding of tumor biology and immunology has allowed the development of specific immunotherapies with minimal toxicity. It is important to highlight the performance of monoclonal antibodies, immune adjuvants, vaccines and cell-based treatments. Although these approaches have shown varying degrees of clinical efficacy, they illustrate the potential to develop new strategies. Targeted immunotherapy is being explored to overcome the heterogeneity of malignant cells and the immune suppression induced by both the tumor and its microenvironment. Nanodelivery strategies seek to minimize systemic exposure to target therapy to malignant tissue and cells. Intracellular penetration has been examined through the use of functionalized particulates. These nano-particulate associated medicines are being developed for use in imaging, diagnostics and cancer targeting. Although nano-particulates are inherently complex medicines, the ability to confer, at least in principle, different types of functionality allows for the plausible consideration these nanodelivery strategies can be exploited for use as combination medicines. The development of targeted nanodelivery systems in which therapeutic and imaging agents are merged into a single platform is an attractive strategy. Currently, several nanoplatform-based formulations, such as polymeric nanoparticles, micelles, liposomes and dendrimers are in preclinical and clinical stages of development. Herein, nanodelivery strategies presently investigated for cancer immunotherapy, cancer targeting mechanisms and nanocarrier functionalization methods will be described. We also intend to discuss the emerging nano-based approaches suitable to be used as imaging techniques and as cancer treatment options.
Collapse
Affiliation(s)
- João Conniot
- Faculdade de Farmácia, Instituto de Investigação do Medicamento (iMed.ULisboa), Universidade de Lisboa Lisboa, Portugal
| | - Joana M Silva
- Faculdade de Farmácia, Instituto de Investigação do Medicamento (iMed.ULisboa), Universidade de Lisboa Lisboa, Portugal
| | - Joana G Fernandes
- Faculdade de Farmácia, Instituto de Investigação do Medicamento (iMed.ULisboa), Universidade de Lisboa Lisboa, Portugal
| | - Liana C Silva
- Faculdade de Farmácia, Instituto de Investigação do Medicamento (iMed.ULisboa), Universidade de Lisboa Lisboa, Portugal
| | - Rogério Gaspar
- Faculdade de Farmácia, Instituto de Investigação do Medicamento (iMed.ULisboa), Universidade de Lisboa Lisboa, Portugal
| | - Steve Brocchini
- EPSRC Centre for Innovative Manufacturing in Emergent Macromolecular Therapies, UCL School of Pharmacy London, UK
| | - Helena F Florindo
- Faculdade de Farmácia, Instituto de Investigação do Medicamento (iMed.ULisboa), Universidade de Lisboa Lisboa, Portugal
| | - Teresa S Barata
- EPSRC Centre for Innovative Manufacturing in Emergent Macromolecular Therapies, UCL School of Pharmacy London, UK
| |
Collapse
|
26
|
Shima F, Uto T, Akagi T, Baba M, Akashi M. Size effect of amphiphilic poly(γ-glutamic acid) nanoparticles on cellular uptake and maturation of dendritic cells in vivo. Acta Biomater 2013; 9:8894-901. [PMID: 23770225 DOI: 10.1016/j.actbio.2013.06.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2012] [Revised: 05/30/2013] [Accepted: 06/07/2013] [Indexed: 01/13/2023]
Abstract
We prepared size-regulated nanoparticles (NPs) composed of amphiphilic poly(γ-glutamic acid) (γ-PGA). In this study, 40, 100 and 200 nm γ-PGA-graft-l-phenylalanine ethylester (γ-PGA-Phe) NPs were employed. The size of NPs significantly influenced the uptake and activation behaviors of antigen-presenting cells (APCs). When 40 nm γ-PGA-Phe NPs were applied to these cells in vitro, they were highly activated compared with 100 and 200 nm NPs, while cellular uptake was size dependent. The size of the γ-PGA-Phe NPs also significantly affected their migration to the lymph nodes and uptake behavior of NPs by dendritic cells (DCs) in vivo. The 40 nm γ-PGA-Phe NPs migrated more rapidly to the lymph nodes and were taken up by a greater number of DCs compared with 100 and 200 nm NPs. On the other hand, when the amount of γ-PGA-Phe NPs taken up per DC was evaluated, it was higher for 100 and 200 nm NPs than for 40 nm NPs, which suggests that the larger γ-PGA-Phe NPs can deliver a large amount of antigen to a single DC compared with smaller NPs. Furthermore, when examined the maturation of DCs in lymph nodes, 40 nm γ-PGA-Phe NPs efficiently stimulated DCs. These results suggest that the activation, uptake behavior by APCs, migration to lymph nodes, and DC maturation can be controlled by the size of γ-PGA-Phe NPs.
Collapse
Affiliation(s)
- Fumiaki Shima
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita 565-0871, Japan; Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Kawaguchi Center Building, 4-1-8 Honcho, Kawaguchi 332-0012, Japan
| | | | | | | | | |
Collapse
|
27
|
Ungaro F, Conte C, Quaglia F, Tornesello ML, Buonaguro FM, Buonaguro L. VLPs and particle strategies for cancer vaccines. Expert Rev Vaccines 2013; 12:1173-1193. [PMID: 24124878 DOI: 10.1586/14760584.2013.836909] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2023]
Abstract
Effective delivery of tumor antigens to APCs is one of the key steps for eliciting a strong and durable immune response to tumors. Several cancer vaccines have been evaluated in clinical trials, based on soluble peptides, but results have not been fully satisfactory. To improve immunogenicity particles provide a valid strategy to display and/or incorporate epitopes which can be efficiently targeted to APCs for effective induction of adaptive immunity. In the present review, we report some leading technologies for developing particulate vaccines employed in cancer immunotherapy, highlighting the key parameters for a rational design to elicit both humoral and cellular responses.
Collapse
Affiliation(s)
- Francesca Ungaro
- Department of Pharmacy, University of Napoli Federico II, Via Domenico Montesano 49, 80131, Napoli, Italy
| | | | | | | | | | | |
Collapse
|
28
|
Tan S, Sasada T, Bershteyn A, Yang K, Ioji T, Zhang Z. Combinational delivery of lipid-enveloped polymeric nanoparticles carrying different peptides for anti-tumor immunotherapy. Nanomedicine (Lond) 2013; 9:635-47. [PMID: 23905577 DOI: 10.2217/nnm.13.67] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
AIM The authors aimed to investigate whether nanotechnology-based delivery of antigenic peptides is feasible for efficiently inducing anti-tumor cytotoxic T lymphocyte responses through vaccination. MATERIALS & METHODS Three different murine melanoma antigens were entrapped in lipid-coated poly(D,L-lactide-co-glycolide) nanoparticles (NPs) by the double emulsion method. RESULTS The loading efficiency of hydrophilic peptides was greatly improved when lipids were introduced to formulate lipid-coated NPs. The lipid-coated NPs carrying a single peptide and/or combinations of multiple lipid-coated NPs carrying antigenic peptides were characterized in vitro and in vivo in a C57/BL6 (B6) mouse model. Both the single melanoma antigen peptide-loaded NPs and combinational delivery of lipid-coated NPs carrying different peptides could induce antigen-specific T-cell responses. However, single peptide-loaded NPs failed to significantly delay the growth of subcutaneously inoculated B16 melanoma cells in a prophylactic setting. By contrast, the combinational delivery of lipid-coated NPs carrying different peptides significantly suppressed growth of inoculated B16 melanoma cells.
Collapse
Affiliation(s)
- Songwei Tan
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, China
| | | | | | | | | | | |
Collapse
|
29
|
Li N, Peng LH, Chen X, Zhang TY, Shao GF, Liang WQ, Gao JQ. Antigen-loaded nanocarriers enhance the migration of stimulated Langerhans cells to draining lymph nodes and induce effective transcutaneous immunization. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2013; 10:215-23. [PMID: 23792655 DOI: 10.1016/j.nano.2013.06.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 05/31/2013] [Accepted: 06/11/2013] [Indexed: 10/26/2022]
Abstract
UNLABELLED This study aims to investigate the efficacy of chitosan nanoparticles (CS-NPs) as a vehicle for transcutaneous antigen delivery in anti-tumor therapy. Ovalbumin (OVA) or gp100 (melanocyte-associated antigen gp100 protein)-loaded CS-sodium tripolyphosphate (TPP)-grafted NPs were prepared by crosslinking low-molecular-weight CS with TPP. Compared with the FITC-OVA solution, the encapsulated fluorescein isothiocyanate (FITC)-OVA-loaded NPs expressed much stronger cellular uptake ability in vitro and higher ability to migrate to lymph nodes in vivo. After transcutaneous administration, OVA-loaded NPs, with imiquimod as an adjuvant, increased the anti-OVA immunoglobulin G titer to levels similar to those induced by the OVA solution. The gp100-loaded NPs promoted the survival of tumor-bearing mice. These results provided evidence of CS-NPs as promising carriers for transcutaneous vaccine delivery, partly contributing to the increased uptake of NPs by skin antigen-presenting cells as well as their enhanced migration to the surrounding lymph nodes. FROM THE CLINICAL EDITOR In this study the efficacy of chitosan nanoparticle based vehicles for transcutaneous antigen delivery is investigated in anti-tumor therapy. Authors demonstrate that such nanoparticles may be efficient carriers partly due to their increased uptake by antigen-presenting cells in the skin and their enhanced migration to surrounding lymph nodes.
Collapse
Affiliation(s)
- Ni Li
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China; The Affiliated Lihuili Hospital, Ningbo University School of Medicine, Ningbo, China
| | - Li-Hua Peng
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| | - Xi Chen
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Tian-Yuan Zhang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Guo-Feng Shao
- The Affiliated Lihuili Hospital, Ningbo University School of Medicine, Ningbo, China
| | - Wen-Quan Liang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Jian-Qing Gao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China; The Novel Transdermal Research Center of Jiangsu Province, Changzhou, China.
| |
Collapse
|
30
|
Polymeric nanogels as vaccine delivery systems. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2013; 9:159-73. [DOI: 10.1016/j.nano.2012.06.001] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Revised: 04/11/2012] [Accepted: 06/18/2012] [Indexed: 01/22/2023]
|
31
|
|
32
|
Grant EV, Thomas M, Fortune J, Klibanov AM, Letvin NL. Enhancement of plasmid DNA immunogenicity with linear polyethylenimine. Eur J Immunol 2012; 42:2937-48. [DOI: 10.1002/eji.201242410] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Revised: 06/05/2012] [Accepted: 08/01/2012] [Indexed: 11/11/2022]
Affiliation(s)
- Evita V. Grant
- Division of Viral Pathogenesis; Beth Israel Deaconess Medical Center; Harvard Medical School; Boston Massachusetts USA
| | - Mini Thomas
- Department of Chemistry; Purdue University; West Lafayette Indiana USA
| | - Jennifer Fortune
- Department of Chemistry and Division of Biological Engineering; Massachusetts Institute of Technology; Cambridge Massachusetts USA
| | - Alexander M. Klibanov
- Department of Chemistry and Division of Biological Engineering; Massachusetts Institute of Technology; Cambridge Massachusetts USA
| | - Norman L. Letvin
- Division of Viral Pathogenesis; Beth Israel Deaconess Medical Center; Harvard Medical School; Boston Massachusetts USA
| |
Collapse
|
33
|
Moon HJ, Lee JS, Choi YK, Park JY, Talactac MR, Chowdhury MY, Poo H, Sung MH, Lee JH, Jung JU, Kim CJ. Induction of type I interferon by high-molecular poly-γ-glutamate protects B6.A2G-Mx1 mice against influenza A virus. Antiviral Res 2012; 94:98-102. [DOI: 10.1016/j.antiviral.2012.02.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2011] [Revised: 02/17/2012] [Accepted: 02/21/2012] [Indexed: 11/27/2022]
|
34
|
Poly-γ-glutamic acid nanoparticles and aluminum adjuvant used as an adjuvant with a single dose of Japanese encephalitis virus-like particles provide effective protection from Japanese encephalitis virus. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2011; 19:17-22. [PMID: 22089248 DOI: 10.1128/cvi.05412-11] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
To maintain immunity against Japanese encephalitis virus (JEV), a formalin-inactivated Japanese encephalitis (JE) vaccine should be administered several times. The repeated vaccination is not helpful in the case of a sudden outbreak of JEV or when urgent travel to a high-JEV-risk region is required; however, there are few single-injection JE vaccine options. In the present study, we investigated the efficacy of a single dose of a new effective JE virus-like particle preparation containing the JE envelope protein (JE-VLP). Although single administration with JE-VLP protected less than 50% of mice against lethal JEV infection, adding poly(γ-glutamic acid) nanoparticles (γ-PGA-NPs) or aluminum adjuvant (alum) to JE-VLP significantly protected more than 90% of the mice. A single injection of JE-VLP with either γ-PGA-NPs or alum induced a significantly greater anti-JEV neutralizing antibody titer than JE-VLP alone. The enhanced titers were maintained for more than 6 months, resulting in long-lasting protection of 90% of the immunized mice. Although the vaccine design needs further modification to reach 100% protection, a single dose of JE-VLP with γ-PGA-NPs may be a useful step in developing a next-generation vaccine to stop a JE outbreak or to immunize travelers or military personnel.
Collapse
|
35
|
Mukai Y, Yoshinaga T, Yoshikawa M, Matsuo K, Yoshikawa T, Matsuo K, Niki K, Yoshioka Y, Okada N, Nakagawa S. Induction of endoplasmic reticulum-endosome fusion for antigen cross-presentation induced by poly (γ-glutamic acid) nanoparticles. THE JOURNAL OF IMMUNOLOGY 2011; 187:6249-55. [PMID: 22095716 DOI: 10.4049/jimmunol.1001093] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
We previously reported that poly (γ-glutamic acid)-based nanoparticles (γ-PGA NPs) are excellent vaccine carriers for inducing efficient cross-presentation in dendritic cells, thereby producing strong antitumor immunity in vivo. Analyzing the mechanism of cross-presentation induced by γ-PGA NPs will be useful toward designing novel vaccine carriers. In this study, we show an intracellular mechanism of efficient cross-presentation induced by OVA-loaded γ-PGA NPs. Cross-presentation induced by γ-PGA NPs depended on cytoplasmic proteasomes and TAP, similar to the classical MHC class I presentation pathway for endogenous Ags. Intracellular behavior analyzed by confocal laser scanning microscopy revealed that encapsulated OVA and γ-PGA accumulated in both the endoplasmic reticulum (ER) and endosome compartments within 2 h. At the same time, electron microscopy analysis clearly showed that intracellular γ-PGA NPs and encapsulated Au NPs were enveloped in endosome-like vesicles, not in the ER. These findings strongly suggest that γ-PGA NPs enhance ER-endosome fusion for cross-presentation. Moreover, inhibition of ER translocon sec61 significantly decreased the γ-PGA NP/OVA-mediated cross-presentation efficiency, indicating that sec61 is important for transporting Ags from the fused ER-endosome to the cytoplasm. These findings imply that the ER-endosome complex is key for the efficient cross-presentation of Ags encapsulated in γ-PGA NPs.
Collapse
Affiliation(s)
- Yohei Mukai
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Kuo YC, Yu HW. Polyethyleneimine/poly-(γ-glutamic acid)/poly(lactide-co-glycolide) nanoparticles for loading and releasing antiretroviral drug. Colloids Surf B Biointerfaces 2011; 88:158-64. [DOI: 10.1016/j.colsurfb.2011.06.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Revised: 06/21/2011] [Accepted: 06/21/2011] [Indexed: 01/21/2023]
|
37
|
Akagi T, Baba M, Akashi M. Biodegradable Nanoparticles as Vaccine Adjuvants and Delivery Systems: Regulation of Immune Responses by Nanoparticle-Based Vaccine. ADVANCES IN POLYMER SCIENCE 2011. [DOI: 10.1007/12_2011_150] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
38
|
Kurosaki T, Kitahara T, Nakamura T, Nishida K, Fumoto S, Kodama Y, Nakagawa H, Higuchi N, Sasaki H. Development of effective cancer vaccine using targeting system of antigen protein to APCs. Pharm Res 2011; 29:483-9. [PMID: 21887598 DOI: 10.1007/s11095-011-0571-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2011] [Accepted: 08/12/2011] [Indexed: 10/17/2022]
Abstract
PURPOSE To develop a novel cancer vaccine using the targeting system of antigen protein to antigen-presenting cells (APCs) for efficient and safe cancer therapy. METHODS The novel delivery system was constructed with antigen protein, benzalkonium chloride (BK), and γ-polyglutamic acid (γ-PGA), using ovalbumin (OVA) as a model antigen protein and evaluating its immune induction effects and utilities for cancer vaccine. RESULTS BK and γ-PGA enabled encapsulation of OVA and formed stable anionic particles at nanoscale, OVA/BK/γ-PGA complex. Complex was taken up by dendritic cell line DC2.4 cells efficiently. We subcutaneously administered the complex to mice and examined induction of IgGs. The complex induced not only Th2-type immunoglobulins but also Th1-type immunoglobulins. OVA/BK/γ-PGA complex inhibited tumor growth of E.G7 cells expressing OVA regularly; administered OVA/BK/γ-PGA complex completely rejected tumor cells. CONCLUSION The novel vaccine could be platform technology for a cancer vaccine.
Collapse
Affiliation(s)
- Tomoaki Kurosaki
- Department of Drug Delivery Research Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Transport of saquinavir across human brain-microvascular endothelial cells by poly(lactide-co-glycolide) nanoparticles with surface poly-(γ-glutamic acid). Int J Pharm 2011; 416:365-75. [DOI: 10.1016/j.ijpharm.2011.06.037] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Revised: 05/27/2011] [Accepted: 06/22/2011] [Indexed: 11/21/2022]
|
40
|
Matsuo K, Ishii Y, Matsuo K, Yoshinaga T, Akashi M, Mukai Y, Yoshioka Y, Okada N, Nakagawa S. The utility of poly(γ-glutamic acid) nanoparticles as antigen delivery carriers in dendritic cell-based cancer immunotherapy. Biol Pharm Bull 2011; 33:2003-7. [PMID: 21139241 DOI: 10.1248/bpb.33.2003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cytotoxic T-lymphocytes (CTLs) specific for tumor-associated antigens (TAAs) act in the immune surveillance system as major effector cells to eliminate malignant cells. Immunization with TAA-loaded dendritic cells (DCs) has great potential for treating cancer, because DCs are potent antigen-presenting cells capable of inducing antigen-specific CTLs by the primary activation of naive T-lymphocytes. The establishment of a non-cytotoxic and efficient antigen delivery method is required to improve the efficacy of DC-based cancer immunotherapy. We developed biodegradable poly(γ-glutamic acid) nanoparticles (γ-PGA NPs) that can efficiently entrap various proteins as antigen delivery carriers. γ-PGA NPs efficiently delivered entrapped antigenic proteins into DCs without cytotoxicity and presented antigens to DCs via major histocompatibility complex class I and II molecules. Immunization with TAA-loaded DCs using γ-PGA NPs inhibited tumor growth by inducing TAA-specific CTLs. These findings indicate that γ-PGA NPs can function as useful antigen delivery carriers in DC-based cancer immunotherapy.
Collapse
Affiliation(s)
- Keisuke Matsuo
- Department of Biotechnology and Therapeutics, Osaka University, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Matsuo K, Koizumi H, Akashi M, Nakagawa S, Fujita T, Yamamoto A, Okada N. Intranasal immunization with poly(γ-glutamic acid) nanoparticles entrapping antigenic proteins can induce potent tumor immunity. J Control Release 2011; 152:310-6. [DOI: 10.1016/j.jconrel.2011.03.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2010] [Revised: 03/04/2011] [Accepted: 03/08/2011] [Indexed: 12/22/2022]
|
42
|
Zhang Z, Tongchusak S, Mizukami Y, Kang YJ, Ioji T, Touma M, Reinhold B, Keskin DB, Reinherz EL, Sasada T. Induction of anti-tumor cytotoxic T cell responses through PLGA-nanoparticle mediated antigen delivery. Biomaterials 2011; 32:3666-78. [DOI: 10.1016/j.biomaterials.2011.01.067] [Citation(s) in RCA: 192] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Accepted: 01/26/2011] [Indexed: 12/18/2022]
|
43
|
Suppression of phagocytic cells in retinal disorders using amphiphilic poly(γ-glutamic acid) nanoparticles containing dexamethasone. J Control Release 2011; 151:65-73. [DOI: 10.1016/j.jconrel.2010.11.029] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2010] [Revised: 11/22/2010] [Accepted: 11/26/2010] [Indexed: 11/19/2022]
|
44
|
Yaeger KA, Kurt RA. Construction and Evaluation of a Combined Cyclophosphamide/Nanoparticle Anticancer Vaccine. ACTA ACUST UNITED AC 2011. [DOI: 10.4236/jct.2011.23053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
45
|
Crownover EF, Convertine AJ, Stayton PS. pH-responsive polymer–antigen vaccine bioconjugates. Polym Chem 2011. [DOI: 10.1039/c1py00060h] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
46
|
Foster S, Duvall CL, Crownover EF, Hoffman AS, Stayton PS. Intracellular delivery of a protein antigen with an endosomal-releasing polymer enhances CD8 T-cell production and prophylactic vaccine efficacy. Bioconjug Chem 2010; 21:2205-12. [PMID: 21043513 PMCID: PMC3075329 DOI: 10.1021/bc100204m] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Protein-based vaccines have significant potential as infectious disease and anticancer therapeutics, but clinical impact has been limited in some applications by their inability to generate a coordinated cellular immune response. Here, a pH-responsive carrier incorporating poly(propylacrylic acid) (PPAA) was evaluated to test whether improved cytosolic delivery of a protein antigen could enhance CD8+ cytotoxic lymphocyte generation and prophylactic tumor vaccine responses. PPAA was directly conjugated to the model ovalbumin antigen via reducible disulfide linkages and was also tested in a particulate formulation after condensation with cationic poly(dimethylaminoethyl methacrylate) (PDMAEMA). Intracellular trafficking studies revealed that both PPAA-containing formulations were stably internalized and evaded exocytotic pathways, leading to increased intracellular accumulation and potential access to the cytosolic MHC-1 antigen presentation pathway. In an EG.7-OVA mouse tumor protection model, both PPAA-containing carriers robustly inhibited tumor growth and led to an approximately 3.5-fold increase in the longevity of tumor-free survival relative to controls. Mechanistically, this response was attributed to the 8-fold increase in production of ovalbumin-specific CD8+ T-lymphocytes and an 11-fold increase in production of antiovalbumin IgG. Significantly, this is one of the first demonstrated examples of in vivo immunotherapeutic efficacy using soluble protein-polymer conjugates. These results suggest that carriers enhancing cytosolic delivery of protein antigens could lead to more robust CD8+ T-cell response and demonstrate the potential of pH-responsive PPAA-based carriers for therapeutic vaccine applications.
Collapse
Affiliation(s)
| | | | - Emily F. Crownover
- Department of Bioengineering and the Center for Intracellular Delivery of Biologics, University of Washington, Seattle WA 98195
| | - Allan S. Hoffman
- Department of Bioengineering and the Center for Intracellular Delivery of Biologics, University of Washington, Seattle WA 98195
| | - Patrick S. Stayton
- Department of Bioengineering and the Center for Intracellular Delivery of Biologics, University of Washington, Seattle WA 98195
| |
Collapse
|
47
|
Jain A, Yan W, Miller KR, O'Carra R, Woodward JG, Mumper RJ. Tresyl-based conjugation of protein antigen to lipid nanoparticles increases antigen immunogenicity. Int J Pharm 2010; 401:87-92. [PMID: 20837122 PMCID: PMC2967624 DOI: 10.1016/j.ijpharm.2010.09.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2010] [Revised: 09/03/2010] [Accepted: 09/03/2010] [Indexed: 12/16/2022]
Abstract
The present studies were aimed at investigating the engineering of NPs with protein-conjugated-surfactant at their surface. In order to increase the immunogenicity of a protein antigen, Brij 78 was functionalized by tresyl chloride and then further reacted with the primary amine of the model proteins ovalbumin (OVA) or horseradish peroxide (HRP). The reaction yielded Brij 78-OVA and Brij 78-HRP conjugates which were then used directly to form NP-OVA or NP-HRP using a one-step warm oil-in-water microemulsion precursor method with emulsifying wax as the oil phase, and Brij 78 and the Brij 78-OVA or Brij 78-HRP conjugate as surfactants. Similarly, Brij 700 was conjugated to HIV p24 antigen to yield Brij 700-p24 conjugate. The utility of these NPs for enhancing the immune responses to protein-based vaccines was evaluated in vivo using ovalbumin (OVA) as model protein and p24 as a relevant HIV antigen. In separate in vivo studies, female BALB/c mice were immunized by subcutaneous (s.c.) injection with NP-OVA and NP-p24 formulations along with several control formulations. These results suggested that with multiple antigens, covalent attachment of the antigen to the NP significantly enhanced antigen-specific immune responses. This facile covalent conjugation and incorporation method may be utilized to further incorporate other protein antigens, even multiple antigens, into an enhanced vaccine delivery system.
Collapse
Affiliation(s)
- Anekant Jain
- Division of Molecular Pharmaceutics, and the Center for Nanotechnology in Drug Delivery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27514
| | - Weili Yan
- Department of Pharmaceutical Sciences, Auburn University, Harrison School of Pharmacy, Auburn, AL 36849
| | - Keith R. Miller
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, NC 27514
| | - Ronan O'Carra
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536
| | - Jerold G. Woodward
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY 40536
| | - Russell J. Mumper
- Division of Molecular Pharmaceutics, and the Center for Nanotechnology in Drug Delivery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27514
| |
Collapse
|
48
|
Kanwar JR, Mohan RR, Kanwar RK, Roy K, Bawa R. Applications of aptamers in nanodelivery systems in cancer, eye and inflammatory diseases. Nanomedicine (Lond) 2010; 5:1435-45. [PMID: 21128724 DOI: 10.2217/nnm.10.115] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Aptamers are an interesting class of molecules that have potential in many facets of human health. They are characterized by high affinity and specificity to their targets, are small in size, have similar properties to antibodies, but are made synthetically. All of these properties, among others, give aptamers the potential to diagnose, image and treat like no other molecules. By combining the unique properties of aptamers with the ever expanding field of nanotechnology and all it has to offer, we are entering a very promising new area of targeted nanodelivery treatments. These treatments have found success in the complex disease processes of cancer, eye and inflammatory diseases.
Collapse
Affiliation(s)
- Jagat R Kanwar
- Laboratory of Immunology & Molecular Biomedical Research, Centre for Biotechnology & Interdisciplinary Biosciences (BioDeakin), Institute for Technology & Research Innovation (ITRI), Geelong Technology Precinct, Deakin University, Pigdons Road, Waurn Ponds, Geelong, Victoria 3217, Australia.
| | | | | | | | | |
Collapse
|
49
|
|
50
|
Krishnamachari Y, Geary SM, Lemke CD, Salem AK. Nanoparticle delivery systems in cancer vaccines. Pharm Res 2010; 28:215-36. [PMID: 20721603 DOI: 10.1007/s11095-010-0241-4] [Citation(s) in RCA: 165] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2010] [Accepted: 08/06/2010] [Indexed: 11/24/2022]
Abstract
Therapeutic strategies that involve the manipulation of the host's immune system are gaining momentum in cancer research. Antigen-loaded nanocarriers are capable of being actively taken up by antigen-presenting cells (APCs) and have shown promising potential in cancer immunotherapy by initiating a strong immunostimulatory cascade that results in potent antigen-specific immune responses against the cancer. Such carrier systems offer versatility in that they can simultaneously co-deliver adjuvants with the antigens to enhance APC activation and maturation. Furthermore, modifying the surface properties of these nanocarriers affords active targeting properties to APCs and/or enhanced accumulation in solid tumors. Here, we review some recent advances in these colloidal and particulate nanoscale systems designed for cancer immunotherapy and the potential for these systems to translate into clinical cancer vaccines.
Collapse
Affiliation(s)
- Yogita Krishnamachari
- Department of Pharmaceutical Sciences & Experimental Therapeutics College of Pharmacy, University of Iowa, Iowa City, Iowa 52242, USA
| | | | | | | |
Collapse
|