1
|
Reay SL, Marina Ferreira A, Hilkens CMU, Novakovic K. The Paradoxical Immunomodulatory Effects of Chitosan in Biomedicine. Polymers (Basel) 2024; 17:19. [PMID: 39795422 PMCID: PMC11723117 DOI: 10.3390/polym17010019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/19/2024] [Accepted: 12/22/2024] [Indexed: 01/13/2025] Open
Abstract
Chitosan is widely explored in the field of biomedicine due to its abundance and reported properties, including biocompatibility, biodegradability, non-toxicity, mucoadhesion, and anti-microbial activity. Although our understanding of the immune response to chitosan has evolved, confusion remains regarding whether chitosan is a pro- or anti-inflammatory biomaterial. Tackling this knowledge gap is essential for the translation of chitosan-based biomaterials to clinical use. Herein, we provide an overview of the immune responses to chitosan, exploring the roles of endotoxin contamination and physiochemical properties in immunomodulation. Ultimately, this literature review concludes that various physiochemical properties, including molecular weight, degree of deacetylation and polydispersity, endotoxin contamination, and cellular environment, interplay in the complex process of chitosan immunomodulation, which can lead to both pro- and anti-inflammatory effects.
Collapse
Affiliation(s)
- Sophie L. Reay
- School of Engineering, Newcastle University, Newcastle Upon Tyne NE1 7RU, UK; (A.M.F.); (K.N.)
| | - Ana Marina Ferreira
- School of Engineering, Newcastle University, Newcastle Upon Tyne NE1 7RU, UK; (A.M.F.); (K.N.)
| | - Catharien M. U. Hilkens
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE1 7RU, UK;
| | - Katarina Novakovic
- School of Engineering, Newcastle University, Newcastle Upon Tyne NE1 7RU, UK; (A.M.F.); (K.N.)
| |
Collapse
|
2
|
Khazaei S, Varela-Calviño R, Rad-Malekshahi M, Quattrini F, Jokar S, Rezaei N, Balalaie S, Haririan I, Csaba N, Garcia-Fuentes M. Self-assembled peptide/polymer hybrid nanoplatform for cancer immunostimulating therapies. Drug Deliv Transl Res 2024; 14:455-473. [PMID: 37721693 PMCID: PMC10761384 DOI: 10.1007/s13346-023-01410-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2023] [Indexed: 09/19/2023]
Abstract
Integrating peptide epitopes in self-assembling materials is a successful strategy to obtain nanovaccines with high antigen density and improved efficacy. In this study, self-assembling peptides containing MAGE-A3/PADRE epitopes were designed to generate functional therapeutic nanovaccines. To achieve higher stability, peptide/polymer hybrid nanoparticles were formulated by controlled self-assembly of the engineered peptides. The nanoparticles showed good biocompatibility to both human red blood- and dendritic cells. Incubation of the nanoparticles with immature dendritic cells triggered immune effects that ultimately activated CD8 + cells. The antigen-specific and IgG antibody responses of healthy C57BL/6 mice vaccinated with the nanoparticles were analyzed. The in vivo results indicate a specific response to the nanovaccines, mainly mediated through a cellular pathway. This research indicates that the immunogenicity of peptide epitope vaccines can be effectively enhanced by developing self-assembled peptide-polymer hybrid nanostructures.
Collapse
Affiliation(s)
- Saeedeh Khazaei
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, CiMUS Research Center and Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Ruben Varela-Calviño
- Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Mazda Rad-Malekshahi
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Federico Quattrini
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, CiMUS Research Center and Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Safura Jokar
- Department of Nuclear Pharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeed Balalaie
- Peptide Chemistry Research Center, K. N. Toosi University of Technology, Tehran, Iran
| | - Ismaeil Haririan
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Noemi Csaba
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, CiMUS Research Center and Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Marcos Garcia-Fuentes
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, CiMUS Research Center and Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain.
| |
Collapse
|
3
|
Zhou H, Wan F, Jian Y, Guo F, Zhang M, Shi S, Yang L, Li S, Liu Y, Ding W. Chitosan/dsRNA polyplex nanoparticles advance environmental RNA interference efficiency through activating clathrin-dependent endocytosis. Int J Biol Macromol 2023; 253:127021. [PMID: 37741481 DOI: 10.1016/j.ijbiomac.2023.127021] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 09/11/2023] [Accepted: 09/20/2023] [Indexed: 09/25/2023]
Abstract
Chitosan, as a promising gene nanocarrier for enhancing RNA interference (RNAi) efficiency, displays tremendous application prospects in addressing dsRNA delivery concerns. However, the molecular mechanism of chitosan/dsRNA polyplex nanoparticles (PNs) for advancing dsRNA delivery efficiency remains largely unknown. Here, chitosan/dsRNA PNs were prepared by an electrostatic attraction method. The results showed that the chitosan/dsRNA PNs significantly advance stability, and cellular uptake efficiency of dsRNA, and RNAi efficiency. RNA-Seq and qPCR assays further revealed that chitosan/dsRNA PNs upregulated the key clathrin heavy chain (CHC) gene for activating clathrin-dependent endocytosis (CDE) pathway. Additionally, inhibition of CDE hindered the robust RNAi responses of chitosan/dsRNA PNs using an inhibitor (chlorpromazine) and an RNAi-of-RNAi strategy. Ultimately, microscale thermophoresis assay confirmed that chitosan/dsRNA PNs directly bound to CHC protein, which was a core component in CDE, to advance RNAi efficiency. To our knowledge, our findings firstly illuminate the molecular mechanism how chitosan nanoparticles-based RNAi deliver dsRNA for enhancing RNAi efficiency. Above mechanism will advance the extensive utilization of nanocarrier-based RNAi in pest management and gene delivery.
Collapse
Affiliation(s)
- Hong Zhou
- Institute of Pesticide Science, College of Plant Protection, Southwest University, Chongqing 400715, PR China
| | - Fenglin Wan
- Institute of Pesticide Science, College of Plant Protection, Southwest University, Chongqing 400715, PR China
| | - Yufan Jian
- Institute of Pesticide Science, College of Plant Protection, Southwest University, Chongqing 400715, PR China
| | - Fuyou Guo
- Institute of Pesticide Science, College of Plant Protection, Southwest University, Chongqing 400715, PR China
| | - Miao Zhang
- Institute of Pesticide Science, College of Plant Protection, Southwest University, Chongqing 400715, PR China
| | - Shiyao Shi
- Institute of Pesticide Science, College of Plant Protection, Southwest University, Chongqing 400715, PR China
| | - Liang Yang
- Institute of Pesticide Science, College of Plant Protection, Southwest University, Chongqing 400715, PR China
| | - Shili Li
- Institute of Pesticide Science, College of Plant Protection, Southwest University, Chongqing 400715, PR China
| | - Ying Liu
- Institute of Pesticide Science, College of Plant Protection, Southwest University, Chongqing 400715, PR China
| | - Wei Ding
- Institute of Pesticide Science, College of Plant Protection, Southwest University, Chongqing 400715, PR China.
| |
Collapse
|
4
|
Shah S, Chougule MB, Kotha AK, Kashikar R, Godugu C, Raghuvanshi RS, Singh SB, Srivastava S. Nanomedicine based approaches for combating viral infections. J Control Release 2021; 338:80-104. [PMID: 34375690 PMCID: PMC8526416 DOI: 10.1016/j.jconrel.2021.08.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 08/04/2021] [Accepted: 08/05/2021] [Indexed: 12/12/2022]
Abstract
Millions of people die each year from viral infections across the globe. There is an urgent need to overcome the existing gap and pitfalls of the current antiviral therapy which include increased dose and dosing frequency, bioavailability challenges, non-specificity, incidences of resistance and so on. These stumbling blocks could be effectively managed by the advent of nanomedicine. Current review emphasizes over an enhanced understanding of how different lipid, polymer and elemental based nanoformulations could be potentially and precisely used to bridle the said drawbacks in antiviral therapy. The dawn of nanotechnology meeting vaccine delivery, role of RNAi therapeutics in antiviral treatment regimen, various regulatory concerns towards clinical translation of nanomedicine along with current trends and implications including unexplored research avenues for advancing the current drug delivery have been discussed in detail.
Collapse
Affiliation(s)
- Saurabh Shah
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Mahavir Bhupal Chougule
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, MS, USA; Department Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Arun K Kotha
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, MS, USA; Department Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Rama Kashikar
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, MS, USA; Department Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Chandraiah Godugu
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Rajeev Singh Raghuvanshi
- Indian Pharmacopoeia Commission, Ministry of Health & Family Welfare, Government of India, India
| | - Shashi Bala Singh
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Saurabh Srivastava
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India.
| |
Collapse
|
5
|
Nasirmoghadas P, Mousakhani A, Behzad F, Beheshtkhoo N, Hassanzadeh A, Nikoo M, Mehrabi M, Kouhbanani MAJ. Nanoparticles in cancer immunotherapies: An innovative strategy. Biotechnol Prog 2020; 37:e3070. [PMID: 32829506 DOI: 10.1002/btpr.3070] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/14/2020] [Accepted: 08/20/2020] [Indexed: 12/21/2022]
Abstract
Cancer has been one of the most significant causes of mortality, worldwide. Cancer immunotherapy has recently emerged as a competent, cancer-fighting clinical strategy. Nevertheless, due to the difficulty of such treatments, costs, and off-target adverse effects, the implementation of cancer immunotherapy described by the antigen-presenting cell (APC) vaccine and chimeric antigen receptor T cell therapy ex vivo in large clinical trials have been limited. Nowadays, the nanoparticles theranostic system as a promising target-based modality provides new opportunities to improve cancer immunotherapy difficulties and reduce their adverse effects. Meanwhile, the appropriate engineering of nanoparticles taking into consideration nanoparticle characteristics, such as, size, shape, and surface features, as well as the use of these physicochemical properties for suitable biological interactions, provides new possibilities for the application of nanoparticles in cancer immunotherapy. In this review article, we focus on the latest state-of-the-art nanoparticle-based antigen/adjuvant delivery vehicle strategies to professional APCs and engineering specific T lymphocyte required for improving the efficiency of tumor-specific immunotherapy.
Collapse
Affiliation(s)
- Pourya Nasirmoghadas
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Akbar Mousakhani
- Department of Plant Sciences, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Farahnaz Behzad
- Research Institute for Fundamental Sciences (RIFS), University of Tabriz, Tabriz, Iran
| | - Nasrin Beheshtkhoo
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Hassanzadeh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Marzieh Nikoo
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Helal Iran Pharmaceutical and Clinical Complex, Tehran, Iran
| | - Mohsen Mehrabi
- Department of Medical Nanotechnology, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Mohammad Amin Jadidi Kouhbanani
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Abstract
Mucosal surfaces are the interface between the host’s internal milieu and the external environment, and they have dual functions, serving as physical barriers to foreign antigens and as accepting sites for vital materials. Mucosal vaccines are more favored to prevent mucosal infections from the portal of entry. Although mucosal vaccination has many advantages, licensed mucosal vaccines are scarce. The most widely studied mucosal routes are oral and intranasal. Licensed oral and intranasal vaccines are composed mostly of whole cell killed or live attenuated microorganisms serving as both delivery systems and built-in adjuvants. Future mucosal vaccines should be made with more purified antigen components, which will be relatively less immunogenic. To induce robust protective immune responses against well-purified vaccine antigens, an effective mucosal delivery system is an essential requisite. Recent developments in biomaterials and nanotechnology have enabled many innovative mucosal vaccine trials. For oral vaccination, the vaccine delivery system should be able to stably carry antigens and adjuvants and resist harsh physicochemical conditions in the stomach and intestinal tract. Besides many nano/microcarrier tools generated by using natural and chemical materials, the development of oral vaccine delivery systems using food materials should be more robustly researched to expand vaccine coverage of gastrointestinal infections in developing countries. For intranasal vaccination, the vaccine delivery system should survive the very active mucociliary clearance mechanisms and prove safety because of the anatomical location of nasal cavity separated by a thin barrier. Future mucosal vaccine carriers, regardless of administration routes, should have certain common characteristics. They should maintain stability in given environments, be mucoadhesive, and have the ability to target specific tissues and cells.
Collapse
|
7
|
Dhandapani RK, Gurusamy D, Howell JL, Palli SR. Development of CS-TPP-dsRNA nanoparticles to enhance RNAi efficiency in the yellow fever mosquito, Aedes aegypti. Sci Rep 2019; 9:8775. [PMID: 31217512 PMCID: PMC6584730 DOI: 10.1038/s41598-019-45019-z] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 05/29/2019] [Indexed: 01/13/2023] Open
Abstract
Mosquito-borne diseases are a major threat to human health and are responsible for millions of deaths globally each year. Vector control is one of the most important approaches used in reducing the incidence of these diseases. However, increasing mosquito resistance to chemical insecticides presents challenges to this approach. Therefore, new strategies are necessary to develop the next generation vector control methods. Because of the target specificity of dsRNA, RNAi-based control measures are an attractive alternative to current insecticides used to control disease vectors. In this study, Chitosan (CS) was cross-linked to sodium tripolyphosphate (TPP) to produce nano-sized polyelectrolyte complexes with dsRNA. CS-TPP-dsRNA nanoparticles were prepared by ionic gelation method. The encapsulation efficiency, protection of dsRNA from nucleases, cellular uptake, in vivo biodistribution, larval mortality and gene knockdown efficiency of CS-TPP-dsRNA nanoparticles were determined. The results showed that at a 5:1 weight ratio of CS-TPP to dsRNA, nanoparticles of less than 200 nm mean diameter and a positive surface charge were formed. Confocal microscopy revealed the distribution of the fed CS-TPP-dsRNA nanoparticles in midgut, fat body and epidermis of yellow fever mosquito, Aedes aegypti larvae. Bioassays showed significant mortality of larvae fed on CS-TPP-dsRNA nanoparticles. These assays also showed knockdown of a target gene in CS-TPP-dsRNA nanoparticle fed larvae. These data suggest that CS-TPP nanoparticles may be used for delivery of dsRNA to mosquito larvae.
Collapse
Affiliation(s)
| | - Dhandapani Gurusamy
- Department of Entomology, University of Kentucky, Lexington, Kentucky, 40546, USA
| | - Jeffrey L Howell
- Department of Entomology, University of Kentucky, Lexington, Kentucky, 40546, USA
| | - Subba Reddy Palli
- Department of Entomology, University of Kentucky, Lexington, Kentucky, 40546, USA.
| |
Collapse
|
8
|
Iscaro A, Howard NF, Muthana M. Nanoparticles: Properties and Applications in Cancer Immunotherapy. Curr Pharm Des 2019; 25:1962-1979. [PMID: 31566122 DOI: 10.2174/1381612825666190708214240] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 06/19/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Tumours are no longer regarded as isolated masses of aberrantly proliferating epithelial cells. Rather, their properties depend on complex interactions between epithelial cancer cells and the surrounding stromal compartment within the tumour microenvironment. In particular, leukocyte infiltration plays a role in controlling tumour development and is now considered one of the hallmarks of cancer. Thus, in the last few years, immunotherapy has become a promising strategy to fight cancer, as its goal is to reprogram or activate antitumour immunity to kill tumour cells, without damaging the normal cells and provide long-lasting results where other therapies fail. However, the immune-related adverse events due to the low specificity in tumour cell targeting, strongly limit immunotherapy efficacy. In this regard, nanomedicine offers a platform for the delivery of different immunotherapeutic agents specifically to the tumour site, thus increasing efficacy and reducing toxicity. Indeed, playing with different material types, several nanoparticles can be formulated with different shape, charge, size and surface chemical modifications making them the most promising platform for biomedical applications. AIM In this review, we will summarize the different types of cancer immunotherapy currently in clinical trials or already approved for cancer treatment. Then, we will focus on the most recent promising strategies to deliver immunotherapies directly to the tumour site using nanoparticles. CONCLUSION Nanomedicine seems to be a promising approach to improve the efficacy of cancer immunotherapy. However, additional investigations are needed to minimize the variables in the production processes in order to make nanoparticles suitable for clinical use.
Collapse
Affiliation(s)
- Alessandra Iscaro
- Department of Oncology & Metabolism, University of Sheffield, Medical School, Beech Hill Road, Sheffield, United Kingdom
| | - Nutter F Howard
- Department of Oncology & Metabolism, University of Sheffield, Medical School, Beech Hill Road, Sheffield, United Kingdom
| | - Munitta Muthana
- Department of Oncology & Metabolism, University of Sheffield, Medical School, Beech Hill Road, Sheffield, United Kingdom
| |
Collapse
|
9
|
Saliba H, Heurtault B, Bouharoun-Tayoun H, Flacher V, Frisch B, Fournel S, Chamat S. Enhancing tumor specific immune responses by transcutaneous vaccination. Expert Rev Vaccines 2017; 16:1079-1094. [DOI: 10.1080/14760584.2017.1382357] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Hanadi Saliba
- Laboratory of Design and Application of Bioactive Molecules, University of Strasbourg, Illkirch Cedex, France
- Laboratory of Immunology, Lebanese University, Fanar, Lebanon
| | - Béatrice Heurtault
- Laboratory of Design and Application of Bioactive Molecules, University of Strasbourg, Illkirch Cedex, France
| | | | - Vincent Flacher
- Laboratory of Immunopathology and Therapeutic Chemistry, Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
| | - Benoît Frisch
- Laboratory of Design and Application of Bioactive Molecules, University of Strasbourg, Illkirch Cedex, France
| | - Sylvie Fournel
- Laboratory of Design and Application of Bioactive Molecules, University of Strasbourg, Illkirch Cedex, France
| | - Soulaima Chamat
- Laboratory of Immunology, Lebanese University, Fanar, Lebanon
- Faculty of Medicine, Lebanese University, Hadath, Lebanon
| |
Collapse
|
10
|
Song W, Musetti SN, Huang L. Nanomaterials for cancer immunotherapy. Biomaterials 2017; 148:16-30. [PMID: 28961532 DOI: 10.1016/j.biomaterials.2017.09.017] [Citation(s) in RCA: 198] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Revised: 09/07/2017] [Accepted: 09/17/2017] [Indexed: 02/08/2023]
Abstract
Cancer immunotherapy is quickly growing to be the fourth most important cancer therapy, after surgery, radiation therapy, and chemotherapy. Immunotherapy is the most promising cancer management strategy because it orchestrates the body's own immune system to target and eradicate cancer cells, which may result in durable antitumor responses and reduce metastasis and recurrence more than traditional treatments. Nanomaterials hold great promise in further improving the efficiency of cancer immunotherapy - in many cases, they are even necessary for effective delivery. In this review, we briefly summarize the basic principles of cancer immunotherapy and explain why and where to apply nanomaterials in cancer immunotherapy, with special emphasis on cancer vaccines and tumor microenvironment modulation.
Collapse
Affiliation(s)
- Wantong Song
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China
| | - Sara N Musetti
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Leaf Huang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
11
|
Grimaldi AM, Incoronato M, Salvatore M, Soricelli A. Nanoparticle-based strategies for cancer immunotherapy and immunodiagnostics. Nanomedicine (Lond) 2017; 12:2349-2365. [PMID: 28868980 DOI: 10.2217/nnm-2017-0208] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Although recent successes in clinical trials are strengthening research focused on cancer immunology, the poor immunogenicity and off-target side effects of immunotherapeutics remain major challenges in translating these promising approaches to clinically feasible therapies in the treatment of a large range of tumors. Nanotechnology offers target-based approaches, which have shown significant improvements in the rapidly advancing field of cancer immunotherapy. Here, we first discuss the chemical and physical features of nanoparticulate systems that can be tuned to address the anticancer immune response, and then review recent, key examples of the exploited strategies, ranging from nanovaccines to NPs revising the tumor immunosuppressive microenvironment, up to immunotherapeutic multimodal NPs. Finally, the paper concludes by identifying the promising and outstanding challenges the field of emerging nanotechnologies is facing for cancer immunotherapy.
Collapse
Affiliation(s)
| | | | | | - Andrea Soricelli
- IRCCS SDN, Via Gianturco 113, 80143, Naples, Italy.,Department of Motor Sciences & Healthiness, University of Naples Parthenope, via Medina 40, 80133, Naples, Italy
| |
Collapse
|
12
|
Gunay G, Sardan Ekiz M, Ferhati X, Richichi B, Nativi C, Tekinay AB, Guler MO. Antigenic GM3 Lactone Mimetic Molecule Integrated Mannosylated Glycopeptide Nanofibers for the Activation and Maturation of Dendritic Cells. ACS APPLIED MATERIALS & INTERFACES 2017; 9:16035-16042. [PMID: 28445638 DOI: 10.1021/acsami.7b04094] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The ability of dendritic cells to coordinate innate and adaptive immune responses makes them essential targets for vaccination strategies. Presentation of specific antigens by dendritic cells is required for the activation of the immune system against many pathogens and tumors, and nanoscale materials can be functionalized for active targeting of dendritic cells. In this work, we integrated an immunogenic, carbohydrate melanoma-associated antigen-mimetic GM3-lactone molecule into mannosylated peptide amphiphile nanofibers to target dendritic cells through DC-SIGN receptor. Based on morphological and functional analyses, when dendritic cells were treated with peptide nanofiber carriers, they showed significant increase in antigen internalization and a corresponding increase in the surface expression of the activation and maturation markers CD86, CD83 and HLA-DR, in addition to exhibiting a general morphology consistent with dendritic cell maturation. These results indicate that mannosylated peptide amphiphile nanofiber carriers are promising candidates to target dendritic cells for antigen delivery.
Collapse
Affiliation(s)
- Gokhan Gunay
- Institute of Materials Science and Nanotechnology, National Nanotechnology Research Center (UNAM), Bilkent University , Ankara 06800, Turkey
- Neuroscience Graduate Program, Bilkent University , Ankara 06800, Turkey
| | - Melis Sardan Ekiz
- Institute of Materials Science and Nanotechnology, National Nanotechnology Research Center (UNAM), Bilkent University , Ankara 06800, Turkey
| | - Xhenti Ferhati
- Department of Chemistry "Ugo Schiff", University of Florence , Sesto Fiorentino, Florence 50019, Italy
| | - Barbara Richichi
- Department of Chemistry "Ugo Schiff", University of Florence , Sesto Fiorentino, Florence 50019, Italy
| | - Cristina Nativi
- Department of Chemistry "Ugo Schiff", University of Florence , Sesto Fiorentino, Florence 50019, Italy
| | - Ayse B Tekinay
- Institute of Materials Science and Nanotechnology, National Nanotechnology Research Center (UNAM), Bilkent University , Ankara 06800, Turkey
- Neuroscience Graduate Program, Bilkent University , Ankara 06800, Turkey
| | - Mustafa O Guler
- Institute of Materials Science and Nanotechnology, National Nanotechnology Research Center (UNAM), Bilkent University , Ankara 06800, Turkey
- Institute for Molecular Engineering, University of Chicago , Chicago, Illinois 60637, United States
| |
Collapse
|
13
|
von Roemeling C, Jiang W, Chan CK, Weissman IL, Kim BY. Breaking Down the Barriers to Precision Cancer Nanomedicine. Trends Biotechnol 2017; 35:159-171. [DOI: 10.1016/j.tibtech.2016.07.006] [Citation(s) in RCA: 170] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 07/01/2016] [Accepted: 07/15/2016] [Indexed: 02/04/2023]
|
14
|
Tuettenberg A, Steinbrink K, Schuppan D. Myeloid cells as orchestrators of the tumor microenvironment: novel targets for nanoparticular cancer therapy. Nanomedicine (Lond) 2016; 11:2735-2751. [DOI: 10.2217/nnm-2016-0208] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Macrophages, myeloid-derived suppressor cells and tolerogenic dendritic cells are central players of a heterogeneous myeloid cell population, with the ability to suppress innate and adaptive immune responses and thus to promote tumor growth. Their influx and local proliferation are mainly induced by the cancers themselves, and their numbers in the tumor microenvironment and the peripheral blood correlate with decreased survival. Therapeutic targeting these innate immune cells, either aiming at their elimination or polarization toward tumor suppressive cells is an attractive novel approach to control tumor progression and block metastasis. We review the current understanding of cancer immunology including immune surveillance and immune editing in the context of these prominent innate suppressor cells, and their targetability by nanoparticular immunotherapy with small molecules or siRNA.
Collapse
Affiliation(s)
- Andrea Tuettenberg
- Department of Dermatology & Research Center for Immunotherapy (FZI) University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Kerstin Steinbrink
- Department of Dermatology & Research Center for Immunotherapy (FZI) University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Detlef Schuppan
- Institute of Translational Immunology & Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg-University, Mainz, Germany
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
15
|
Glass JJ, Kent SJ, De Rose R. Enhancing dendritic cell activation and HIV vaccine effectiveness through nanoparticle vaccination. Expert Rev Vaccines 2016; 15:719-29. [PMID: 26783186 DOI: 10.1586/14760584.2016.1141054] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Novel vaccination approaches are needed to prevent and control human immunodeficiency virus (HIV) infection. A growing body of literature demonstrates the potential of nanotechnology to modulate the human immune system and generate targeted, controlled immune responses. In this Review, we summarize important advances in how 'nanovaccinology' can be used to develop safe and effective vaccines for HIV. We highlight the central role of dendritic cells in the immune response to vaccination and describe how nanotechnology can be used to enhance delivery to and activation of these important antigen-presenting cells. Strategies employed to improve biodistribution are discussed, including improved lymph node delivery and mucosal penetration concepts, before detailing methods to enhance the humoral and/or cellular immune response to vaccines. We conclude with a commentary on the current state of nanovaccinology.
Collapse
Affiliation(s)
- Joshua J Glass
- a ARC Centre of Excellence in Convergent Bio-Nano Science and Technology , The University of Melbourne , Melbourne , Australia.,b Department of Microbiology and Immunology , Peter Doherty Institute for Infection and Immunity, The University of Melbourne , Melbourne , Australia
| | - Stephen J Kent
- a ARC Centre of Excellence in Convergent Bio-Nano Science and Technology , The University of Melbourne , Melbourne , Australia.,b Department of Microbiology and Immunology , Peter Doherty Institute for Infection and Immunity, The University of Melbourne , Melbourne , Australia.,c Melbourne Sexual Health Centre and Department of Infectious Diseases , Alfred Health, Central Clinical School, Monash University , Melbourne , Australia
| | - Robert De Rose
- a ARC Centre of Excellence in Convergent Bio-Nano Science and Technology , The University of Melbourne , Melbourne , Australia.,b Department of Microbiology and Immunology , Peter Doherty Institute for Infection and Immunity, The University of Melbourne , Melbourne , Australia
| |
Collapse
|
16
|
Silva AL, Soema PC, Slütter B, Ossendorp F, Jiskoot W. PLGA particulate delivery systems for subunit vaccines: Linking particle properties to immunogenicity. Hum Vaccin Immunother 2016; 12:1056-69. [PMID: 26752261 PMCID: PMC4962933 DOI: 10.1080/21645515.2015.1117714] [Citation(s) in RCA: 176] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Among the emerging subunit vaccines are recombinant protein- and synthetic peptide-based vaccine formulations. However, proteins and peptides have a low intrinsic immunogenicity. A common strategy to overcome this is to co-deliver (an) antigen(s) with (an) immune modulator(s) by co-encapsulating them in a particulate delivery system, such as poly(lactic-co-glycolic acid) (PLGA) particles. Particulate PLGA formulations offer many advantages for antigen delivery as they are biocompatible and biodegradable; can protect the antigens from degradation and clearance; allow for co-encapsulation of antigens and immune modulators; can be targeted to antigen presenting cells; and their particulate nature can increase uptake and cross-presentation by mimicking the size and shape of an invading pathogen. In this review we discuss the pros and cons of using PLGA particulate formulations for subunit vaccine delivery and provide an overview of formulation parameters that influence their adjuvanticity and the ensuing immune response.
Collapse
Affiliation(s)
- A L Silva
- a Division of Drug Delivery Technology , Leiden Academic Center for Drug Research, Leiden University , Leiden , The Netherlands
| | - P C Soema
- b Intravacc (Institute for Translational Vaccinology) , Bilthoven , The Netherlands
| | - B Slütter
- a Division of Drug Delivery Technology , Leiden Academic Center for Drug Research, Leiden University , Leiden , The Netherlands.,c Cluster BioTherapeutics, Leiden Academic Center for Drug Research, Leiden University , Leiden , The Netherlands
| | - F Ossendorp
- d Department of Immunohematology and Blood Transfusion , Leiden University Medical Center , Leiden , The Netherlands
| | - W Jiskoot
- a Division of Drug Delivery Technology , Leiden Academic Center for Drug Research, Leiden University , Leiden , The Netherlands
| |
Collapse
|
17
|
Huh JB, Kim JH, Kim S, Lee SH, Shim KM, Kim SE, Kang SS, Jeong CM. Effects of PMMA and Cross-Linked Dextran Filler for Soft Tissue Augmentation in Rats. Int J Mol Sci 2015; 16:28523-33. [PMID: 26633376 PMCID: PMC4691059 DOI: 10.3390/ijms161226112] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 10/27/2015] [Accepted: 11/20/2015] [Indexed: 11/23/2022] Open
Abstract
This study was conducted for evaluation of the ability to maintain efficacy and biocompatibility of cross-linked dextran in hydroxypropyl methylcellulose (DiHM) and cross-linked dextran mixed with PMMA in hydroxypropyl methylcellulose (PDiHM), compared with hyaluronic acid (HA) filler. Saline and HA solution was administered in the negative and positive control groups, and DiHM and PDiHM were administered in the test groups (n = 10 in each group). The site of cranial subcutaneous injection was the mid-point of the interpupillary line, and the site of intraoral submucosal injection was the ridge crest 2 mm below the cervical line of the mandibular left incisor. Before and immediately after filler injection, intraoral photos and lateral cephalometric radiographs were taken for analysis and comparison of the effect of the filler on the injection sites. The filler injected areas were converted into sequential size changes (%) of the baseline. Histomorphologic examination was performed after 12 weeks. The smallest value in the filler injected area was observed during the experimental period in the normal saline group (p < 0.001), which was almost absorbed at 4 weeks (7.19% ± 12.72%). The HA group exhibited a steady decrease in sequential size and showed a lower value than the DiHM and PDiHM groups (saline < HA < DHiM, PDHiM, p < 0.001). DiHM and PDiHM tended to increase for the first 4 weeks and later decreased until 12 weeks. In this study on DiHM and PDiHM, there was no histological abnormality in cranial skin and oral mucosa. DiHM and PDiHM filler materials with injection system provide an excellent alternative surgical method for use in oral and craniofacial fields.
Collapse
Affiliation(s)
- Jung-Bo Huh
- Department of Prosthodontics, Dental Research Institute, School of Dentistry, Pusan National University, YangSan 676-870, Korea.
| | - Joo-Hyun Kim
- Department of Prosthodontics, Dental Research Institute, School of Dentistry, Pusan National University, YangSan 676-870, Korea.
| | - Soyun Kim
- School of Dentistry, Pusan National University, YangSan 676-870, Korea.
| | - So-Hyoun Lee
- Department of Prosthodontics, Dental Research Institute, School of Dentistry, Pusan National University, YangSan 676-870, Korea.
| | - Kyung Mi Shim
- Department of Veterinary Surgery, College of Veterinary Medicine, Chonnam National University, Gwangju 500-757, Korea.
| | - Se Eun Kim
- Department of Veterinary Surgery, College of Veterinary Medicine, Chonnam National University, Gwangju 500-757, Korea.
| | - Seong Soo Kang
- Department of Veterinary Surgery, College of Veterinary Medicine, Chonnam National University, Gwangju 500-757, Korea.
| | - Chang-Mo Jeong
- Department of Prosthodontics, Dental Research Institute, School of Dentistry, Pusan National University, YangSan 676-870, Korea.
| |
Collapse
|
18
|
Investigation of the stabilizer elimination during the washing step of charged PLGA microparticles utilizing a novel HPLC-UV-ELSD method. Eur J Pharm Biopharm 2015; 94:468-72. [DOI: 10.1016/j.ejpb.2015.06.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 05/28/2015] [Accepted: 06/07/2015] [Indexed: 11/24/2022]
|
19
|
Phanse Y, Dunphy BM, Perry JL, Airs PM, Paquette CCH, Carlson JO, Xu J, Luft JC, DeSimone JM, Beaty BJ, Bartholomay LC. Biodistribution and Toxicity Studies of PRINT Hydrogel Nanoparticles in Mosquito Larvae and Cells. PLoS Negl Trop Dis 2015; 9:e0003735. [PMID: 25996390 PMCID: PMC4440723 DOI: 10.1371/journal.pntd.0003735] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 04/02/2015] [Indexed: 12/20/2022] Open
Abstract
Mosquito-borne diseases continue to remain major threats to human and animal health and impediments to socioeconomic development. Increasing mosquito resistance to chemical insecticides is a great public health concern, and new strategies/technologies are necessary to develop the next-generation of vector control tools. We propose to develop a novel method for mosquito control that employs nanoparticles (NPs) as a platform for delivery of mosquitocidal dsRNA molecules to silence mosquito genes and cause vector lethality. Identifying optimal NP chemistry and morphology is imperative for efficient mosquitocide delivery. Toward this end, fluorescently labeled polyethylene glycol NPs of specific sizes, shapes (80 nm x 320 nm, 80 nm x 5000 nm, 200 nm x 200 nm, and 1000 nm x 1000 nm) and charges (negative and positive) were fabricated by Particle Replication in Non-Wetting Templates (PRINT) technology. Biodistribution, persistence, and toxicity of PRINT NPs were evaluated in vitro in mosquito cell culture and in vivo in Anopheles gambiae larvae following parenteral and oral challenge. Following parenteral challenge, the biodistribution of the positively and negatively charged NPs of each size and shape was similar; intense fluorescence was observed in thoracic and abdominal regions of the larval body. Positively charged NPs were more associated with the gastric caeca in the gastrointestinal tract. Negatively charged NPs persisted through metamorphosis and were observed in head, body and ovaries of adults. Following oral challenge, NPs were detected in the larval mid- and hindgut. Positively charged NPs were more efficiently internalized in vitro than negatively charged NPs. Positively charged NPs trafficked to the cytosol, but negatively charged NPs co-localized with lysosomes. Following in vitro and in vivo challenge, none of the NPs tested induced any cytotoxic effects.
Collapse
Affiliation(s)
- Yashdeep Phanse
- Department of Entomology, Iowa State University, Ames, Iowa, United States of America
| | - Brendan M. Dunphy
- Department of Entomology, Iowa State University, Ames, Iowa, United States of America
| | - Jillian L. Perry
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, North Carolina, United States of America
| | - Paul M. Airs
- Department of Entomology, Iowa State University, Ames, Iowa, United States of America
| | - Cynthia C. H. Paquette
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Jonathan O. Carlson
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Jing Xu
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, North Carolina, United States of America
| | - J. Christopher Luft
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, North Carolina, United States of America
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Joseph M. DeSimone
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, North Carolina, United States of America
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Institute for Nanomedicine and Institute for Advanced Materials, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, United States of America
- Sloan-Kettering Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Barry J. Beaty
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Lyric C. Bartholomay
- Department of Entomology, Iowa State University, Ames, Iowa, United States of America
- * E-mail:
| |
Collapse
|
20
|
Briggs T, Matos J, Collins G, Arinzeh TL. Evaluating protein incorporation and release in electrospun composite scaffolds for bone tissue engineering applications. J Biomed Mater Res A 2015; 103:3117-27. [DOI: 10.1002/jbm.a.35444] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 02/06/2015] [Accepted: 02/19/2015] [Indexed: 11/11/2022]
Affiliation(s)
- Tonye Briggs
- Department of Biomedical Engineering; New Jersey Institute of Technology; Newark New Jersey 07102
| | - Jeffrey Matos
- Department of Biomedical Engineering; New Jersey Institute of Technology; Newark New Jersey 07102
| | - George Collins
- Department of Biomedical Engineering; New Jersey Institute of Technology; Newark New Jersey 07102
| | | |
Collapse
|
21
|
Vera M, Barcia E, Negro S, Marcianes P, García-García L, Slowing K, Fernández-Carballido A. New celecoxib multiparticulate systems to improve glioblastoma treatment. Int J Pharm 2014; 473:518-27. [DOI: 10.1016/j.ijpharm.2014.07.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 07/21/2014] [Accepted: 07/22/2014] [Indexed: 10/25/2022]
|
22
|
Chitosan as an adjuvant-like substrate for dendritic cell culture to enhance antitumor effects. Biomaterials 2014; 35:8867-8875. [DOI: 10.1016/j.biomaterials.2014.07.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 07/10/2014] [Indexed: 11/22/2022]
|
23
|
Vasiliev YM. Chitosan-based vaccine adjuvants: incomplete characterization complicates preclinical and clinical evaluation. Expert Rev Vaccines 2014; 14:37-53. [PMID: 25262982 DOI: 10.1586/14760584.2015.956729] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
A number of preclinical and clinical studies with chitosan-adjuvanted antigen- and DNA-based vaccines have been carried out. Various chitosans and their modifications, in different forms (solutions, powders, gels and particles), have been evaluated with various antigens administered via different routes. Chitosan is a generic name for a wide array of glucosamine-based substances derived from biological sources, and standardization is necessary. However, in most of the studies published to date, molecular weight, viscosity, deacetylation degree and/or purity level (especially endotoxins) are not provided for the initial chitosan substance and/or final formulation and the preparation procedure is not detailed. Evaluation of adjuvant properties is challenging, given that the only available data are insufficient to demonstrate immunogenicity for chitosans with characteristics within certain intervals to elucidate mechanisms of action or to exclude impurities as the active substance. These and other issues of chitosan-based vaccine adjuvants are summarized and a step-by-step evaluation approach for chitosan-based vaccine adjuvants is outlined.
Collapse
Affiliation(s)
- Yuri M Vasiliev
- Mechnikov Research Institute of Vaccines and Sera, M. Kazeny lane, 5a, Moscow, 105064, Russian Federation
| |
Collapse
|
24
|
Sionkowska A, Płanecka A, Lewandowska K, Michalska M. The influence of UV-irradiation on thermal and mechanical properties of chitosan and silk fibroin mixtures. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2014; 140:301-5. [PMID: 25218587 DOI: 10.1016/j.jphotobiol.2014.08.017] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 07/29/2014] [Accepted: 08/24/2014] [Indexed: 11/29/2022]
Abstract
In the present paper the results regarding the influence of UV-irradiation with 254 nm wavelength on the thermal and mechanical properties and the intrinsic viscosity of chitosan/silk fibroin mixtures are presented. The mixture of chitosan and silk fibroin in solution and thin films made of chitosan/silk fibroin mixture obtained by solvent evaporation were submitted to the treatment with UV irradiation (wavelength 254 nm) for different time intervals. Mechanical properties of thin films made of chitosan/silk fibroin blends before and after UV-irradiation have been investigated by mechanical testing machine and compared with mechanical properties of chitosan films. The changes in such mechanical properties as ultimate breaking strength, percentage of elongation at break and Young Modulus have been investigated. The results have shown, that the mechanical properties of the blends were greatly affected by time of exposure to UV irradiation. Ultimate tensile strength and ultimate percentage of elongation decreased after UV irradiation of the blend. Increasing UV irradiation led to the decrease in Young's Modulus of the chitosan/silk fibroin blend. Viscosity of chitosan/silk fibroin mixtures decreased after UV-irradiation. Thermal properties of the mixtures have been only slightly altered by UV-irradiation.
Collapse
Affiliation(s)
- Alina Sionkowska
- Nicolaus Copernicus University, Faculty of Chemistry, Biomaterials and Cosmetics Department, ul. Gagarina 7, 87-100 Toruń, Poland.
| | - Anna Płanecka
- Nicolaus Copernicus University, Faculty of Chemistry, Biomaterials and Cosmetics Department, ul. Gagarina 7, 87-100 Toruń, Poland
| | - Katarzyna Lewandowska
- Nicolaus Copernicus University, Faculty of Chemistry, Biomaterials and Cosmetics Department, ul. Gagarina 7, 87-100 Toruń, Poland
| | - Marta Michalska
- Nicolaus Copernicus University, Faculty of Chemistry, Biomaterials and Cosmetics Department, ul. Gagarina 7, 87-100 Toruń, Poland
| |
Collapse
|
25
|
Kansal S, Tandon R, Verma A, Misra P, Choudhary AK, Verma R, Verma PRP, Dube A, Mishra PR. Coating doxorubicin-loaded nanocapsules with alginate enhances therapeutic efficacy against Leishmania in hamsters by inducing Th1-type immune responses. Br J Pharmacol 2014; 171:4038-50. [PMID: 24837879 DOI: 10.1111/bph.12754] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 04/09/2014] [Accepted: 04/23/2014] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE The aim of the present study was to evaluate the immunomodulatory and chemotherapeutic potential of alginate-(SA) coated nanocapsule (NCs) loaded with doxorubicin (SA-NCs-DOX) against visceral leishmaniasis in comparison with nano-emulsions containing doxorubicin (NE-DOX). EXPERIMENTAL APPROACH NE-DOX was prepared using low-energy emulsification methods. Stepwise addition of protamine sulphate and SA in a layer-by-layer manner was used to form SA-NCs-DOX. SA-NCs-DOX, NE-DOX and Free DOX were compared for their cytotoxicity against Leishmania donovani-infected macrophages in vitro and generation of T-cell responses in infected hamsters in vivo. KEY RESULTS Size and ζ potential of the NE-DOX and SA-NCs-DOX formulations were 310 ± 2.1 nm and (-)32.6 ± 2.1 mV, 342 ± 4.1 nm and (-)29.3 ± 1.2 mV respectively. SA-NCs-DOX was better (1.5 times) taken up by J774A.1 macrophages compared with NE-DOX. SA-NCs -DOX showed greater efficacy than NE-DOX against intramacrophagic amastigotes. SA-NCs-DOX treatment exhibited enhanced apoptotic efficiency than NE-DOX and free DOX as evident by cell cycle analysis, decrease in mitochondrial membrane potential, ROS and NO production. T-cell responses, when assessed through lymphoproliferative responses, NO production along with enhanced levels of iNOS, TNF-α, IFN-γ and IL-12 were found to be up-regulated after SA-NCs-DOX, compared with responses to NE-DOX in vivo. Parasitic burden was decreased in Leishmania-infected hamsters treated with SA-NCs-DOX, compared with NE-DOX. CONCLUSIONS AND IMPLICATIONS Our results provide insights into the development of an alternative approach to improved management of leishmaniasis through a combination of chemotherapy with stimulation of the innate immune system.
Collapse
Affiliation(s)
- S Kansal
- Pharmaceutics Division, CSIR-Central Drug Research Institute, BS-10/1 sector-10 Jankipuram Extension, Lucknow, India
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Bhargava A, Bunkar N, Khare NK, Mishra D, Mishra PK. Nanoengineered strategies to optimize dendritic cells for gastrointestinal tumor immunotherapy: from biology to translational medicine. Nanomedicine (Lond) 2014; 9:2187-2202. [PMID: 25405796 DOI: 10.2217/nnm.14.115] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Nanomedicine may play an important role in improving the clinical efficacy of dendritic cell-based immunotherapy against GI tract malignancies. Dendritic cell-based vaccines have proven their effectiveness against different established GI tract tumors, yet their success is mainly hindered by the strong tumor-induced suppressive microenvironment. The sustained and targeted release of tumor antigens to dendritic cells using different nanoengineered approaches would be an efficient strategy to overcome established immune tolerance. Encapsulation would result in low diffusivity, restricted movement, effective crosspresentation and enhanced T-cell responses. These nanotherapy-based approaches will certainly help with the designing of clinically translatable dendritic cell-based therapeutic vaccines and facilitate the selective removal of residual disease in gastrointestinal cancer patients following standard treatments.
Collapse
Affiliation(s)
- Arpit Bhargava
- Translational Research Laboratory, School of Biological Sciences, Dr H. S. Gour Central University, Sagar, India
| | | | | | | | | |
Collapse
|
27
|
Chen X, Liu Y, Wang L, Liu Y, Zhang W, Fan B, Ma X, Yuan Q, Ma G, Su Z. Enhanced Humoral and Cell-Mediated Immune Responses Generated by Cationic Polymer-Coated PLA Microspheres with Adsorbed HBsAg. Mol Pharm 2014; 11:1772-84. [DOI: 10.1021/mp400597z] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Xiaoming Chen
- National
Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China
- Graduated University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Yuying Liu
- College
of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, PR China
| | - Lianyan Wang
- National
Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Yuan Liu
- National
Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China
- Graduated University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Weifeng Zhang
- National
Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China
- Graduated University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Bei Fan
- Hualan Biological Engineering Inc., Henan 453003, PR China
| | - Xiaowei Ma
- Hualan Biological Engineering Inc., Henan 453003, PR China
| | - Qipeng Yuan
- College
of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, PR China
| | - Guanghui Ma
- National
Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Zhiguo Su
- National
Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China
| |
Collapse
|
28
|
Neffe AT, Wischke C, Racheva M, Lendlein A. Progress in biopolymer-based biomaterials and their application in controlled drug delivery. Expert Rev Med Devices 2014; 10:813-33. [DOI: 10.1586/17434440.2013.839209] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
29
|
|
30
|
Hotaling NA, Ratner DM, Cummings RD, Babensee JE. Presentation Modality of Glycoconjugates Modulates Dendritic Cell Phenotype. Biomater Sci 2014; 2:1426-1439. [PMID: 26146546 DOI: 10.1039/c4bm00138a] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The comparative dendritic cell (DC) response to glycoconjugates presented in soluble, phagocytosable, or non-phagocytosable display modalities is poorly understood. This is particularly problematic, as the probing of immobilized glycans presented on the surface of microarrays is a common screen for potential candidates for glycan-based therapeutics. However, the assumption that carbohydrate-protein interactions on a flat surface can be translatable to development of efficacious therapies, such as vaccines, which are delivered in soluble or phagocytosable particles, has not been validated. Thus, a preliminary investigation was performed in which mannose or glucose was conjugated to cationized bovine serum albumin and presented to DCs in soluble, phagocytosable, or non-phagocytosable display modalities. The functional DC response to the glycoconjugates was assessed via a high throughput assay. Dendritic cell phenotypic outcomes were placed into a multivariate, general linear model (GLM) and shown to be statistically different amongst display modalities when comparing similar surface areas. The GLM showed that glycoconjugates that were adsorbed to wells were the most pro-inflammatory while soluble conjugates were the least. DC interactions with mannose conjugates were found to be calcium dependent and could be inhibited via anti-DC-SIGN antibodies. The results of this study aim to resolve conflicts in reports from multiple laboratories showing differential DC profiles in response to similar, if not identical, ligands delivered via different modalities. Additionally, this study begins to bridge the gap between microarray binding data and functional cell responses by highlighting the phenotypes induced from adsorbed glycoconjugates as compared to those in solution or displayed on microparticles.
Collapse
Affiliation(s)
- N A Hotaling
- Wallace H. Coulter Dept. of Biomedical Engineering, Georgia Institute of Technology, Atlanta GA, 30332
| | - D M Ratner
- Dept. of Bioengineering, University of Washington, Seattle WA, 98195
| | - R D Cummings
- Dept. of Biochemistry, Emory University, Atlanta GA 30322
| | - J E Babensee
- Wallace H. Coulter Dept. of Biomedical Engineering, Georgia Institute of Technology, Atlanta GA, 30332
| |
Collapse
|
31
|
Phanse Y, Carrillo-Conde BR, Ramer-Tait AE, Roychoudhury R, Pohl NLB, Narasimhan B, Wannemuehler MJ, Bellaire BH. Functionalization of polyanhydride microparticles with di-mannose influences uptake by and intracellular fate within dendritic cells. Acta Biomater 2013; 9:8902-9. [PMID: 23796408 DOI: 10.1016/j.actbio.2013.06.024] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 04/30/2013] [Accepted: 06/14/2013] [Indexed: 12/18/2022]
Abstract
Innovative vaccine delivery platforms can facilitate the development of effective single-dose treatment regimens to control emerging and re-emerging infectious diseases. Polyanhydride microparticles are promising vaccine delivery vehicles due to their ability to stably maintain antigens, provide tailored release kinetics and function as adjuvants. A major obstacle for the use of microparticle-based vaccines, however, is their limited uptake by dendritic cells (DCs). In this study, we functionalized the microparticle surface with di-mannose in order to target C-type lectin receptors (CLRs) on DCs. Polyanhydride particles based on sebacic acid (SA), 1,6-bis(p-carboxyphenoxy)hexane (CPH) and 1,8-bis(p-carboxyphenoxy)-3,6-dioxaoctane (CPTEG) were evaluated. Co-incubation of di-mannose-functionalized microparticles up-regulated the expression of CLRs on DCs. More importantly, di-mannose functionalization increased the uptake, as measured by the percentage of cells internalizing particles. The uptake of CPH:SA microparticles increased ∼20-fold, from 0.82% (non-functionalized) to 20.2%, and internalization of CPTEG:CPH microparticles increased ∼7-fold from 1.35% (non-functionalized) to 9.3% upon di-mannose functionalization. Both di-mannose-functionalized and non-functionalized particles trafficked to lysosomes. Together, these studies demonstrate that employing rational vaccine design principles, such as the targeting of CLRs on antigen-presenting cells, can enhance delivery of encapsulated antigens and potentially induce a more robust adaptive immune response.
Collapse
Affiliation(s)
- Yashdeep Phanse
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA 50011, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Hafner AM, Corthésy B, Merkle HP. Particulate formulations for the delivery of poly(I:C) as vaccine adjuvant. Adv Drug Deliv Rev 2013; 65:1386-99. [PMID: 23751781 DOI: 10.1016/j.addr.2013.05.013] [Citation(s) in RCA: 188] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 05/30/2013] [Accepted: 05/31/2013] [Indexed: 12/18/2022]
Abstract
Current research and development of antigens for vaccination often center on purified recombinant proteins, viral subunits, synthetic oligopeptides or oligosaccharides, most of them suffering from being poorly immunogenic and subject to degradation. Hence, they call for efficient delivery systems and potent immunostimulants, jointly denoted as adjuvants. Particulate delivery systems like emulsions, liposomes, nanoparticles and microspheres may provide protection from degradation and facilitate the co-formulation of both the antigen and the immunostimulant. Synthetic double-stranded (ds) RNA, such as polyriboinosinic acid-polyribocytidylic acid, poly(I:C), is a mimic of viral dsRNA and, as such, a promising immunostimulant candidate for vaccines directed against intracellular pathogens. Poly(I:C) signaling is primarily dependent on Toll-like receptor 3 (TLR3), and on melanoma differentiation-associated gene-5 (MDA-5), and strongly drives cell-mediated immunity and a potent type I interferon response. However, stability and toxicity issues so far prevented the clinical application of dsRNAs as they undergo rapid enzymatic degradation and bear the potential to trigger undue immune stimulation as well as autoimmune disorders. This review addresses these concerns and suggests strategies to improve the safety and efficacy of immunostimulatory dsRNA formulations. The focus is on technological means required to lower the necessary dosage of poly(I:C), to target surface-modified microspheres passively or actively to antigen-presenting cells (APCs), to control their interaction with non-professional phagocytes and to modulate the resulting cytokine secretion profile.
Collapse
|
33
|
Mishra DK, Dhote V, Mishra PK. Transdermal immunization: biological framework and translational perspectives. Expert Opin Drug Deliv 2013; 10:183-200. [PMID: 23256860 DOI: 10.1517/17425247.2013.746660] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
INTRODUCTION The renaissance in drug delivery research during the past decade led to several new approaches toward vaccine development. Transdermal immunization (TI) is a promising modality with both practical and immunological merits. Compared with conventional routes of administration, this needle-free delivery approach with ability to target the rich immunologically milieu of the skin provides a dual-edged benefit. It not only elicits an effective immune response in both systemic and mucosal compartments but has the potential to make vaccine delivery more equitable, safer and efficient. AREAS COVERED Over the years, numerous studies have explored physical, chemical and nanocarrier-based strategies to develop vaccines using this attractive route of delivery. The review provides insight into the various facets including research at interface that might drive novel basic scientific ideas to translational outcomes. EXPERT OPINION As we continue to develop TI as a vaccine delivery method, it is important to consider the practical application of this method and device strategies that best fit the public health needs. In the authors' view, nanoengineering-based approaches holds a great promise to overcome the associated challenges in TI and might help to translate early laboratory successes into the development of effective clinical prophylactics.
Collapse
Affiliation(s)
- Dinesh Kumar Mishra
- Guru Ghasidas Central University, SLT Institute of Pharmaceutical Sciences, Department of Pharmaceutics, Bilaspur (CG) 495009, India.
| | | | | |
Collapse
|
34
|
Kuo YC, Wang CC. Cationic solid lipid nanoparticles with primary and quaternary amines for release of saquinavir and biocompatibility with endothelia. Colloids Surf B Biointerfaces 2013; 101:101-5. [DOI: 10.1016/j.colsurfb.2012.06.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Revised: 06/01/2012] [Accepted: 06/06/2012] [Indexed: 11/16/2022]
|
35
|
Adjuvant Effect of Microencapsulated NOD Ligands Studied in a Human Phagocytic Cell Line. J Appl Biomater Funct Mater 2012; 10:229-36. [DOI: 10.5301/jabfm.2012.10438] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/03/2012] [Indexed: 11/20/2022] Open
Abstract
Background Modern subunit vaccines, which are of high purity compared with traditional vaccines, are often incapable of inducing strong immune responses as necessary to build an immunological memory. The desired level of immune response can be achieved only by codelivering immune-modulating agents along with the antigenic epitopes present in these high-purity formulations. This study aimed to explore the adjuvant effect of nucleotide oligomerization domain (nod) receptor agonists as immunomodulators encapsulated in polymeric microparticles as carriers. Methods Microparticles (MP) prepared from poly[(rac-lactide)-co-glycolide] (PLGA) (Mn = 5 kDa, PD = 3.2) by the water-in-oil-in-water (w/o/w) emulsion/solvent evaporation technique were characterized in terms of size, surface morphology, payload and endotoxin content. As NOD agonists, N-acetylmuramyl–L-alanyl–D-isoglutamine (MDP; NOD 2) and γ-d-glutamyl-meso-diaminopimelic acid (iE-DAP; NOD 1) were encapsulated. The immunomodulatory potential of these ligand-loaded MP was evaluated with a human acute monocytic leukemia cell line (THP-1X Blue-CD14 cells). Results The MP prepared had a phagocytosable size (<10 μm) with a unimodal size distribution and low endotoxin content (<0.5 EU/mL). A dose-dependent cell activation could be established for MDP-loaded microparticles. Conclusions MP with suitable characteristics for phagocytosis can be prepared and loaded with NOD agonists. The capability of these ligand-loaded microparticles to activate monocytes suggests their broader exploration as vaccine carriers.
Collapse
|
36
|
Wischke C, Mathew S, Roch T, Frentsch M, Lendlein A. Potential of NOD receptor ligands as immunomodulators in particulate vaccine carriers. J Control Release 2012; 164:299-306. [DOI: 10.1016/j.jconrel.2012.06.034] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Revised: 06/13/2012] [Accepted: 06/27/2012] [Indexed: 10/28/2022]
|
37
|
Mao S, Guo C, Shi Y, Li LC. Recent advances in polymeric microspheres for parenteral drug delivery--part 2. Expert Opin Drug Deliv 2012; 9:1209-23. [PMID: 22924745 DOI: 10.1517/17425247.2012.717926] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Currently marketed microsphere products are manufactured with the use of organic solvents which have a negative impact on the environment and stability of biological molecules. With recent advances in fabrication technologies, solvent free methods have demonstrated potential for the preparation of microspheres. AREAS COVERED New technical advances recently achieved in solvent based microsphere manufacturing processes have allowed for major improvement in product quality and properties. Novel solvent free fabrication methods combined with newly functionalized biodegradable polymers have been explored for their application in the preparation of microspheres containing biological molecules. EXPERT OPINION Novel fabrication methods for microspheres have been recently reported but technical challenges and development risks remain high for scale up from bench to industrial commercialization. While the applications of microspheres for delivery of proteins, genes and vaccines have shown promise for clinical use, the approval of newly functionalized polymers as carriers may still face scrutiny on safety and biocompatibility, which can be key factors in securing the regulatory approval of the product.
Collapse
Affiliation(s)
- Shirui Mao
- Shenyang Pharmaceutical University, School of Pharmacy, China
| | | | | | | |
Collapse
|
38
|
Cruz LJ, Tacken PJ, Rueda F, Domingo JC, Albericio F, Figdor CG. Targeting nanoparticles to dendritic cells for immunotherapy. Methods Enzymol 2012; 509:143-63. [PMID: 22568905 DOI: 10.1016/b978-0-12-391858-1.00008-3] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Dendritic cells (DCs) are key players in the initiation of adaptive immune responses and are currently exploited in immunotherapy for treatment of cancer and infectious diseases. Development of targeted nanodelivery systems carrying vaccine components, including antigens and adjuvants, to DCs in vivo represents a promising strategy to enhance immune responses. Delivering particulate vaccines specifically to DCs and preventing nonspecific uptake by other endocytotic cells are challenging. Size represents a critical parameter determining whether particulate vaccines can penetrate lymph nodes and reach resident DCs. Specific delivery is further enhanced by actively targeting DC-specific receptors. This chapter discusses the rationale for the use of particle-based vaccines and provides an overview of antigen-delivery vehicles currently under investigation. In addition, we discuss how vaccine delivery systems may be developed, focusing on liposomes, PLGA polymers, and gold nanoparticles, to obtain safe and efficacious vaccines.
Collapse
Affiliation(s)
- Luis J Cruz
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | | | | | | | | | | |
Collapse
|
39
|
Bioley G, Bussat P, Lassus A, Schneider M, Terrettaz J, Corthésy B. The phagocytosis of gas-filled microbubbles by human and murine antigen-presenting cells. Biomaterials 2011; 33:333-42. [PMID: 21983137 DOI: 10.1016/j.biomaterials.2011.09.045] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Accepted: 09/21/2011] [Indexed: 01/12/2023]
Abstract
This study was designed to evaluate the potential of gas-filled microbubbles (MB) to be internalized by antigen-presenting cells (APC). Fluorescently labeled MB were prepared, thus permitting to track binding to, and internalization in, APC. Both human and mouse cells, including monocytes and dendritic cells (DC), prove capable to phagocyte MB in vitro. Observation by confocal laser scanning microscopy showed that interaction between MB and target cells resulted in a rapid internalization in cellular compartments and to a lesser extent in the cytoplasm. Capture of MB by APC resulted in phagolysosomal targeting as verified by double staining with anti-lysosome-associated membrane protein-1 monoclonal antibody and decrease of internalization by phagocytosis inhibitors. Fluorescent MB injected subcutaneously (s.c.) in mice were found to be associated with CD11c(+)DC in lymph nodes draining the injection sites 24 h after administration. Altogether, our study demonstrates that MB can successfully target APC both in vitro and in vivo, and thus may serve as a potent Ag delivery system without requirement for ultrasound-based sonoporation. This adds to the potential of applications of MB already extensively used for diagnostic imaging in humans.
Collapse
Affiliation(s)
- Gilles Bioley
- R&D Laboratory, Immunology and Allergy, University State Hospital, Rue du Bugnon 46, 1011 Lausanne, Switzerland
| | | | | | | | | | | |
Collapse
|
40
|
Sionkowska A. Current research on the blends of natural and synthetic polymers as new biomaterials: Review. Prog Polym Sci 2011. [DOI: 10.1016/j.progpolymsci.2011.05.003] [Citation(s) in RCA: 532] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
41
|
Palumbo RN, Nagarajan L, Wang C. Recombinant monomeric CD40 ligand for delivering polymer particles to dendritic cells. Biotechnol Prog 2011; 27:830-7. [PMID: 21538973 DOI: 10.1002/btpr.595] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2010] [Revised: 11/24/2010] [Indexed: 01/23/2023]
Abstract
Dendritic cells (DCs) are considered the most efficient antigen-presenting cells and are therefore ideal targets for in vivo delivery of antigen for vaccines. We are investigating the strategy of using CD40 ligand (CD40L) as a targeting moiety because this protein has the potential to not only target DCs, but also stimulate cell maturation, leading to more potent immune responses. We have shown that a recombinant, monomeric CD40 ligand fusion protein conjugated to polystyrene micro- and nanoparticles led to significantly enhanced uptake by DCs in vitro. This enhancement was observed for particles of both sizes and in both a murine DC cell line and primary DCs. The uptake appeared to be specifically mediated by CD40L binding to CD40 expressed on DCs. Enhanced uptake of nanoparticles in draining lymph nodes of mice was not observed, however, 48 hours after subcutaneous injection. These findings suggest that CD40 ligand may be a potentially useful targeting moiety for delivery of particulate vaccines to DCs, and that further optimization of both CD40L and the polymer carriers is necessary to achieve efficacy in vivo.
Collapse
Affiliation(s)
- R Noelle Palumbo
- Dept. of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | |
Collapse
|
42
|
Krajišnik D, Daković A, Milojević M, Malenović A, Kragović M, Bogdanović DB, Dondur V, Milić J. Properties of diclofenac sodium sorption onto natural zeolite modified with cetylpyridinium chloride. Colloids Surf B Biointerfaces 2011; 83:165-72. [DOI: 10.1016/j.colsurfb.2010.11.024] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2010] [Revised: 11/01/2010] [Accepted: 11/10/2010] [Indexed: 10/18/2022]
|
43
|
Arias MA, Loxley A, Eatmon C, Van Roey G, Fairhurst D, Mitchnick M, Dash P, Cole T, Wegmann F, Sattentau Q, Shattock R. Carnauba wax nanoparticles enhance strong systemic and mucosal cellular and humoral immune responses to HIV-gp140 antigen. Vaccine 2011; 29:1258-69. [PMID: 21145913 PMCID: PMC3098439 DOI: 10.1016/j.vaccine.2010.11.084] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2010] [Revised: 10/21/2010] [Accepted: 11/24/2010] [Indexed: 01/28/2023]
Abstract
Induction of humoral responses to HIV at mucosal compartments without inflammation is important for vaccine design. We developed charged wax nanoparticles that efficiently adsorb protein antigens and are internalized by DC in the absence of inflammation. HIV-gp140-adsorbed nanoparticles induced stronger in vitro T-cell proliferation responses than antigen alone. Such responses were greatly enhanced when antigen was co-adsorbed with TLR ligands. Immunogenicity studies in mice showed that intradermal vaccination with HIV-gp140 antigen-adsorbed nanoparticles induced high levels of specific IgG. Importantly, intranasal immunization with HIV-gp140-adsorbed nanoparticles greatly enhanced serum and vaginal IgG and IgA responses. Our results show that HIV-gp140-carrying wax nanoparticles can induce strong cellular/humoral immune responses without inflammation and may be of potential use as effective mucosal adjuvants for HIV vaccine candidates.
Collapse
Affiliation(s)
- Mauricio A. Arias
- Division of Clinical Sciences, St. George's University of London, London SW17 0RE, UK
| | - Andrew Loxley
- Particle Sciences, Inc., 3894 Courtney Street, Suite 180, Bethlehem, Pennsylvania 18017, USA
| | - Christy Eatmon
- Particle Sciences, Inc., 3894 Courtney Street, Suite 180, Bethlehem, Pennsylvania 18017, USA
| | - Griet Van Roey
- Division of Clinical Sciences, St. George's University of London, London SW17 0RE, UK
| | - David Fairhurst
- Particle Sciences, Inc., 3894 Courtney Street, Suite 180, Bethlehem, Pennsylvania 18017, USA
| | - Mark Mitchnick
- Particle Sciences, Inc., 3894 Courtney Street, Suite 180, Bethlehem, Pennsylvania 18017, USA
| | - Philip Dash
- Division of Clinical Sciences, St. George's University of London, London SW17 0RE, UK
| | - Tom Cole
- Division of Clinical Sciences, St. George's University of London, London SW17 0RE, UK
| | - Frank Wegmann
- The Sir William Dunn School of Pathology, University of Oxford, South Park Road, Oxford OX1 3RE, UK
| | - Quentin Sattentau
- The Sir William Dunn School of Pathology, University of Oxford, South Park Road, Oxford OX1 3RE, UK
| | - Robin Shattock
- Division of Clinical Sciences, St. George's University of London, London SW17 0RE, UK
| |
Collapse
|
44
|
Weber C, Drogoz A, David L, Domard A, Charles MH, Verrier B, Delair T. Polysaccharide-based vaccine delivery systems: Macromolecular assembly, interactions with antigen presenting cells, and in vivo immunomonitoring. J Biomed Mater Res A 2010; 93:1322-34. [PMID: 19859973 DOI: 10.1002/jbm.a.32605] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Using a strategy of macromolecular assembly, a colloidal vaccine delivery system was obtained from chitosan and dextran sulfate and loaded with an antigenic protein (p24, the capsid protein of HIV-1). The colloidal polyelectrolyte complexes (PECs) were obtained by charge neutralization of the polyanion and polycation at a charge ratio (n(+)/n(-)) of 2 (CHDS). The conditions of assembly were tuned to maintain the colloidal properties of the carrier in high salt environment. The relative molar masses of the two polyions and the degree of acetylation (DA) of chitosan were essential parameters to achieve this goal, and this could be related to the nanometric scale organization of the colloids observed by Small Angle X-rays Scattering experiments. The binding of p24 to the colloidal carrier was achieved and the release of the antigen was investigated. Antigen presenting cells [dendritic cells (DCs)], obtained from monocytes, could internalize the colloids. Immature DCs (iDCs) were not matured by the colloidal PECs either loaded or not loaded with p24, as proved by Fluorescent Activated Cell Sorting (FACS) analysis. Despite this lack of in vitro interaction, a specific immune response was observed in mice with a high production of antibodies, after subcutaneous injection. The analysis of the interleukin production shows that both the cellular and the humoral responses were stimulated. This work brings a physico-chemical insight on polysaccharide-based antigen delivery systems and opens up new perspectives for their use as vaccine carriers.
Collapse
Affiliation(s)
- Caroline Weber
- Institut de Biologie et Chimie des Protéines, 69367 Lyon Cedex 07, France
| | | | | | | | | | | | | |
Collapse
|
45
|
Han R, Zhu J, Yang X, Xu H. Surface modification of poly(D,L-lactic-co-glycolic acid) nanoparticles with protamine enhanced cross-presentation of encapsulated ovalbumin by bone marrow-derived dendritic cells. J Biomed Mater Res A 2010; 96:142-9. [PMID: 21105162 DOI: 10.1002/jbm.a.32860] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2010] [Accepted: 04/26/2010] [Indexed: 11/09/2022]
Abstract
Cross-presentation is the key process in stimulation of cytotoxic T lymphocyte (CTL) immune response in eliminating many infectious diseases and tumors. Previous studies have shown that surface modification of poly(D,L-lactic-co-glycolic acid) (PLGA) particles with polycations enhanced their adjuvant ability resulting in a strong antibody response to the encapsulated antigen. However, the in vitro cross-presentation by protamine-coated PLGA nanoparticles (NPs) has not been addressed yet. In this study, a model antigen ovalbumin (OVA) was encapsulated into PLGA nanoparticles, with (OVA-NPs/protamine) or without protamine coating (OVA-NPs). These nanoparticles were then used to stimulate murine bone marrow-derived dendritic cells (BMDCs). Flow cytometry analysis revealed an increase in endocytosis of protamine-coated PLGA nanoparticles by BMDCs at 37°C. Compared with OVA-NPs-treated BMDCs, stimulation with OVA-NPs/protamine led to significantly upregulation of CD80, CD86, and CD83, increased secretion of IL-12p70, and decreased production of IL-4 by BMDCs. Furthermore, OVA-NPs/protamine-treated BMDCs also showed an enhanced cross-presentation to B3Z T cell hybridoma in vitro. Transmission electron microscopy (TEM) study showed that protamine-coated PLGA nanoparticles escaped from lysosomes through the interaction with lysosomal membrane. These results demonstrated that protamine-coated PLGA nanoparticles could enhance the cross-presentation of encapsulated exogenous antigen by facilitating antigen uptake and lysosomal escape, suggesting the feasibility to be a potent adjuvant for cellular vaccines.
Collapse
Affiliation(s)
- Ruiling Han
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | | | | | | |
Collapse
|
46
|
Chen H, Li P, Yin Y, Cai X, Huang Z, Chen J, Dong L, Zhang J. The promotion of type 1 T helper cell responses to cationic polymers in vivo via toll-like receptor-4 mediated IL-12 secretion. Biomaterials 2010; 31:8172-80. [DOI: 10.1016/j.biomaterials.2010.07.056] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2010] [Accepted: 07/13/2010] [Indexed: 10/19/2022]
|
47
|
Fay F, Quinn DJ, Gilmore BF, McCarron PA, Scott CJ. Gene delivery using dimethyldidodecylammonium bromide-coated PLGA nanoparticles. Biomaterials 2010; 31:4214-22. [DOI: 10.1016/j.biomaterials.2010.01.143] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2009] [Accepted: 01/29/2010] [Indexed: 02/07/2023]
|
48
|
Kinetics of styrene miniemulsion polymerization using poly[(stearyl methacrylate-co-(N,N-dimethylamino)ethyl methacrylate] as surfactant. Colloids Surf A Physicochem Eng Asp 2010. [DOI: 10.1016/j.colsurfa.2010.02.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
49
|
Gill RF, Pirockinaite G, O'Sullivan NL, Montgomery PC. Nasal-associated lymphoid tissue is not an absolute requirement for the induction of rat tear IgA antibody responses. Curr Eye Res 2010; 35:1-8. [PMID: 20021248 DOI: 10.3109/02713680903395281] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
UNLABELLED PURPOSE/AIM OF STUDY: The purpose of this work was to determine whether rat nasal-associated lymphoid tissue is required for the induction of tear IgA responses. MATERIALS AND METHODS Particulate antigen in the form of DNP-BSA encapsulated in cationic microparticles was applied topically to the eyes (ocular topically) of rats that had the nasolacrimal ducts temporarily plugged with chromic gut suture material. Eye washes and serum were monitored for development of antigen specific IgA and IgG, respectively. To track the particulate uptake, fluorescent latex beads were applied topically to the eyes of plugged and unplugged animals. The nasal-associated lymphoid tissue and the draining lymph nodes were then examined for the presence of the fluorescent beads. RESULTS It was found that the chromic gut suture was effective in blocking the passage of antigen into the nasopharyngeal cavity for at least 24 hr. Tear antigen-specific IgA levels found in the eyes of plugged animals were not significantly lower from those of unplugged animals. Serum IgG antibody levels were also similar between the two groups. In animals with plugged nasolacrimal ducts, fluorescent beads were found predominately in the superficial cervical lymph nodes, which have been shown to drain the surface of the eye. CONCLUSIONS These results indicate that particulate antigen can be taken up by the conjunctiva and transported to the draining lymph nodes, showing that antigen does not need to access nasal-associated lymphoid tissue to induce tear IgA antibody responses.
Collapse
Affiliation(s)
- Randall F Gill
- Department of Immunology and Microbiology, Wayne State University School of Medicine, Detroit, Michigan 48201, USA.
| | | | | | | |
Collapse
|
50
|
Bal SM, Slütter B, van Riet E, Kruithof AC, Ding Z, Kersten GF, Jiskoot W, Bouwstra JA. Efficient induction of immune responses through intradermal vaccination with N-trimethyl chitosan containing antigen formulations. J Control Release 2010; 142:374-83. [DOI: 10.1016/j.jconrel.2009.11.018] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2009] [Revised: 11/11/2009] [Accepted: 11/15/2009] [Indexed: 10/20/2022]
|