1
|
Tunbridge MJ, Luo X, Thomson AW. Negative Vaccination Strategies for Promotion of Transplant Tolerance. Transplantation 2024; 108:1715-1729. [PMID: 38361234 PMCID: PMC11265982 DOI: 10.1097/tp.0000000000004911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Organ transplantation requires the use of immunosuppressive medications that lack antigen specificity, have many adverse side effects, and fail to induce immunological tolerance to the graft. The safe induction of tolerance to allogeneic tissue without compromising host responses to infection or enhancing the risk of malignant disease is a major goal in transplantation. One promising approach to achieve this goal is based on the concept of "negative vaccination." Vaccination (or actively acquired immunity) involves the presentation of both a foreign antigen and immunostimulatory adjuvant to the immune system to induce antigen-specific immunity. By contrast, negative vaccination, in the context of transplantation, involves the delivery of donor antigen before or after transplantation, together with a "negative adjuvant" to selectively inhibit the alloimmune response. This review will explore established and emerging negative vaccination strategies for promotion of organ or pancreatic islet transplant tolerance. These include donor regulatory myeloid cell infusion, which has progressed to early-phase clinical trials, apoptotic donor cell infusion that has advanced to nonhuman primate models, and novel nanoparticle antigen-delivery systems.
Collapse
Affiliation(s)
- Matthew J. Tunbridge
- Adelaide Medical School, University of Adelaide, Adelaide, Australia
- Department of Medicine (Nephrology), Duke University Medical Center, Durham, North Carolina, USA
| | - Xunrong Luo
- Department of Medicine (Nephrology), Duke University Medical Center, Durham, North Carolina, USA
| | - Angus W. Thomson
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
2
|
Pham JPA, Coronel MM. Unlocking Transplant Tolerance with Biomaterials. Adv Healthc Mater 2024:e2400965. [PMID: 38843866 DOI: 10.1002/adhm.202400965] [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: 03/14/2024] [Revised: 05/31/2024] [Indexed: 07/04/2024]
Abstract
For patients suffering from organ failure due to injury or autoimmune disease, allogeneic organ transplantation with chronic immunosuppression is considered the god standard in terms of clinical treatment. However, the true "holy grail" of transplant immunology is operational tolerance, in which the recipient exhibits a sustained lack of alloreactivity toward unencountered antigen presented by the donor graft. This outcome is resultant from critical changes to the phenotype and genotype of the immune repertoire predicated by the activation of specific signaling pathways responsive to soluble and mechanosensitive cues. Biomaterials have emerged as a medium for interfacing with and reprogramming these endogenous pathways toward tolerance in precise, minimally invasive, and spatiotemporally defined manners. By viewing seminal and contemporary breakthroughs in transplant tolerance induction through the lens of biomaterials-mediated immunomodulation strategies-which include intrinsic material immunogenicity, the depot effect, graft coatings, induction and delivery of tolerogenic immune cells, biomimicry of tolerogenic immune cells, and in situ reprogramming-this review emphasizes the stunning diversity of approaches in the field and spotlights exciting future directions for research to come.
Collapse
Affiliation(s)
- John-Paul A Pham
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- Elizabeth Caswell Diabetes Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - María M Coronel
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- Elizabeth Caswell Diabetes Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
3
|
Abousalman-Rezvani Z, Refaat A, Dehghankelishadi P, Roghani-Mamaqani H, Esser L, Voelcker NH. Insights into Targeted and Stimulus-Responsive Nanocarriers for Brain Cancer Treatment. Adv Healthc Mater 2024; 13:e2302902. [PMID: 38199238 DOI: 10.1002/adhm.202302902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 12/10/2023] [Indexed: 01/12/2024]
Abstract
Brain cancers, especially glioblastoma multiforme, are associated with poor prognosis due to the limited efficacy of current therapies. Nanomedicine has emerged as a versatile technology to treat various diseases, including cancers, and has played an indispensable role in combatting the COVID-19 pandemic as evidenced by the role that lipid nanocarrier-based vaccines have played. The tunability of nanocarrier physicochemical properties -including size, shape, surface chemistry, and drug release kinetics- has resulted in the development of a wide range of nanocarriers for brain cancer treatment. These nanocarriers can improve the pharmacokinetics of drugs, increase blood-brain barrier transfer efficiency, and specifically target brain cancer cells. These unique features would potentially allow for more efficient treatment of brain cancer with fewer side effects and better therapeutic outcomes. This review provides an overview of brain cancers, current therapeutic options, and challenges to efficient brain cancer treatment. The latest advances in nanomedicine strategies are investigated with an emphasis on targeted and stimulus-responsive nanocarriers and their potential for clinical translation.
Collapse
Affiliation(s)
- Zahra Abousalman-Rezvani
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Melbourne, VIC 3052, Australia
- Biomedical Manufacturing, Commonwealth Scientific and Industrial Research Organization, Research Way, Melbourne, VIC 3168, Australia
| | - Ahmed Refaat
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Melbourne, VIC 3052, Australia
- Pharmaceutics Department, Faculty of Pharmacy - Alexandria University, 1 El-Khartoum Square, Alexandria, 21021, Egypt
| | - Pouya Dehghankelishadi
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Melbourne, VIC 3052, Australia
| | - Hossein Roghani-Mamaqani
- Faculty of Polymer Engineering, Sahand University of Technology, Tabriz, P.O. Box: 51335/1996, Iran
| | - Lars Esser
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Melbourne, VIC 3052, Australia
- Biomedical Manufacturing, Commonwealth Scientific and Industrial Research Organization, Research Way, Melbourne, VIC 3168, Australia
| | - Nicolas H Voelcker
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Melbourne, VIC 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Rd, Melbourne, VIC 3168, Australia
- Department of Materials Science & Engineering, Faculty of Engineering, Monash University, 14 Alliance Ln, Melbourne, VIC 3168, Australia
| |
Collapse
|
4
|
Liu Q, Chen G, Liu X, Tao L, Fan Y, Xia T. Tolerogenic Nano-/Microparticle Vaccines for Immunotherapy. ACS NANO 2024. [PMID: 38323542 DOI: 10.1021/acsnano.3c11647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
Autoimmune diseases, allergies, transplant rejections, generation of antidrug antibodies, and chronic inflammatory diseases have impacted a large group of people across the globe. Conventional treatments and therapies often use systemic or broad immunosuppression with serious efficacy and safety issues. Tolerogenic vaccines represent a concept that has been extended from their traditional immune-modulating function to induction of antigen-specific tolerance through the generation of regulatory T cells. Without impairing immune homeostasis, tolerogenic vaccines dampen inflammation and induce tolerogenic regulation. However, achieving the desired potency of tolerogenic vaccines as preventive and therapeutic modalities calls for precise manipulation of the immune microenvironment and control over the tolerogenic responses against the autoantigens, allergens, and/or alloantigens. Engineered nano-/microparticles possess desirable design features that can bolster targeted immune regulation and enhance the induction of antigen-specific tolerance. Thus, particle-based tolerogenic vaccines hold great promise in clinical translation for future treatment of aforementioned immune disorders. In this review, we highlight the main strategies to employ particles as exciting tolerogenic vaccines, with a focus on the particles' role in facilitating the induction of antigen-specific tolerance. We describe the particle design features that facilitate their usage and discuss the challenges and opportunities for designing next-generation particle-based tolerogenic vaccines with robust efficacy to promote antigen-specific tolerance for immunotherapy.
Collapse
Affiliation(s)
- Qi Liu
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Guoqiang Chen
- State Key Laboratory of Biochemical Engineering, Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100190, China
| | - Xingchi Liu
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Lu Tao
- State Key Laboratory of Biochemical Engineering, Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100190, China
| | - Yubo Fan
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Tian Xia
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
- Division of NanoMedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
| |
Collapse
|
5
|
Mehryab F, Rabbani S, Shekari F, Nazari A, Goshtasbi N, Haeri A. Sirolimus-loaded exosomes as a promising vascular delivery system for the prevention of post-angioplasty restenosis. Drug Deliv Transl Res 2024; 14:158-176. [PMID: 37518365 DOI: 10.1007/s13346-023-01390-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2023] [Indexed: 08/01/2023]
Abstract
Restenosis remains the main reason for treatment failure of arterial disease. Sirolimus (SIR) as a potent anti-proliferative agent is believed to prevent the phenomenon. The application of exosomes provides an extended-release delivery platform for SIR intramural administration. Herein, SIR was loaded into fibroblast-derived exosomes isolated by ultracentrifugation. Different parameters affecting drug loading were optimized, and exosome samples were characterized regarding physicochemical, pharmaceutical, and biological properties. Cytotoxicity, scratch wound assays, and quantitative real-time PCR for inflammation- and migration-associated genes were performed. Restenosis was induced by carotid injury in a rat carotid model and then exosomes were locally administered. After 14 days, animals were investigated by computed tomography (CT) angiography, morphometric, and immunohistochemical analyses. Western blotting confirmed the presence of specific protein markers in exosomes. Characterization of empty and SIR-loaded exosomes verified round and nanoscale structure of vesicles. Among prepared formulations, desired entrapment efficiency (EE) of 76% was achieved by protein:drug proportion of 2:1 and simple incubation for 30 min at 37 °C. Also, the optimal formulation released about 30% of the drug content during the first 24 h, followed by a prolonged release for several days. In vitro studies revealed the uptake and functional efficacy of the optimized formulation. In vivo studies revealed that %restenosis was in the following order: saline > empty exosomes > SIR-loaded exosomes. Furthermore, Ki67, alpha smooth muscle actin (α-SMA), and matrix metalloproteinase (MMP) markers were less expressed in the SIR-exosomes-treated arteries. These findings confirmed that exosomal SIR could be a hopeful strategy for the prevention of restenosis.
Collapse
Affiliation(s)
- Fatemeh Mehryab
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, PO Box: 14155-6153, Tehran, Iran
| | - Shahram Rabbani
- Research Center for Advanced Technologies in Cardiovascular Medicine, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Abdoreza Nazari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Nazanin Goshtasbi
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, PO Box: 14155-6153, Tehran, Iran
| | - Azadeh Haeri
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, PO Box: 14155-6153, Tehran, Iran.
- Protein Technology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Scotland BL, Shaw JR, Dharmaraj S, Caprio N, Cottingham AL, Joy Martín Lasola J, Sung JJ, Pearson RM. Cell and biomaterial delivery strategies to induce immune tolerance. Adv Drug Deliv Rev 2023; 203:115141. [PMID: 37980950 PMCID: PMC10842132 DOI: 10.1016/j.addr.2023.115141] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/09/2023] [Accepted: 11/10/2023] [Indexed: 11/21/2023]
Abstract
The prevalence of immune-mediated disorders, including autoimmune conditions and allergies, is steadily increasing. However, current therapeutic approaches are often non-specific and do not address the underlying pathogenic condition, often resulting in impaired immunity and a state of generalized immunosuppression. The emergence of technologies capable of selectively inhibiting aberrant immune activation in a targeted, antigen (Ag)-specific manner by exploiting the body's intrinsic tolerance pathways, all without inducing adverse side effects, holds significant promise to enhance patient outcomes. In this review, we will describe the body's natural mechanisms of central and peripheral tolerance as well as innovative delivery strategies using cells and biomaterials targeting innate and adaptive immune cells to promote Ag-specific immune tolerance. Additionally, we will discuss the challenges and future opportunities that warrant consideration as we navigate the path toward clinical implementation of tolerogenic strategies to treat immune-mediated diseases.
Collapse
Affiliation(s)
- Brianna L Scotland
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD 21201, United States
| | - Jacob R Shaw
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore Street, Baltimore, MD 21201, United States
| | - Shruti Dharmaraj
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD 21201, United States
| | - Nicholas Caprio
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD 21201, United States
| | - Andrea L Cottingham
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD 21201, United States
| | - Jackline Joy Martín Lasola
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore Street, Baltimore, MD 21201, United States
| | - Junsik J Sung
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD 21201, United States
| | - Ryan M Pearson
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD 21201, United States; Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore Street, Baltimore, MD 21201, United States; Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S. Greene Street, Baltimore, MD 21201, United States.
| |
Collapse
|
7
|
Lin G, Wang J, Yang YG, Zhang Y, Sun T. Advances in dendritic cell targeting nano-delivery systems for induction of immune tolerance. Front Bioeng Biotechnol 2023; 11:1242126. [PMID: 37877041 PMCID: PMC10593475 DOI: 10.3389/fbioe.2023.1242126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 09/25/2023] [Indexed: 10/26/2023] Open
Abstract
Dendritic cells (DCs) are the major specialized antigen-presenting cells (APCs), play a key role in initiating the body's immune response, maintain the balance of immunity. DCs can also induce immune tolerance by rendering effector T cells absent and anergy, and promoting the expansion of regulatory T cells. Induction of tolerogenic DCs has been proved to be a promising strategy for the treatment of autoimmune diseases, organ transplantation, and allergic diseases by various laboratory researches and clinical trials. The development of nano-delivery systems has led to advances in situ modulation of the tolerance phenotype of DCs. By changing the material composition, particle size, zeta-potential, and surface modification of nanoparticles, nanoparticles can be used for the therapeutic payloads targeted delivery to DCs, endowing them with great potential in the induction of immune tolerance. This paper reviews how nano-delivery systems can be modulated for targeted delivery to DCs and induce immune tolerance and reviews their potential in the treatment of autoimmune diseases, organ transplantation, and allergic diseases.
Collapse
Affiliation(s)
- Guojiao Lin
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| | - Jialiang Wang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
- International Center of Future Science, Jilin University, Changchun, China
| | - Yuning Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
- International Center of Future Science, Jilin University, Changchun, China
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, China
| |
Collapse
|
8
|
Xia Y, Fu S, Ma Q, Liu Y, Zhang N. Application of Nano-Delivery Systems in Lymph Nodes for Tumor Immunotherapy. NANO-MICRO LETTERS 2023; 15:145. [PMID: 37269391 PMCID: PMC10239433 DOI: 10.1007/s40820-023-01125-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 05/07/2023] [Indexed: 06/05/2023]
Abstract
Immunotherapy has become a promising research "hotspot" in cancer treatment. "Soldier" immune cells are not uniform throughout the body; they accumulate mostly in the immune organs such as the spleen and lymph nodes (LNs), etc. The unique structure of LNs provides the microenvironment suitable for the survival, activation, and proliferation of multiple types of immune cells. LNs play an important role in both the initiation of adaptive immunity and the generation of durable anti-tumor responses. Antigens taken up by antigen-presenting cells in peripheral tissues need to migrate with lymphatic fluid to LNs to activate the lymphocytes therein. Meanwhile, the accumulation and retaining of many immune functional compounds in LNs enhance their efficacy significantly. Therefore, LNs have become a key target for tumor immunotherapy. Unfortunately, the nonspecific distribution of the immune drugs in vivo greatly limits the activation and proliferation of immune cells, which leads to unsatisfactory anti-tumor effects. The efficient nano-delivery system to LNs is an effective strategy to maximize the efficacy of immune drugs. Nano-delivery systems have shown beneficial in improving biodistribution and enhancing accumulation in lymphoid tissues, exhibiting powerful and promising prospects for achieving effective delivery to LNs. Herein, the physiological structure and the delivery barriers of LNs were summarized and the factors affecting LNs accumulation were discussed thoroughly. Moreover, developments in nano-delivery systems were reviewed and the transformation prospects of LNs targeting nanocarriers were summarized and discussed.
Collapse
Affiliation(s)
- Yiming Xia
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Shunli Fu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Qingping Ma
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Yongjun Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China.
| | - Na Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China.
| |
Collapse
|
9
|
Cheng R, Santos HA. Smart Nanoparticle-Based Platforms for Regulating Tumor Microenvironment and Cancer Immunotherapy. Adv Healthc Mater 2023; 12:e2202063. [PMID: 36479842 PMCID: PMC11468886 DOI: 10.1002/adhm.202202063] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/18/2022] [Indexed: 12/12/2022]
Abstract
Tumor development and metastasis are closely related to the tumor microenvironment (TME). Recently, several studies indicate that modulating TME can enhance cancer immunotherapy. Among various approaches to modulating TME, nanoparticles (NPs) with unique inherent advantages and smart modified characteristics are promising candidates in delivering drugs to cancer cells, amplifying the therapeutic effects, and leading to a cascade of immune responses. In this review, several smart NP-based platforms are briefly introduced, such as responsive NPs, targeting NPs, and the composition of TME, including dendritic cells, macrophages, fibroblasts, endothelial cells, myeloid-derived suppressor cells, and regulatory T cells. Moreover, the recent applications of smart NP-based platforms in regulating TME and cancer immunotherapy are briefly introduced. Last, the advantages and disadvantages of these smart NP-based platforms in potential clinical translation are discussed.
Collapse
Affiliation(s)
- Ruoyu Cheng
- Department of Biomedical EngineeringUniversity Medical Center GroningenUniversity of GroningenAnt. Deusinglaan 1Groningen9713 AVThe Netherlands
- W. J. Kolff Institute for Biomedical Engineering and Materials ScienceUniversity Medical Center GroningenUniversity of GroningenAnt. Deusinglaan 1Groningen9713 AVThe Netherlands
- Drug Research ProgramDivision of Pharmaceutical Chemistry and TechnologyFaculty of PharmacyUniversity of HelsinkiHelsinkiFI‐00014Finland
| | - Hélder A. Santos
- Department of Biomedical EngineeringUniversity Medical Center GroningenUniversity of GroningenAnt. Deusinglaan 1Groningen9713 AVThe Netherlands
- W. J. Kolff Institute for Biomedical Engineering and Materials ScienceUniversity Medical Center GroningenUniversity of GroningenAnt. Deusinglaan 1Groningen9713 AVThe Netherlands
- Drug Research ProgramDivision of Pharmaceutical Chemistry and TechnologyFaculty of PharmacyUniversity of HelsinkiHelsinkiFI‐00014Finland
| |
Collapse
|
10
|
Zhang DX, Tieu T, Esser L, Wojnilowicz M, Lee CH, Cifuentes-Rius A, Thissen H, Voelcker NH. Differential Surface Engineering Generates Core-Shell Porous Silicon Nanoparticles for Controlled and Targeted Delivery of an Anticancer Drug. ACS APPLIED MATERIALS & INTERFACES 2022; 14:54539-54549. [PMID: 36469497 DOI: 10.1021/acsami.2c16370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
An approach to differentially modify the internal surface of porous silicon nanoparticles (pSiNPs) with hydrophobic dodecene and the external surface with antifouling poly-N-(2-hydroxypropyl) acrylamide (polyHPAm) as well as a cell-targeting peptide was developed. Specifically, to generate these core-shell pSiNPs, the interior surface of a porous silicon (pSi) film was hydrosilylated with 1-dodecene, followed by ultrasonication to create pSiNPs. The new external surfaces were modified by silanization with a polymerization initiator, and surface-initiated atom transfer radical polymerization was performed to introduce polyHPAm brushes. Afterward, a fraction of the polymer side chain hydroxyl groups was activated to conjugate cRGDfK─a peptide with a high affinity and selectivity for the ανβ3 integrin receptor that is overexpressed in prostate and melanoma cancers. Finally, camptothecin, a hydrophobic anti-cancer drug, was successfully loaded into the pores. This drug delivery system showed excellent colloidal stability in a cell culture medium, and the in vitro drug release kinetics could be fine-tuned by the combination of internal and external surface modifications. In vitro studies by confocal microscopy and flow cytometry revealed improved cellular association attributed to cRGDfK. Furthermore, the cell viability results showed that the drug-loaded and peptide-functionalized nanoparticles had enhanced cytotoxicity toward a C4-2B prostate carcinoma cell line in both 2D cell culture and a 3D spheroid model.
Collapse
Affiliation(s)
- De-Xiang Zhang
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Manufacturing, Clayton, Victoria 3168, Australia
| | - Terence Tieu
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Manufacturing, Clayton, Victoria 3168, Australia
| | - Lars Esser
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Manufacturing, Clayton, Victoria 3168, Australia
| | - Marcin Wojnilowicz
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Manufacturing, Clayton, Victoria 3168, Australia
| | - Chieh-Hua Lee
- Department of Biological Science and Technology, China Medical University, Taichung 40402, Taiwan
| | - Anna Cifuentes-Rius
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Helmut Thissen
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Manufacturing, Clayton, Victoria 3168, Australia
| | - Nicolas H Voelcker
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Manufacturing, Clayton, Victoria 3168, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of Australian National Fabrication Facility, Clayton, Victoria 3168, Australia
| |
Collapse
|
11
|
Immunosuppressive cells in cancer: mechanisms and potential therapeutic targets. J Hematol Oncol 2022; 15:61. [PMID: 35585567 PMCID: PMC9118588 DOI: 10.1186/s13045-022-01282-8] [Citation(s) in RCA: 203] [Impact Index Per Article: 67.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/03/2022] [Indexed: 02/08/2023] Open
Abstract
Immunotherapies like the adoptive transfer of gene-engineered T cells and immune checkpoint inhibitors are novel therapeutic modalities for advanced cancers. However, some patients are refractory or resistant to these therapies, and the mechanisms underlying tumor immune resistance have not been fully elucidated. Immunosuppressive cells such as myeloid-derived suppressive cells, tumor-associated macrophages, tumor-associated neutrophils, regulatory T cells (Tregs), and tumor-associated dendritic cells are critical factors correlated with immune resistance. In addition, cytokines and factors secreted by tumor cells or these immunosuppressive cells also mediate the tumor progression and immune escape of cancers. Thus, targeting these immunosuppressive cells and the related signals is the promising therapy to improve the efficacy of immunotherapies and reverse the immune resistance. However, even with certain success in preclinical studies or in some specific types of cancer, large perspectives are unknown for these immunosuppressive cells, and the related therapies have undesirable outcomes for clinical patients. In this review, we comprehensively summarized the phenotype, function, and potential therapeutic targets of these immunosuppressive cells in the tumor microenvironment.
Collapse
|
12
|
Mitarotonda R, Giorgi E, Eufrasio-da-Silva T, Dolatshahi-Pirouz A, Mishra YK, Khademhosseini A, Desimone MF, De Marzi M, Orive G. Immunotherapeutic nanoparticles: From autoimmune disease control to the development of vaccines. BIOMATERIALS ADVANCES 2022; 135:212726. [PMID: 35475005 PMCID: PMC9023085 DOI: 10.1016/j.bioadv.2022.212726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 02/09/2022] [Accepted: 02/18/2022] [Indexed: 11/01/2022]
Abstract
The development of nanoparticles (NPs) with potential therapeutic uses represents an area of vast interest in the scientific community during the last years. Recently, the pandemic caused by COVID-19 motivated a race for vaccines creation to overcome the crisis generated. This is a good demonstration that nanotechnology will most likely be the basis of future immunotherapy. Moreover, the number of publications based on nanosystems has significantly increased in recent years and it is expected that most of these developments can go on to experimentation in clinical stages soon. The therapeutic use of NPs to combat different diseases such as cancer, allergies or autoimmune diseases will depend on their characteristics, their targets, and the transported molecules. This review presents an in-depth analysis of recent advances that have been developed in order to obtain novel nanoparticulate based tools for the treatment of allergies, autoimmune diseases and for their use in vaccines. Moreover, it is highlighted that by providing targeted delivery an increase in the potential of vaccines to induce an immune response is expected in the future. Definitively, the here gathered analysis is a good demonstration that nanotechnology will be the basis of future immunotherapy.
Collapse
Affiliation(s)
- Romina Mitarotonda
- Laboratorio de Inmunología, Instituto de Ecología y Desarrollo Sustentable (INEDES) CONICET-UNLu, Departamento de Ciencias Básicas, Universidad Nacional de Luján, Ruta 5 y Avenida Constitución (6700) Lujan, Buenos Aires, Argentina
| | - Exequiel Giorgi
- Laboratorio de Inmunología, Instituto de Ecología y Desarrollo Sustentable (INEDES) CONICET-UNLu, Departamento de Ciencias Básicas, Universidad Nacional de Luján, Ruta 5 y Avenida Constitución (6700) Lujan, Buenos Aires, Argentina
| | - Tatiane Eufrasio-da-Silva
- Department of Health Technology, Technical University of Denmark (DTU), 2800 Kgs. Lyngby, Denmark; Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Department of Dentistry - Regenerative Biomaterials, Philips van Leydenlaan 25, 6525EX Nijmegen, the Netherlands
| | | | - Yogendra Kumar Mishra
- Mads Clausen Institute, NanoSYD, University of Southern Denmark, 6400 Sønderborg, Denmark
| | - Ali Khademhosseini
- Department of Bioengineering, Center for Minimally Invasive Therapeutics (C-MIT), California NanoSystems Institute, University of California, Los Angeles, CA 90095, USA; Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, CA 90095, USA; Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA; Jonsson Comprehensive Cancer Center, Department of Radiology, University of California, Los Angeles, CA 90095, USA
| | - Martin F Desimone
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de la Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica, Buenos Aires, Argentina.
| | - Mauricio De Marzi
- Laboratorio de Inmunología, Instituto de Ecología y Desarrollo Sustentable (INEDES) CONICET-UNLu, Departamento de Ciencias Básicas, Universidad Nacional de Luján, Ruta 5 y Avenida Constitución (6700) Lujan, Buenos Aires, Argentina.
| | - Gorka Orive
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; University Institute for Regenerative Medicine and Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain; Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower, Singapore.
| |
Collapse
|
13
|
Cheng R, Wang S, Moslova K, Mäkilä E, Salonen J, Li J, Hirvonen J, Xia B, Santos HA. Quantitative Analysis of Porous Silicon Nanoparticles Functionalization by 1H NMR. ACS Biomater Sci Eng 2021; 8:4132-4139. [PMID: 34292713 PMCID: PMC9554871 DOI: 10.1021/acsbiomaterials.1c00440] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
![]()
Porous silicon (PSi)
nanoparticles have been applied in various
fields, such as catalysis, imaging, and biomedical applications, because
of their large specific surface area, easily modifiable surface chemistry,
biocompatibility, and biodegradability. For biomedical applications,
it is important to precisely control the surface modification of PSi-based
materials and quantify the functionalization density, which determines
the nanoparticle’s behavior in the biological system. Therefore,
we propose here an optimized solution to quantify the functionalization
groups on PSi, based on the nuclear magnetic resonance (NMR) method
by combining the hydrolysis with standard 1H NMR experiments.
We optimized the hydrolysis conditions to degrade the PSi, providing
mobility to the molecules for NMR detection. The NMR parameters were
also optimized by relaxation delay and the number of scans to provide
reliable NMR spectra. With an internal standard, we quantitatively
analyzed the surficial amine groups and their sequential modification
of polyethylene glycol. Our investigation provides a reliable, fast,
and straightforward method in quantitative analysis of the surficial
modification characterization of PSi requiring a small amount of sample.
Collapse
Affiliation(s)
- Ruoyu Cheng
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland
| | - Shiqi Wang
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland
| | - Karina Moslova
- Department of Chemistry, Faculty of Science, University of Helsinki, Helsinki FI-00014, Finland
| | - Ermei Mäkilä
- Laboratory of Industrial Physics, Department of Physics and Astronomy, University of Turku, Turku FI-20014, Finland
| | - Jarno Salonen
- Laboratory of Industrial Physics, Department of Physics and Astronomy, University of Turku, Turku FI-20014, Finland
| | - Jiachen Li
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland.,College of Science Key Laboratory of Forest Genetics & Biotechnology (Ministry of Education of China), Nanjing Forestry University, Nanjing 210037, P. R. China
| | - Jouni Hirvonen
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland
| | - Bing Xia
- College of Science Key Laboratory of Forest Genetics & Biotechnology (Ministry of Education of China), Nanjing Forestry University, Nanjing 210037, P. R. China
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland.,Helsinki Insititute of Life Science, HiLIFE, University of Helsinki, Helsinki FI-00014, Finland
| |
Collapse
|
14
|
Surface chemistry modification of silica nanoparticles alters the activation of monocytes. Ther Deliv 2021; 12:443-459. [PMID: 33902308 DOI: 10.4155/tde-2021-0006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Aim: Nanoparticles (NPs) interaction with immune system is a growing topic of study. Materials & methods: Bare and amine grafted silica NPs effects on monocytes/macrophages cells were analyzed by flow cytometry, MTT test and LIVE/DEAD® viability/cytotoxicity assay. Results: Bare silica NPs inhibited proliferation and induced monocyte/macrophages activation (increasing CD40/CD80 expression besides pro-inflammatory cytokines and nitrite secretion). Furthermore, silica NPs increased cell membrane damage and reduced the number of living cells. In contrast, amine grafted silica NPs did not alter these parameters. Conclusion: Cell activation properties of bare silica NPs could be hindered after grafting with amine moieties. This strategy is useful to tune the immune system stimulation by NPs or to design NPs suitable to transport therapeutic molecules.
Collapse
|
15
|
Cifuentes-Rius A, Desai A, Yuen D, Johnston APR, Voelcker NH. Inducing immune tolerance with dendritic cell-targeting nanomedicines. NATURE NANOTECHNOLOGY 2021; 16:37-46. [PMID: 33349685 DOI: 10.1038/s41565-020-00810-2] [Citation(s) in RCA: 126] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 10/29/2020] [Indexed: 04/14/2023]
Abstract
Induced tolerogenic dendritic cells are a powerful immunotherapy for autoimmune disease that have shown promise in laboratory models of disease and early clinical trials. In contrast to conventional immunosuppressive treatments, tolerogenic immunotherapy leverages the cells and function of the immune system to quell the autoreactive lymphocytes responsible for damage and disease. The principle techniques of isolating and reprogramming dendritic cells (DCs), central to this approach, are well characterized. However, the broader application of this technology is limited by its high cost and bespoke nature. Nanomedicine offers an alternative route by performing this reprogramming process in situ. Here, we review the challenges and opportunities in using nanoparticles as a delivery mechanism to target DCs and induce immunomodulation, emphasizing their versatility. We then highlight their potential to solve critical problems in organ transplantation and increasingly prevalent autoimmune disorders such as type 1 diabetes mellitus and multiple sclerosis, where new immunotherapy approaches have begun to show promise.
Collapse
Affiliation(s)
- Anna Cifuentes-Rius
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville Campus, Parkville, Victoria, Australia.
| | - Anal Desai
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville Campus, Parkville, Victoria, Australia
| | - Daniel Yuen
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville Campus, Parkville, Victoria, Australia
| | - Angus P R Johnston
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville Campus, Parkville, Victoria, Australia
| | - Nicolas H Voelcker
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville Campus, Parkville, Victoria, Australia.
- CSIRO Manufacturing, Bayview Avenue, Clayton, Victoria, Australia.
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, Victoria, Australia.
| |
Collapse
|
16
|
Iberg CA, Hawiger D. Natural and Induced Tolerogenic Dendritic Cells. THE JOURNAL OF IMMUNOLOGY 2020; 204:733-744. [PMID: 32015076 DOI: 10.4049/jimmunol.1901121] [Citation(s) in RCA: 159] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 10/04/2019] [Indexed: 11/19/2022]
Abstract
Dendritic cells (DCs) are highly susceptible to extrinsic signals that modify the functions of these crucial APCs. Maturation of DCs induced by diverse proinflammatory conditions promotes immune responses, but certain signals also induce tolerogenic functions in DCs. These "induced tolerogenic DCs" help to moderate immune responses such as those to commensals present at specific anatomical locations. However, also under steady-state conditions, some DCs are characterized by inherent tolerogenic properties. The immunomodulatory mechanisms constitutively present in such "natural tolerogenic DCs" help to promote tolerance to peripheral Ags. By extending tolerance initially established in the thymus, these functions of DCs help to regulate autoimmune and other immune responses. In this review we will discuss the mechanisms and functions of natural and induced tolerogenic DCs and offer further insight into how their possible manipulations may ultimately lead to more precise treatments for various immune-mediated conditions and diseases.
Collapse
Affiliation(s)
- Courtney A Iberg
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63104
| | - Daniel Hawiger
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63104
| |
Collapse
|
17
|
Zhang DX, Esser L, Vasani RB, Thissen H, Voelcker NH. Porous silicon nanomaterials: recent advances in surface engineering for controlled drug-delivery applications. Nanomedicine (Lond) 2020; 14:3213-3230. [PMID: 31855121 DOI: 10.2217/nnm-2019-0167] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Porous silicon (pSi) nanomaterials are increasingly attractive for biomedical applications due to their promising properties such as simple and feasible fabrication procedures, tunable morphology, versatile surface modification routes, biocompatibility and biodegradability. This review focuses on recent advances in surface modification of pSi for controlled drug delivery applications. A range of functionalization strategies and fabrication methods for pSi-polymer hybrids are summarized. Surface engineering solutions such as stimuli-responsive polymer grafting, stealth coatings and active targeting modifications are highlighted as examples to demonstrate what can be achieved. Finally, the current status of engineered pSi nanomaterials for in vivo applications is reviewed and future prospects and challenges in drug-delivery applications are discussed.
Collapse
Affiliation(s)
- De-Xiang Zhang
- Drug Delivery, Disposition & Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia.,Commonwealth Scientific & Industrial Research Organisation (CSIRO), Manufacturing, Clayton, Victoria, 3168, Australia
| | - Lars Esser
- Commonwealth Scientific & Industrial Research Organisation (CSIRO), Manufacturing, Clayton, Victoria, 3168, Australia
| | - Roshan B Vasani
- Drug Delivery, Disposition & Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
| | - Helmut Thissen
- Commonwealth Scientific & Industrial Research Organisation (CSIRO), Manufacturing, Clayton, Victoria, 3168, Australia
| | - Nicolas H Voelcker
- Drug Delivery, Disposition & Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia.,Commonwealth Scientific & Industrial Research Organisation (CSIRO), Manufacturing, Clayton, Victoria, 3168, Australia.,Melbourne Centre for Nanofabrication, Victorian Node of Australian National Fabrication Facility, Clayton, Victoria, 3168, Australia
| |
Collapse
|
18
|
Wei X, Fang Z, Sheng J, Wang Y, Lu P. Honokiol-mesoporous Silica Nanoparticles Inhibit Vascular Restenosis via the Suppression of TGF-β Signaling Pathway. Int J Nanomedicine 2020; 15:5239-5252. [PMID: 32801689 PMCID: PMC7399453 DOI: 10.2147/ijn.s250911] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 06/01/2020] [Indexed: 11/23/2022] Open
Abstract
Introduction The main pathological mechanism of restenosis after percutaneous coronary intervention (PCI) is intimal hyperplasia, which is mainly caused by proliferation and migration of vascular smooth muscle cells (VSMCs). Our previous study found that honokiol (HNK), a small-molecule polyphenol, can inhibit neointimal hyperplasia after balloon injury, but its specific mechanism is still unclear. Moreover, poor water solubility as well as low bioavailability of honokiol has limited its practical use. Methods We used mesoporous silica nanoparticles (MSNPs) as a standard substance to encapsulate HNK and then assemble into honokiol-mesoporous silica nanoparticles, and we investigated the effect of these nanoparticles on the process of restenosis after common carotid artery injury in rats. Results We report a promising delivery system that loads HNK into MSNPs and finally assembles it into a nanocomposite particle. These HNK-MSNPs not merely inhibited proliferation and migration of VSMCs by reducing phosphorylation of Smad3, but also showed a higher suppression of intimal thickening than the free-honokiol-treated group in a rat model of balloon injury. Conclusion To sum up, this drug delivery system supplies a potent nano-platform for improving the biological effects of HNK and provides a promising strategy for preventing vascular restenosis.
Collapse
Affiliation(s)
- Xiao Wei
- Department of Geriatrics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, People's Republic of China
| | - Zhiwei Fang
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
| | - Jing Sheng
- Department of Geriatrics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, People's Republic of China
| | - Yu Wang
- Department of Cardiology, Shidong Hospital of Yangpu District, Shanghai 200438, People's Republic of China
| | - Ping Lu
- Department of Geriatrics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, People's Republic of China
| |
Collapse
|
19
|
Effective Activation of Human Antigen-Presenting Cells and Cytotoxic CD8 + T Cells by a Calcium Phosphate-Based Nanoparticle Vaccine Delivery System. Vaccines (Basel) 2020; 8:vaccines8010110. [PMID: 32121590 PMCID: PMC7157756 DOI: 10.3390/vaccines8010110] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/12/2020] [Accepted: 02/20/2020] [Indexed: 12/20/2022] Open
Abstract
The ability of vaccines to induce T cell responses is crucial for preventing diseases caused by viruses. Nanoparticles (NPs) are considered to be efficient tools for the initiation of potent immune responses. Calcium phosphate (CaP) NPs are a class of biodegradable nanocarriers that are able to deliver immune activating molecules across physiological barriers. Therefore, the aim of this study was to assess whether Toll-like receptor (TLR) ligand and viral antigen functionalized CaP NPs are capable of inducing efficient maturation of human antigen presenting cells (APC). To achieve this, we generated primary human dendritic cells (DCs) and stimulated them with CpG or poly(I:C) functionalized CaP NPs. DCs were profoundly stronger when activated upon NP stimulation compared to treatment with soluble TLR ligands. This is indicated by increased levels of costimulatory molecules and the secretion of proinflammatory cytokines. Consequently, coculture of NP-stimulated APCs with CD8+ T cells resulted in a significant expansion of virus-specific T cells. In summary, our data suggest that functionalized CaP NPs are a suitable tool for activating human virus-specific CD8+ T cells and may represent an excellent vaccine delivery system.
Collapse
|
20
|
Cristescu R, Negut I, Visan AI, Nguyen AK, Sachan A, Goering PL, Chrisey DB, Narayan RJ. Matrix-Assisted Pulsed laser Evaporation-deposited Rapamycin Thin Films Maintain Antiproliferative Activity. Int J Bioprint 2020; 6:188. [PMID: 32782983 PMCID: PMC7415860 DOI: 10.18063/ijb.v6i1.188] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 05/16/2019] [Indexed: 11/23/2022] Open
Abstract
Matrix-assisted pulsed laser evaporation (MAPLE) has many benefits over conventional methods (e.g., dip-coating, spin coating, and Langmuir-Blodgett dip-coating) for manufacturing coatings containing pharmacologic agents on medical devices. In particular, the thickness of the coating that is applied to the surface of the medical device can be tightly controlled. In this study, MAPLE was used to deposit rapamycin-polyvinylpyrrolidone (rapamycin-PVP) thin films onto silicon and borosilicate optical glass substrates. Alamar Blue and PicoGreen studies were used to measure the metabolic health and DNA content of L929 mouse fibroblasts as measures of viability and proliferation, respectively. The cells on the MAPLE-deposited rapamycin-PVP surfaces exhibited 70.6% viability and 53.7% proliferation compared to a borosilicate glass control. These data indicate that the antiproliferative properties of rapamycin were maintained after MAPLE deposition.
Collapse
Affiliation(s)
- Rodica Cristescu
- Department of Lasers, National Institute for Lasers, Plasma and Radiation Physics, P.O. Box MG-36, Bucharest-Magurele, Romania
| | - Irina Negut
- Department of Lasers, National Institute for Lasers, Plasma and Radiation Physics, P.O. Box MG-36, Bucharest-Magurele, Romania
| | - Anita Ioana Visan
- Department of Lasers, National Institute for Lasers, Plasma and Radiation Physics, P.O. Box MG-36, Bucharest-Magurele, Romania
| | - Alexander K. Nguyen
- UNC/NCSU Joint Department of Biomedical Engineering, Raleigh, North Carolina, USA
- Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver Spring, Maryland, United States
| | - Andrew Sachan
- Wake Technical Community College, Raleigh, North Carolina, USA
| | - Peter L. Goering
- Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver Spring, Maryland, United States
| | - Douglas B. Chrisey
- Department of Physics and Engineering Physics, Tulane University, New Orleans, LA, USA
| | - Roger J. Narayan
- UNC/NCSU Joint Department of Biomedical Engineering, Raleigh, North Carolina, USA
| |
Collapse
|
21
|
Mitarotonda R, Giorgi E, Desimone MF, De Marzi MC. Nanoparticles and Immune Cells. Curr Pharm Des 2019; 25:3960-3982. [DOI: 10.2174/1381612825666190926161209] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 09/17/2019] [Indexed: 02/07/2023]
Abstract
Nanoparticles have gained ground in several fields. However, it is important to consider their potentially
hazardous effects on humans, flora, and fauna. Human exposure to nanomaterials can occur unintentionally
in daily life or in industrial settings, and the continuous exposure of the biological components (cells, receptors,
proteins, etc.) of the immune system to these particles can trigger an unwanted immune response (activation or
suppression). Here, we present different studies that have been carried out to evaluate the response of immune
cells in the presence of nanoparticles and their possible applications in the biomedical field.
Collapse
Affiliation(s)
- Romina Mitarotonda
- Laboratorio de Inmunologia, Instituto de Ecologia y Desarrollo Sustentable (INEDES) UNLu-CONICET, Buenos Aires, Argentina
| | - Exequiel Giorgi
- Laboratorio de Inmunologia, Instituto de Ecologia y Desarrollo Sustentable (INEDES) UNLu-CONICET, Buenos Aires, Argentina
| | - Martín F. Desimone
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Tecnicas (CONICET), Instituto de la Quimica y Metabolismo del Farmaco (IQUIMEFA), Facultad de Farmacia y Bioquimica, Buenos Aires, Argentina
| | - Mauricio C. De Marzi
- Laboratorio de Inmunologia, Instituto de Ecologia y Desarrollo Sustentable (INEDES) UNLu-CONICET, Buenos Aires, Argentina
| |
Collapse
|
22
|
Luo M, Lewik G, Ratcliffe JC, Choi CHJ, Mäkilä E, Tong WY, Voelcker NH. Systematic Evaluation of Transferrin-Modified Porous Silicon Nanoparticles for Targeted Delivery of Doxorubicin to Glioblastoma. ACS APPLIED MATERIALS & INTERFACES 2019; 11:33637-33649. [PMID: 31433156 DOI: 10.1021/acsami.9b10787] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
There is a dire need to develop more effective therapeutics to combat brain cancer such as glioblastoma multiforme (GBM). An ideal treatment is expected to target deliver chemotherapeutics to glioma cells across the blood-brain barrier (BBB). The overexpression of transferrin (Tf) receptor (TfR) on the BBB and the GBM cell surfaces but not on the surrounding cells renders TfR a promising target. While porous silicon nanoparticles (pSiNPs) have been intensely studied as a delivery vehicle due to their high biocompatibility, degradability, and drug-loading capacity, the potential to target deliver drugs with transferrin (Tf)-functionalized pSiNPs remains unaddressed. Here, we developed and systematically evaluated Tf-functionalized pSiNPs (Tf@pSiNPs) as a glioma-targeted drug delivery system. These nanoparticles showed excellent colloidal stability and had a low toxicity profile. As compared with nontargeted pSiNPs, Tf@pSiNPs were selective to BBB-forming cells and GBM cells and were efficiently internalized through clathrin receptor-mediated endocytosis. The anticancer drug doxorubicin (Dox) was effectively loaded (8.8 wt %) and released from Tf@pSiNPs in a pH-responsive manner over 24 h. Furthermore, the results demonstrate that Dox delivered by Tf@pSiNPs induced significantly enhanced cytotoxicity to GBM cells across an in vitro BBB monolayer compared with free Dox. Overall, Tf@pSiNPs offer a potential toolbox for enabling targeted therapy to treat GBM.
Collapse
Affiliation(s)
- Meihua Luo
- Monash Institute of Pharmaceutics Science, Monash University , Parkville Campus , 381 Royal Parade , Parkville , VIC 3052 , Australia
- Department of Biomedical Engineering , The Chinese University of Hong Kong , Shatin , New Territories , Hong Kong
| | - Guido Lewik
- Monash Institute of Pharmaceutics Science, Monash University , Parkville Campus , 381 Royal Parade , Parkville , VIC 3052 , Australia
- Faculty of Medicine , Ruhr-University Bochum , Bochum 44801 , Germany
| | - Julian Charles Ratcliffe
- Commonwealth Scientific and Industrial Research Organization (CSIRO) , Clayton , VIC 3168 , Australia
| | - Chung Hang Jonathan Choi
- Department of Biomedical Engineering , The Chinese University of Hong Kong , Shatin , New Territories , Hong Kong
| | - Ermei Mäkilä
- Industrial Physics Laboratory, Department of Physics and Astronomy , University of Turku , Turku 20014 , Finland
| | - Wing Yin Tong
- Monash Institute of Pharmaceutics Science, Monash University , Parkville Campus , 381 Royal Parade , Parkville , VIC 3052 , Australia
- Commonwealth Scientific and Industrial Research Organization (CSIRO) , Clayton , VIC 3168 , Australia
| | - Nicolas H Voelcker
- Monash Institute of Pharmaceutics Science, Monash University , Parkville Campus , 381 Royal Parade , Parkville , VIC 3052 , Australia
- Department of Biomedical Engineering , The Chinese University of Hong Kong , Shatin , New Territories , Hong Kong
- Commonwealth Scientific and Industrial Research Organization (CSIRO) , Clayton , VIC 3168 , Australia
- Melbourne Centre for Nanofabrication , Victorian Node of the Australian National Fabrication Facility , 151 Wellington Road , Clayton , VIC 3168 , Australia
- Materials Science and Engineering , Monash University , 14 Alliance Lane , Clayton , VIC 3800 , Australia
| |
Collapse
|
23
|
Yang G, Phua SZF, Bindra AK, Zhao Y. Degradability and Clearance of Inorganic Nanoparticles for Biomedical Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1805730. [PMID: 30614561 DOI: 10.1002/adma.201805730] [Citation(s) in RCA: 219] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 10/05/2018] [Indexed: 05/07/2023]
Abstract
Inorganic nanoparticles with tunable and diverse properties hold tremendous potential in the field of nanomedicine, while having non-negligible toxicity concerns in healthy tissues/organs that have resulted in their restricted clinical translation to date. In the past decade, the emergence of biodegradable or clearable inorganic nanoparticles has made it possible to completely solve this long-standing conundrum. A comprehensive understanding of the design of these inorganic nanoparticles with their metabolic performance in the body is of crucial importance to advance clinical trials and expand their biological applications in disease diagnosis. Here, a diverse variety of biodegradable or clearable inorganic nanoparticles regarding considerations of the size, morphology, surface chemistry, and doping strategy are highlighted. Their pharmacokinetics, pathways of metabolism in the body, and time required for excretion are discussed. Some inorganic materials intrinsically responsive to various conditions in the tumor microenvironment are also introduced. Finally, an overview of the encountered challenges is provided along with an outlook for applying these inorganic nanoparticles toward future clinical translations.
Collapse
Affiliation(s)
- Guangbao Yang
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Soo Zeng Fiona Phua
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Anivind Kaur Bindra
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Yanli Zhao
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| |
Collapse
|
24
|
Tieu T, Alba M, Elnathan R, Cifuentes‐Rius A, Voelcker NH. Advances in Porous Silicon–Based Nanomaterials for Diagnostic and Therapeutic Applications. ADVANCED THERAPEUTICS 2018. [DOI: 10.1002/adtp.201800095] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Terence Tieu
- Monash Institute of Pharmaceutical Sciences Monash University Parkville Campus, 381 Royal Parade Parkville Victoria 3052 Australia
- T. Tieu, Dr. M. Alba, Prof. N. H. Voelcker CSIRO Manufacturing Bayview Avenue Clayton Victoria 3168 Australia
| | - Maria Alba
- Monash Institute of Pharmaceutical Sciences Monash University Parkville Campus, 381 Royal Parade Parkville Victoria 3052 Australia
- T. Tieu, Dr. M. Alba, Prof. N. H. Voelcker CSIRO Manufacturing Bayview Avenue Clayton Victoria 3168 Australia
| | - Roey Elnathan
- Monash Institute of Pharmaceutical Sciences Monash University Parkville Campus, 381 Royal Parade Parkville Victoria 3052 Australia
| | - Anna Cifuentes‐Rius
- Monash Institute of Pharmaceutical Sciences Monash University Parkville Campus, 381 Royal Parade Parkville Victoria 3052 Australia
| | - Nicolas H. Voelcker
- Monash Institute of Pharmaceutical Sciences Monash University Parkville Campus, 381 Royal Parade Parkville Victoria 3052 Australia
- Prof. N. H. Voelcker Melbourne Centre for Nanofabrication Victorian Node of the Australian National Fabrication Facility 151 Wellington Road Clayton Victoria 3168 Australia
- T. Tieu, Dr. M. Alba, Prof. N. H. Voelcker CSIRO Manufacturing Bayview Avenue Clayton Victoria 3168 Australia
| |
Collapse
|
25
|
Thomson AW, Ezzelarab MB. Regulatory dendritic cells: profiling, targeting, and therapeutic application. Curr Opin Organ Transplant 2018; 23:538-545. [PMID: 30036199 PMCID: PMC6620776 DOI: 10.1097/mot.0000000000000565] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE OF REVIEW There is currently increased focus on improved understanding of how dendritic cell tolerogenicity is determined and maintained, and on their therapeutic potential. We review recent progress in profiling of regulatory dendritic cells (DCreg), innovative approaches to enhancing dendritic cell tolerogenicity in situ, ex-vivo generation of DCreg and initial clinical testing of these cells in organ transplantation. RECENT FINDINGS "Omics' studies indicate that the distinctive properties of DCreg are the result of a specific transcriptional program characterized by activation of tolerance-enhancing genes, rather than the retention of an immature state. In situ dendritic cell-directed targeting of nanovesicles bearing immune regulatory molecules can trigger in-vivo expansion of Ag-specific regulatory cells. Innovative approaches to ex-vivo modification of dendritic cells to enhance their regulatory function and capacity to migrate to secondary lymphoid organs has been described. Cross-dressing (with donor major histocompatibility complex molecules) of graft-infiltrating host dendritic cells that regulate antidonor T-cell responses has been implicated in "spontaneous' liver transplant tolerance. Clinical trials of DCreg therapy have begun in living donor renal and liver transplantation. SUMMARY Further definition of molecules that can be targeted to promote the function and stability of DCreg in vivo may lead to standardization of DCreg manufacturing for therapeutic application.
Collapse
Affiliation(s)
- Angus W Thomson
- Department of Surgery, Thomas E. Starzl Transplantation Institute
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
26
|
Stead SO, Kireta S, McInnes SJP, Kette FD, Sivanathan KN, Kim J, Cueto-Diaz EJ, Cunin F, Durand JO, Drogemuller CJ, Carroll RP, Voelcker NH, Coates PT. Murine and Non-Human Primate Dendritic Cell Targeting Nanoparticles for in Vivo Generation of Regulatory T-Cells. ACS NANO 2018; 12:6637-6647. [PMID: 29979572 DOI: 10.1021/acsnano.8b01625] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Porous silicon nanoparticles (pSiNP), modified to target dendritic cells (DC), provide an alternate strategy for the delivery of immunosuppressive drugs. Here, we aimed to develop a DC-targeting pSiNP displaying c-type lectin, dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN), and CD11c monoclonal antibodies. The in vivo tracking of these fluorescent DC-targeting nanoparticles was assessed in both C57BL/6 mice and common marmosets ( Callithrix jacchus) by intravenous injection (20 mg/kg). Rapamycin and ovalbumin (OVA)323-339 peptide loaded pSiNP were employed to evaluate their ability to generate murine CD4+CD25+FoxP3+ regulatory T-cells in vivo within OVA sensitized mice. In vivo, pSiNP migrated to the liver, kidneys, lungs, and spleen in both mice and marmosets. Flow cytometry confirmed pSiNP uptake by splenic and peripheral blood DC when functionalized with targeting antibodies. C57BL/6 OVA sensitized mice injected with CD11c-pSiNP loaded with rapamycin + OVA323-339 produced a 5-fold higher number of splenic regulatory T-cells compared to control mice, at 40 days post-pSiNP injection. These results demonstrate the importance of the immobilized targeting antibodies to enhance cellular uptake and enable the in vivo generation of splenic regulatory T-cells.
Collapse
Affiliation(s)
- Sebastian O Stead
- Department of Medicine , University of Adelaide , Adelaide 5000 , Australia
| | - Svjetlana Kireta
- Central Northern Adelaide Renal and Transplantation Service (CNARTS) , The Royal Adelaide Hospital , Adelaide 5000 , Australia
| | - Steve J P McInnes
- Future Industries Institute , University of South Australia , Adelaide 5095 , Australia
| | - Francis D Kette
- Department of Medicine , University of Adelaide , Adelaide 5000 , Australia
| | - Kisha N Sivanathan
- Department of Medicine , University of Adelaide , Adelaide 5000 , Australia
| | - Juewan Kim
- Department of Medicine , University of Adelaide , Adelaide 5000 , Australia
| | - Eduardo J Cueto-Diaz
- Case 1701, UMR 5253 CNRS -ENSCM-UM , Institut Charles Gerhardt Montpellier , 34095 Montpellier , cedex 5, France
| | - Frederique Cunin
- Case 1701, UMR 5253 CNRS -ENSCM-UM , Institut Charles Gerhardt Montpellier , 34095 Montpellier , cedex 5, France
| | - Jean-Olivier Durand
- Case 1701, UMR 5253 CNRS -ENSCM-UM , Institut Charles Gerhardt Montpellier , 34095 Montpellier , cedex 5, France
| | - Christopher J Drogemuller
- Department of Medicine , University of Adelaide , Adelaide 5000 , Australia
- Central Northern Adelaide Renal and Transplantation Service (CNARTS) , The Royal Adelaide Hospital , Adelaide 5000 , Australia
| | - Robert P Carroll
- Department of Medicine , University of Adelaide , Adelaide 5000 , Australia
- Central Northern Adelaide Renal and Transplantation Service (CNARTS) , The Royal Adelaide Hospital , Adelaide 5000 , Australia
| | - Nicolas H Voelcker
- Drug Delivery, Disposition, and Dynamics, Monash Institute of Pharmaceutical Sciences , Monash University , 381 Royal Parade , Parkville , Victoria 3052 , Australia
- Commonwealth Scientific and Industrial Research Organization (CSIRO) , Clayton , Victoria 3169 , Australia
- Melbourne Center for Nanofabrication, Victorian Node of the Australian National Fabrication Facility , Clayton , Victoria 3168 , Australia
- Monash Institute of Medical Engineering , Monash University , Clayton , Victoria 3800 , Australia
| | - Patrick T Coates
- Department of Medicine , University of Adelaide , Adelaide 5000 , Australia
- Central Northern Adelaide Renal and Transplantation Service (CNARTS) , The Royal Adelaide Hospital , Adelaide 5000 , Australia
| |
Collapse
|
27
|
Electrospun Composites of Polycaprolactone and Porous Silicon Nanoparticles for the Tunable Delivery of Small Therapeutic Molecules. NANOMATERIALS 2018; 8:nano8040205. [PMID: 29596352 PMCID: PMC5923535 DOI: 10.3390/nano8040205] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 03/26/2018] [Accepted: 03/27/2018] [Indexed: 12/31/2022]
Abstract
This report describes the use of an electrospun composite of poly(ε-caprolactone) (PCL) fibers and porous silicon (pSi) nanoparticles (NPs) as an effective system for the tunable delivery of camptothecin (CPT), a small therapeutic molecule. Both materials are biodegradable, abundant, low-cost, and most importantly, have no known cytotoxic effects. The composites were treated with and without sodium hydroxide (NaOH) to investigate the wettability of the porous network for drug release and cell viability measurements. CPT release and subsequent cell viability was also investigated. We observed that the cell death rate was not only affected by the addition of our CPT carrier, pSi, but also by increasing the rate of dissolution via treatment with NaOH. This is the first example of loading pSi NPs as a therapeutics nanocarrier into electronspun PCL fibers and this system opens up new possibilities for the delivery of molecular therapeutics.
Collapse
|