101
|
Modulation of immune responses in lentiviral vector-mediated gene transfer. Cell Immunol 2018; 342:103802. [PMID: 29735164 PMCID: PMC6695505 DOI: 10.1016/j.cellimm.2018.04.012] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 04/25/2018] [Accepted: 04/26/2018] [Indexed: 02/07/2023]
Abstract
Lentiviral vectors (LV) are widely used vehicles for gene transfer and therapy in pre-clinical animal models and clinical trials with promising safety and efficacy results. However, host immune responses against vector- and/or transgene-derived antigens remain a major obstacle to the success and broad applicability of gene therapy. Here we review the innate and adaptive immunological barriers to successful gene therapy, both in the context of ex vivo and in vivo LV gene therapy, mostly concerning systemic LV delivery and discuss possible means to overcome them, including vector design and production and immune modulatory strategies.
Collapse
|
102
|
CD4+ αβ T cell infiltration into the leptomeninges of lumbar dorsal roots contributes to the transition from acute to chronic mechanical allodynia after adult rat tibial nerve injuries. J Neuroinflammation 2018; 15:81. [PMID: 29544518 PMCID: PMC5855984 DOI: 10.1186/s12974-018-1115-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 03/05/2018] [Indexed: 02/06/2023] Open
Abstract
Background Antigen-specific and MHCII-restricted CD4+ αβ T cells have been shown or suggested to play an important role in the transition from acute to chronic mechanical allodynia after peripheral nerve injuries. However, it is still largely unknown where these T cells infiltrate along the somatosensory pathways transmitting mechanical allodynia to initiate the development of chronic mechanical allodynia after nerve injuries. Therefore, the purpose of this study was to ascertain the definite neuroimmune interface for these T cells to initiate the development of chronic mechanical allodynia after peripheral nerve injuries. Methods First, we utilized both chromogenic and fluorescent immunohistochemistry (IHC) to map αβ T cells along the somatosensory pathways for the transmission of mechanical allodynia after modified spared nerve injuries (mSNIs), i.e., tibial nerve injuries, in adult male Sprague-Dawley rats. We further characterized the molecular identity of these αβ T cells selectively infiltrating into the leptomeninges of L4 dorsal roots (DRs). Second, we identified the specific origins in lumbar lymph nodes (LLNs) for CD4+ αβ T cells selectively present in the leptomeninges of L4 DRs by two experiments: (1) chromogenic IHC in these lymph nodes for CD4+ αβ T cell responses after mSNIs and (2) fluorescent IHC for temporal dynamics of CD4+ αβ T cell infiltration into the L4 DR leptomeninges after mSNIs in prior lymphadenectomized or sham-operated animals to LLNs. Finally, following mSNIs, we evaluated the effects of region-specific targeting of these T cells through prior lymphadenectomy to LLNs and chronic intrathecal application of the suppressive anti-αβTCR antibodies on the development of mechanical allodynia by von Frey hair test and spinal glial or neuronal activation by fluorescent IHC. Results Our results showed that during the sub-acute phase after mSNIs, αβ T cells selectively infiltrate into the leptomeninges of the lumbar DRs along the somatosensory pathways responsible for transmitting mechanical allodynia. Almost all these αβ T cells are CD4 positive. Moreover, the temporal dynamics of CD4+ αβ T cell infiltration into the lumbar DR leptomeninges are specifically determined by LLNs after mSNIs. Prior lymphadenectomy to LLNs specifically reduces the development of mSNI-induced chronic mechanical allodynia. More importantly, intrathecal application of the suppressive anti-αβTCR antibodies reduces the development of mSNI-induced chronic mechanical allodynia. In addition, prior lymphadenectomy to LLNs attenuates mSNI-induced spinal activation of glial cells and PKCγ+ excitatory interneurons. Conclusions The noteworthy results here provide the first evidence that CD4+ αβ T cells selectively infiltrate into the DR leptomeninges of the somatosensory pathways transmitting mechanical allodynia and contribute to the transition from acute to chronic mechanical allodynia after peripheral nerve injuries. Electronic supplementary material The online version of this article (10.1186/s12974-018-1115-7) contains supplementary material, which is available to authorized users.
Collapse
|
103
|
Allen RP, Bolandparvaz A, Ma JA, Manickam VA, Lewis JS. Latent, Immunosuppressive Nature of Poly(lactic- co-glycolic acid) Microparticles. ACS Biomater Sci Eng 2018; 4:900-918. [PMID: 30555893 PMCID: PMC6290919 DOI: 10.1021/acsbiomaterials.7b00831] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Use of biomaterials to spatiotemporally control the activation of immune cells is at the forefront of biomedical engineering research. As more biomaterial strategies are employed for immunomodulation, understanding the immunogenicity of biodegradable materials and their byproducts is paramount in tailoring systems for immune activation or suppression. Poly(D,L-lactic-co-glycolic acid) (PLGA), one of the most commonly studied polymers in tissue engineering and drug delivery, has been previously described on one hand as an immune adjuvant, and on the other as a nonactivating material. In this study, the effect of PLGA microparticles (MPs) on the maturation status of murine bone marrow-derived dendritic cells (DCs), the primary initiators of adaptive immunity, was investigated to decipher the immunomodulatory properties of this biomaterial. Treatment of bone marrow-derived DCs from C57BL/6 mice with PLGA MPs led to a time dependent decrease in the maturation level of these cells, as quantified by decreased expression of the positive stimulatory molecules MHCII, CD80, and CD86 as well as the ability to resist maturation following challenge with lipopolysaccharide (LPS). Moreover, this immunosuppression was dependent on the molecular weight of the PLGA used to fabricate the MPs, as higher molecular weight polymers required longer incubation to produce comparable dampening of maturation molecules. These phenomena were correlated to an increase in lactic acid both intracellularly and extracellularly during DC/PLGA MP coculture, which is postulated to be the primary agent behind the observed immune inhibition. This hypothesis is supported by our results demonstrating that resistance to LPS stimulation may be due to the ability of PLGA MP-derived lactic acid to inhibit the phosphorylation of TAK1 and therefore prevent NF-κB activation. This work is significant as it begins to elucidate how PLGA, a prominent biomaterial with broad applications ranging from tissue engineering to pharmaceutics, could modulate the local immune environment and offers insight on engineering PLGA to exploit its evolving immunogenicity.
Collapse
Affiliation(s)
- Riley P. Allen
- Department of Biomedical Engineering, University of California Davis, 1 Shields Avenue, Davis, California 95616, United States
| | - Amir Bolandparvaz
- Department of Biomedical Engineering, University of California Davis, 1 Shields Avenue, Davis, California 95616, United States
| | - Jeffrey A. Ma
- Department of Biomedical Engineering, University of California Davis, 1 Shields Avenue, Davis, California 95616, United States
| | - Vishal A. Manickam
- Department of Biomedical Engineering, University of California Davis, 1 Shields Avenue, Davis, California 95616, United States
| | - Jamal S. Lewis
- Department of Biomedical Engineering, University of California Davis, 1 Shields Avenue, Davis, California 95616, United States
| |
Collapse
|
104
|
LaMothe RA, Kolte PN, Vo T, Ferrari JD, Gelsinger TC, Wong J, Chan VT, Ahmed S, Srinivasan A, Deitemeyer P, Maldonado RA, Kishimoto TK. Tolerogenic Nanoparticles Induce Antigen-Specific Regulatory T Cells and Provide Therapeutic Efficacy and Transferrable Tolerance against Experimental Autoimmune Encephalomyelitis. Front Immunol 2018; 9:281. [PMID: 29552007 PMCID: PMC5840162 DOI: 10.3389/fimmu.2018.00281] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 01/31/2018] [Indexed: 12/16/2022] Open
Abstract
T cells reacting to self-components can promote tissue damage when escaping tolerogenic control mechanisms which may result in autoimmune disease. The current treatments for these disorders are not antigen (Ag) specific and can compromise host immunity through chronic suppression. We have previously demonstrated that co-administration of encapsulated or free Ag with tolerogenic nanoparticles (tNPs) comprised of biodegradable polymers that encapsulate rapamycin are capable of inhibiting Ag-specific transgenic T cell proliferation and inducing Ag-specific regulatory T cells (Tregs). Here, we further show that tNPs can trigger the expansion of endogenous Tregs specific to a target Ag. The proportion of Ag-specific Treg to total Ag-specific T cells remains constant even after subsequent Ag challenge in combination with a potent TLR7/8 agonist or complete Freund’s adjuvant. tNP-treated mice do not develop experimental autoimmune encephalomyelitis (EAE) after adoptive transfer of encephalitogenic T cells; furthermore, tNP treatment provided therapeutic protection in relapsing EAE that was transferred to naïve animals. These findings describe a potent therapy to expand Ag-specific Tregs in vivo and suppress T cell-mediated autoimmunity.
Collapse
Affiliation(s)
| | | | - Trinh Vo
- Selecta Biosciences Inc., Watertown, MA, United States
| | | | | | - Jodie Wong
- Selecta Biosciences Inc., Watertown, MA, United States
| | - Victor T Chan
- Selecta Biosciences Inc., Watertown, MA, United States
| | - Sinthia Ahmed
- Selecta Biosciences Inc., Watertown, MA, United States
| | | | | | | | | |
Collapse
|
105
|
Abstract
Antigen-specific immune tolerance has been a long-standing goal for immunotherapy for the treatment of autoimmune diseases and allergies and for the prevention of allograft rejection and anti-drug antibodies directed against biologic therapies. Nanoparticles have emerged as powerful tools to initiate and modulate immune responses due to their inherent capacity to target antigen-presenting cells (APCs) and deliver coordinated signals that can elicit an antigen-specific immune response. A wide range of strategies have been described to create tolerogenic nanoparticles (tNPs) that fall into three broad categories. One strategy includes tNPs that provide antigen alone to harness natural tolerogenic processes and environments, such as presentation of antigen in the absence of costimulatory signals, oral tolerance, the tolerogenic environment of the liver, and apoptotic cell death. A second strategy includes tNPs that carry antigen and simultaneously target tolerogenic receptors, such as pro-tolerogenic cytokine receptors, aryl hydrocarbon receptor, FAS receptor, and the CD22 inhibitory receptor. A third strategy includes tNPs that carry a payload of tolerogenic pharmacological agents that can “lock” APCs into a developmental or metabolic state that favors tolerogenic presentation of antigens. These diverse strategies have led to the development of tNPs that are capable of inducing antigen-specific immunological tolerance, not just immunosuppression, in animal models. These novel tNP technologies herald a promising approach to specifically prevent and treat unwanted immune reactions in humans. The first tNP, SEL-212, a biodegradable synthetic vaccine particle encapsulating rapamycin, has reached the clinic and is currently in Phase 2 clinical trials.
Collapse
|
106
|
Sahoo JK, Braegelman AS, Webber MJ. Immunoengineering with Supramolecular Peptide Biomaterials. J Indian Inst Sci 2018. [DOI: 10.1007/s41745-018-0060-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
107
|
Bookstaver ML, Tsai SJ, Bromberg JS, Jewell CM. Improving Vaccine and Immunotherapy Design Using Biomaterials. Trends Immunol 2018; 39:135-150. [PMID: 29249461 PMCID: PMC5914493 DOI: 10.1016/j.it.2017.10.002] [Citation(s) in RCA: 128] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 09/29/2017] [Accepted: 10/03/2017] [Indexed: 12/13/2022]
Abstract
Polymers, lipids, scaffolds, microneedles, and other biomaterials are rapidly emerging as technologies to improve the efficacy of vaccines against infectious disease and immunotherapies for cancer, autoimmunity, and transplantation. New studies are also providing insight into the interactions between these materials and the immune system. This insight can be exploited for more efficient design of vaccines and immunotherapies. Here, we describe recent advances made possible through the unique features of biomaterials, as well as the important questions for further study.
Collapse
Affiliation(s)
- Michelle L Bookstaver
- Fischell Department of Bioengineering, University of Maryland, 8228 Paint Branch Drive, College Park, MD 20742, USA
| | - Shannon J Tsai
- Fischell Department of Bioengineering, University of Maryland, 8228 Paint Branch Drive, College Park, MD 20742, USA
| | - Jonathan S Bromberg
- Department of Surgery, University of Maryland School of Medicine, 29 South Greene Street, Baltimore, MD 21201, USA; Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, 800 West Baltimore Street, Baltimore, MD 21201, USA; Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201, USA; Marlene and Stewart Greenebaum Cancer Center, 22 South Greene Street, Baltimore, MD 21201, USA.
| | - Christopher M Jewell
- Fischell Department of Bioengineering, University of Maryland, 8228 Paint Branch Drive, College Park, MD 20742, USA; Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201, USA; Marlene and Stewart Greenebaum Cancer Center, 22 South Greene Street, Baltimore, MD 21201, USA; United States Department of Veteran Affairs, 10 North Greene Street, Baltimore, MD 21201, USA.
| |
Collapse
|
108
|
Dangi A, Luo X. Harnessing Apoptotic Cells for Transplantation Tolerance: Current Status and Future Perspectives. CURRENT TRANSPLANTATION REPORTS 2017; 4:270-279. [PMID: 29177124 PMCID: PMC5697727 DOI: 10.1007/s40472-017-0167-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
PURPOSE OF REVIEW The use of donor apoptotic cells is an emerging therapy for inducing transplantation tolerance. In this review, we will discuss current understanding of mechanisms of this approach, as well as crucial aspects necessary for successful translation of this approach to clinical transplantation. RECENT FINDINGS Transplantation tolerance by donor apoptotic cells is mediated by their homeostatic interaction with recipient phagocytes, and subsequent expansion of suppressor cell populations as well as inhibition of effector T cells via deletion and anergy. To ensure their tolerogenicity, it is critical to procure non-stressed donor cells, and to induce and arrest their apoptosis at the appropriate stage prior to their administration. Equally important is the monitoring of dynamics of recipient immunological status, and its influences on tolerance efficacy and longevity. Emerging concepts and technologies may significantly streamline tolerogen manufacture and delivery of this approach, and smooth its transition to clinical application. SUMMARY Hijacking homeostatic clearance of donor apoptotic cells is a promising strategy for transplantation tolerance. Timing is now mature for concerted efforts for transitioning this strategy to clinical transplantation.
Collapse
Affiliation(s)
- Anil Dangi
- Center for Kidney Research and Therapeutics, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Division of Nephrology and Hypertension, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Xunrong Luo
- Center for Kidney Research and Therapeutics, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Division of Nephrology and Hypertension, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
109
|
Dacoba TG, Olivera A, Torres D, Crecente-Campo J, Alonso MJ. Modulating the immune system through nanotechnology. Semin Immunol 2017; 34:78-102. [PMID: 29032891 PMCID: PMC5774666 DOI: 10.1016/j.smim.2017.09.007] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 09/18/2017] [Accepted: 09/18/2017] [Indexed: 12/11/2022]
Abstract
Nowadays, nanotechnology-based modulation of the immune system is presented as a cutting-edge strategy, which may lead to significant improvements in the treatment of severe diseases. In particular, efforts have been focused on the development of nanotechnology-based vaccines, which could be used for immunization or generation of tolerance. In this review, we highlight how different immune responses can be elicited by tuning nanosystems properties. In addition, we discuss specific formulation approaches designed for the development of anti-infectious and anti-autoimmune vaccines, as well as those intended to prevent the formation of antibodies against biologicals.
Collapse
Affiliation(s)
- Tamara G Dacoba
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain; Department of Pharmacology, Pharmacy and Pharmaceutical Technology, School of Pharmacy, Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Ana Olivera
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain; Department of Pharmacology, Pharmacy and Pharmaceutical Technology, School of Pharmacy, Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Dolores Torres
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, School of Pharmacy, Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - José Crecente-Campo
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain; Department of Pharmacology, Pharmacy and Pharmaceutical Technology, School of Pharmacy, Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain.
| | - María José Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain; Department of Pharmacology, Pharmacy and Pharmaceutical Technology, School of Pharmacy, Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain.
| |
Collapse
|
110
|
Casey LM, Pearson RM, Hughes KR, Liu JMH, Rose JA, North MG, Wang LZ, Lei M, Miller SD, Shea LD. Conjugation of Transforming Growth Factor Beta to Antigen-Loaded Poly(lactide- co-glycolide) Nanoparticles Enhances Efficiency of Antigen-Specific Tolerance. Bioconjug Chem 2017; 29:813-823. [PMID: 29148731 DOI: 10.1021/acs.bioconjchem.7b00624] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Current strategies for treating autoimmunity involve the administration of broad-acting immunosuppressive agents that impair healthy immunity. Intravenous (i.v.) administration of poly(lactide- co-glycolide) nanoparticles (NPs) containing disease-relevant antigens (Ag-NPs) have demonstrated antigen (Ag)-specific immune tolerance in models of autoimmunity. However, subcutaneous (s.c.) delivery of Ag-NPs has not been effective. This investigation tested the hypothesis that codelivery of the immunomodulatory cytokine, transforming growth factor beta 1 (TGF-β), on Ag-NPs would modulate the immune response to Ag-NPs and improve the efficiency of tolerance induction. TGF-β was coupled to the surface of Ag-NPs such that the loadings of Ag and TGF-β were independently tunable. The particles demonstrated bioactive delivery of Ag and TGF-β in vitro by reducing the inflammatory phenotype of bone marrow-derived dendritic cells and inducing regulatory T cells in a coculture system. Using an in vivo mouse model for multiple sclerosis, experimental autoimmune encephalomyelitis, TGF-β codelivery on Ag-NPs resulted in improved efficacy at lower doses by i.v. administration and significantly reduced disease severity by s.c. administration. This study demonstrates that the codelivery of immunomodulatory cytokines on Ag-NPs may enhance the efficacy of Ag-specific tolerance therapies by programming Ag presenting cells for more efficient tolerance induction.
Collapse
Affiliation(s)
- Liam M Casey
- Department of Chemical Engineering , University of Michigan , 2300 Hayward Avenue , Ann Arbor , Michigan 48105 , United States
| | - Ryan M Pearson
- Department of Biomedical Engineering , University of Michigan , 1119 Carl A. Gerstacker Building, 2200 Bonisteel Boulevard , Ann Arbor , Michigan 48109-2099 , United States.,Cour Pharmaceuticals, Northbrook , Illinois 60062 , United States
| | - Kevin R Hughes
- Department of Biomedical Engineering , University of Michigan , 1119 Carl A. Gerstacker Building, 2200 Bonisteel Boulevard , Ann Arbor , Michigan 48109-2099 , United States
| | - Jeffrey M H Liu
- Department of Biomedical Engineering , University of Michigan , 1119 Carl A. Gerstacker Building, 2200 Bonisteel Boulevard , Ann Arbor , Michigan 48109-2099 , United States
| | - Justin A Rose
- Department of Chemical Engineering , University of Michigan , 2300 Hayward Avenue , Ann Arbor , Michigan 48105 , United States
| | - Madeleine G North
- Department of Biomedical Engineering , University of Michigan , 1119 Carl A. Gerstacker Building, 2200 Bonisteel Boulevard , Ann Arbor , Michigan 48109-2099 , United States
| | - Leon Z Wang
- Department of Biomedical Engineering , University of Michigan , 1119 Carl A. Gerstacker Building, 2200 Bonisteel Boulevard , Ann Arbor , Michigan 48109-2099 , United States
| | - Mei Lei
- Department of Biomedical Engineering , University of Michigan , 1119 Carl A. Gerstacker Building, 2200 Bonisteel Boulevard , Ann Arbor , Michigan 48109-2099 , United States
| | - Stephen D Miller
- Department of Microbiology-Immunology, Feinberg School of Medicine , Northwestern University , 6-713 Tarry Building, 303 East Chicago Avenue , Chicago , Illinois 60611 , United States.,The Robert H. Lurie Comprehensive Cancer Center of Northwestern University , Chicago , Illinois 60611 , United States
| | - Lonnie D Shea
- Department of Chemical Engineering , University of Michigan , 2300 Hayward Avenue , Ann Arbor , Michigan 48105 , United States.,Department of Biomedical Engineering , University of Michigan , 1119 Carl A. Gerstacker Building, 2200 Bonisteel Boulevard , Ann Arbor , Michigan 48109-2099 , United States
| |
Collapse
|
111
|
Pujol-Autonell I, Mansilla MJ, Rodriguez-Fernandez S, Cano-Sarabia M, Navarro-Barriuso J, Ampudia RM, Rius A, Garcia-Jimeno S, Perna-Barrull D, Martinez-Caceres E, Maspoch D, Vives-Pi M. Liposome-based immunotherapy against autoimmune diseases: therapeutic effect on multiple sclerosis. Nanomedicine (Lond) 2017; 12:1231-1242. [PMID: 28593827 DOI: 10.2217/nnm-2016-0410] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM Based on the ability of apoptosis to induce immunological tolerance, liposomes were generated mimicking apoptotic cells, and they arrest autoimmunity in Type 1 diabetes. Our aim was to validate the immunotherapy in other autoimmune disease: multiple sclerosis. MATERIALS & METHODS Phosphatidylserine-rich liposomes were loaded with disease-specific autoantigen. Therapeutic capability of liposomes was assessed in vitro and in vivo. RESULTS Liposomes induced a tolerogenic phenotype in dendritic cells, and arrested autoimmunity, thus decreasing the incidence, delaying the onset and reducing the severity of experimental disease, correlating with an increase in a probably regulatory CD25+ FoxP3- CD4+ T-cell subset. CONCLUSION This is the first work that confirms phosphatidylserine-liposomes as a powerful tool to arrest multiple sclerosis, demonstrating its relevance for clinical application.
Collapse
Affiliation(s)
- Irma Pujol-Autonell
- Immunology Division, Germans Trias i Pujol University Hospital & Research Institute, Department of Cellular Biology, Physiology & Immunology, Autonomous University of Barcelona, 08916 Badalona, Spain
| | - Maria-Jose Mansilla
- Immunology Division, Germans Trias i Pujol University Hospital & Research Institute, Department of Cellular Biology, Physiology & Immunology, Autonomous University of Barcelona, 08916 Badalona, Spain
| | - Silvia Rodriguez-Fernandez
- Immunology Division, Germans Trias i Pujol University Hospital & Research Institute, Department of Cellular Biology, Physiology & Immunology, Autonomous University of Barcelona, 08916 Badalona, Spain
| | - Mary Cano-Sarabia
- Catalan Institute of Nanoscience & Nanotechnology (ICN2), CSIC & The Barcelona Institute of Science & Technology, 08193 Bellaterra, Barcelona, Spain
| | - Juan Navarro-Barriuso
- Immunology Division, Germans Trias i Pujol University Hospital & Research Institute, Department of Cellular Biology, Physiology & Immunology, Autonomous University of Barcelona, 08916 Badalona, Spain
| | - Rosa-Maria Ampudia
- Immunology Division, Germans Trias i Pujol University Hospital & Research Institute, Department of Cellular Biology, Physiology & Immunology, Autonomous University of Barcelona, 08916 Badalona, Spain
| | - Aleix Rius
- Immunology Division, Germans Trias i Pujol University Hospital & Research Institute, Department of Cellular Biology, Physiology & Immunology, Autonomous University of Barcelona, 08916 Badalona, Spain
| | - Sonia Garcia-Jimeno
- Catalan Institute of Nanoscience & Nanotechnology (ICN2), CSIC & The Barcelona Institute of Science & Technology, 08193 Bellaterra, Barcelona, Spain
| | - David Perna-Barrull
- Immunology Division, Germans Trias i Pujol University Hospital & Research Institute, Department of Cellular Biology, Physiology & Immunology, Autonomous University of Barcelona, 08916 Badalona, Spain
| | - Eva Martinez-Caceres
- Immunology Division, Germans Trias i Pujol University Hospital & Research Institute, Department of Cellular Biology, Physiology & Immunology, Autonomous University of Barcelona, 08916 Badalona, Spain
| | - Daniel Maspoch
- Catalan Institute of Nanoscience & Nanotechnology (ICN2), CSIC & The Barcelona Institute of Science & Technology, 08193 Bellaterra, Barcelona, Spain.,Institucio Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
| | - Marta Vives-Pi
- Immunology Division, Germans Trias i Pujol University Hospital & Research Institute, Department of Cellular Biology, Physiology & Immunology, Autonomous University of Barcelona, 08916 Badalona, Spain.,CIBER of Diabetes & Associated Metabolic Diseases (CIBERDEM), ISCIII, Madrid, Spain
| |
Collapse
|
112
|
Co-delivery of autoantigen and dexamethasone in incomplete Freund's adjuvant ameliorates experimental autoimmune encephalomyelitis. J Control Release 2017; 266:156-165. [PMID: 28963036 DOI: 10.1016/j.jconrel.2017.09.034] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 09/10/2017] [Accepted: 09/25/2017] [Indexed: 12/15/2022]
Abstract
Current therapies for autoimmune diseases focus on treating the symptoms rather than the underlying disease cause. A major setback in improving current therapeutics for autoimmunity is the lack of antigen specificity. Successful antigen-specific immunotherapy (ASIT) would allow for improved treatment of autoimmune diseases. In this work, dexamethasone was co-delivered with autoantigen (PLP) in vivo to create effective ASIT for the treatment of experimental autoimmune encephalomyelitis (EAE). Using an emulsion of incomplete Freund's adjuvant (IFA) as a co-delivery vehicle, it was discovered that the controlled release of autoantigen was important for the suppression of clinical disease symptoms. Analysis of the immune response via cytokines revealed that dexamethasone was important for shifting the immune response away from inflammation. Co-delivery of both autoantigen and dexamethasone increased B-cell populations and antibody production, signifying an increased humoral immune response. Overall, this data indicated that the co-delivery of PLP and dexamethasone with a water-in-oil emulsion is effective in treating a murine autoimmune model.
Collapse
|
113
|
Balmert SC, Donahue C, Vu JR, Erdos G, Falo LD, Little SR. In vivo induction of regulatory T cells promotes allergen tolerance and suppresses allergic contact dermatitis. J Control Release 2017; 261:223-233. [DOI: 10.1016/j.jconrel.2017.07.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 07/06/2017] [Indexed: 11/26/2022]
|
114
|
Solomon M, Muro S. Lysosomal enzyme replacement therapies: Historical development, clinical outcomes, and future perspectives. Adv Drug Deliv Rev 2017; 118:109-134. [PMID: 28502768 PMCID: PMC5828774 DOI: 10.1016/j.addr.2017.05.004] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 04/26/2017] [Accepted: 05/08/2017] [Indexed: 01/06/2023]
Abstract
Lysosomes and lysosomal enzymes play a central role in numerous cellular processes, including cellular nutrition, recycling, signaling, defense, and cell death. Genetic deficiencies of lysosomal components, most commonly enzymes, are known as "lysosomal storage disorders" or "lysosomal diseases" (LDs) and lead to lysosomal dysfunction. LDs broadly affect peripheral organs and the central nervous system (CNS), debilitating patients and frequently causing fatality. Among other approaches, enzyme replacement therapy (ERT) has advanced to the clinic and represents a beneficial strategy for 8 out of the 50-60 known LDs. However, despite its value, current ERT suffers from several shortcomings, including various side effects, development of "resistance", and suboptimal delivery throughout the body, particularly to the CNS, lowering the therapeutic outcome and precluding the use of this strategy for a majority of LDs. This review offers an overview of the biomedical causes of LDs, their socio-medical relevance, treatment modalities and caveats, experimental alternatives, and future treatment perspectives.
Collapse
Affiliation(s)
- Melani Solomon
- Institute for Bioscience and Biotechnology Research, University Maryland, College Park, MD 20742, USA
| | - Silvia Muro
- Institute for Bioscience and Biotechnology Research, University Maryland, College Park, MD 20742, USA; Fischell Department of Bioengineering, University Maryland, College Park, MD 20742, USA.
| |
Collapse
|
115
|
Cho JJ, Stewart JM, Drashansky TT, Brusko MA, Zuniga AN, Lorentsen KJ, Keselowsky BG, Avram D. An antigen-specific semi-therapeutic treatment with local delivery of tolerogenic factors through a dual-sized microparticle system blocks experimental autoimmune encephalomyelitis. Biomaterials 2017; 143:79-92. [PMID: 28772190 DOI: 10.1016/j.biomaterials.2017.07.029] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/11/2017] [Accepted: 07/21/2017] [Indexed: 01/07/2023]
Abstract
Antigen-specific treatments are highly desirable for autoimmune diseases in contrast to treatments which induce systemic immunosuppression. A novel antigen-specific therapy has been developed which, when administered semi-therapeutically, is highly efficacious in the treatment of the mouse model for multiple sclerosis, experimental autoimmune encephalomyelitis (EAE). The treatment uses dual-sized, polymeric microparticles (dMPs) loaded with specific antigen and tolerizing factors for intra- and extra-cellular delivery, designed to recruit and modulate dendritic cells toward a tolerogenic phenotype without systemic release. This approach demonstrated robust efficacy and provided complete protection against disease. Therapeutic efficacy required encapsulation of the factors in controlled-release microparticles and was antigen-specific. Disease blocking was associated with a reduction of infiltrating CD4+ T cells, inflammatory cytokine-producing pathogenic CD4+ T cells, and activated macrophages and microglia in the central nervous system. Furthermore, CD4+ T cells isolated from dMP-treated mice were anergic in response to disease-specific, antigen-loaded splenocytes. Additionally, the frequency of CD86hiMHCIIhi dendritic cells in draining lymph nodes of EAE mice treated with Ag-specific dMPs was reduced. Our findings highlight the efficacy of microparticle-based drug delivery platform to mediate antigen-specific tolerance, and suggest that such a multi-factor combinatorial approach can act to block autoimmunity.
Collapse
Affiliation(s)
- Jonathan J Cho
- Division of Pulmonary Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Joshua M Stewart
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Theodore T Drashansky
- Division of Pulmonary Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Maigan A Brusko
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Ashley N Zuniga
- Division of Pulmonary Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Kyle J Lorentsen
- Division of Pulmonary Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Benjamin G Keselowsky
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA.
| | - Dorina Avram
- Division of Pulmonary Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
116
|
Gammon JM, Adapa AR, Jewell CM. Control of autoimmune inflammation using liposomes to deliver positive allosteric modulators of metabotropic glutamate receptors. J Biomed Mater Res A 2017. [DOI: 10.1002/jbm.a.36151] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Joshua M. Gammon
- Fischell Department of BioengineeringUniversity of MarylandCollege Park Maryland
| | - Arjun R. Adapa
- Fischell Department of BioengineeringUniversity of MarylandCollege Park Maryland
| | - Christopher M. Jewell
- Fischell Department of BioengineeringUniversity of MarylandCollege Park Maryland
- Department of Microbiology and ImmunologyUniversity of Maryland Medical SchoolBaltimore Maryland
- Marlene and Stewart Greenebaum Cancer CenterBaltimore Maryland
- University States Department of Veteran AffairsBaltimore Maryland
| |
Collapse
|
117
|
Hess KL, Oh E, Tostanoski LH, Andorko JI, Susumu K, Deschamps JR, Medintz IL, Jewell CM. Engineering Immunological Tolerance Using Quantum Dots to Tune the Density of Self-Antigen Display. ADVANCED FUNCTIONAL MATERIALS 2017; 27:1700290. [PMID: 29503604 PMCID: PMC5828250 DOI: 10.1002/adfm.201700290] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Treatments for autoimmunity - diseases where the immune system mistakenly attacks self-molecules - are not curative and leave patients immunocompromised. New studies aimed at more specific treatments reveal development of inflammation or tolerance is influenced by the form self-antigens are presented. Using a mouse model of multiple sclerosis (MS), we show for the first time that quantum dots (QDs) can be used to generate immunological tolerance by controlling the density of self-antigen on QDs. These assemblies display dense arrangements of myelin self-peptide associated with disease in MS, are uniform in size (<20 nm), and allow direct visualization in immune tissues. Peptide-QDs rapidly concentrate in draining lymph nodes, co-localizing with macrophages expressing scavenger receptors involved in tolerance. Treatment with peptide-QDs reduces disease incidence 10-fold. Strikingly, the degree of tolerance - and the underlying expansion of regulatory T cells - correlates with the density of myelin molecules presented on QDs. A key discovery is that higher numbers of tolerogenic particles displaying lower levels of self-peptide are more effective for inducing tolerance than fewer particles each displaying higher densities of peptide. QDs conjugated with self-antigens could serve as a new platform to induce tolerance, while visualizing QD therapeutics in tolerogenic tissue domains.
Collapse
Affiliation(s)
- Krystina L Hess
- Fischell Department of Bioengineering, University of Maryland, 8228 Paint Branch Drive, College Park, MD 20742, USA
| | - Eunkeu Oh
- Optical Sciences Division, Code 5600, U.S. Naval Research Laboratory, 4555 Overlook Ave, SW, Washington DC 20375, USA
| | - Lisa H Tostanoski
- Fischell Department of Bioengineering, University of Maryland, 8228 Paint Branch Drive, College Park, MD 20742, USA
| | - James I Andorko
- Fischell Department of Bioengineering, University of Maryland, 8228 Paint Branch Drive, College Park, MD 20742, USA
| | - Kimihiro Susumu
- Optical Sciences Division, Code 5600, U.S. Naval Research Laboratory, 4555 Overlook Ave, SW, Washington DC 20375, USA
| | - Jeffrey R Deschamps
- Center for Bio/Molecular Science and Engineering Code 6900 U.S. Naval Research Laboratory, 4555 Overlook Ave SW, Washington DC 20375, USA
| | - Igor L Medintz
- Center for Bio/Molecular Science and Engineering Code 6900 U.S. Naval Research Laboratory, 4555 Overlook Ave SW, Washington DC 20375, USA
| | - Christopher M Jewell
- Fischell Department of Bioengineering, University of Maryland, 8228 Paint Branch Drive, College Park, MD 20742, USA
| |
Collapse
|
118
|
Andorko JI, Jewell CM. Designing biomaterials with immunomodulatory properties for tissue engineering and regenerative medicine. Bioeng Transl Med 2017; 2:139-155. [PMID: 28932817 PMCID: PMC5579731 DOI: 10.1002/btm2.10063] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 04/14/2017] [Accepted: 04/24/2017] [Indexed: 12/29/2022] Open
Abstract
Recent research in the vaccine and immunotherapy fields has revealed that biomaterials have the ability to activate immune pathways, even in the absence of other immune-stimulating signals. Intriguingly, new studies reveal these responses are influenced by the physicochemical properties of the material. Nearly all of this work has been done in the vaccine and immunotherapy fields, but there is tremendous opportunity to apply this same knowledge to tissue engineering and regenerative medicine. This review discusses recent findings that reveal how material properties-size, shape, chemical functionality-impact immune response, and links these changes to emerging opportunities in tissue engineering and regenerative medicine. We begin by discussing what has been learned from studies conducted in the contexts of vaccines and immunotherapies. Next, research is highlighted that elucidates the properties of materials that polarize innate immune cells, including macrophages and dendritic cells, toward either inflammatory or wound healing phenotypes. We also discuss recent studies demonstrating that scaffolds used in tissue engineering applications can influence cells of the adaptive immune system-B and T cell lymphocytes-to promote regenerative tissue microenvironments. Through greater study of the intrinsic immunogenic features of implantable materials and scaffolds, new translational opportunities will arise to better control tissue engineering and regenerative medicine applications.
Collapse
Affiliation(s)
- James I. Andorko
- Fischell Department of BioengineeringUniversity of MarylandCollege ParkMD 20742
| | - Christopher M. Jewell
- Fischell Department of BioengineeringUniversity of MarylandCollege ParkMD 20742
- Department of Microbiology and ImmunologyUniversity of Maryland Medical SchoolBaltimoreMD 21201
- Marlene and Stewart Greenebaum Cancer CenterBaltimoreMD 21201
- United States Department of Veterans AffairsBaltimoreMD 21201
| |
Collapse
|
119
|
Tostanoski LH, Jewell CM. Engineering self-assembled materials to study and direct immune function. Adv Drug Deliv Rev 2017; 114:60-78. [PMID: 28392305 PMCID: PMC6262758 DOI: 10.1016/j.addr.2017.03.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 03/21/2017] [Accepted: 03/22/2017] [Indexed: 12/19/2022]
Abstract
The immune system is an awe-inspiring control structure that maintains a delicate and constantly changing balance between pro-immune functions that fight infection and cancer, regulatory or suppressive functions involved in immune tolerance, and homeostatic resting states. These activities are determined by integrating signals in space and time; thus, improving control over the densities, combinations, and durations with which immune signals are delivered is a central goal to better combat infectious disease, cancer, and autoimmunity. Self-assembly presents a unique opportunity to synthesize materials with well-defined compositions and controlled physical arrangement of molecular building blocks. This review highlights strategies exploiting these capabilities to improve the understanding of how precisely-displayed cues interact with immune cells and tissues. We present work centered on fundamental properties that regulate the nature and magnitude of immune response, highlight pre-clinical and clinical applications of self-assembled technologies in vaccines, cancer, and autoimmunity, and describe some of the key manufacturing and regulatory hurdles facing these areas.
Collapse
Key Words
- Autoimmunity and tolerance
- Biomaterial
- Cancer
- Immunomodulation
- Manufacturing, regulatory approval and FDA
- Nanoparticle, microparticle, micelle, liposome, polyplex, lipoplex, polyelectrolyte multilayer
- Nanotechnology
- Non-covalent, hydrophobic, hydrogen bonding, and electrostatic interaction
- Self-assembly
- Sensor, diagnostic, and theranostic
- Vaccine and immunotherapy
Collapse
Affiliation(s)
- Lisa H Tostanoski
- Fischell Department of Bioengineering, University of Maryland, 8228 Paint Branch Drive, College Park, MD 20742, USA
| | - Christopher M Jewell
- Fischell Department of Bioengineering, University of Maryland, 8228 Paint Branch Drive, College Park, MD 20742, USA; Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201, USA; Marlene and Stewart Greenebaum Cancer Center, 22 S. Greene St., Baltimore, MD 21201, USA; United States Department of Veterans Affairs, 10 North Greene Street, Baltimore, MD 21201, USA.
| |
Collapse
|
120
|
Baekkeskov S, Hubbell JA, Phelps EA. Bioengineering strategies for inducing tolerance in autoimmune diabetes. Adv Drug Deliv Rev 2017. [PMID: 28625830 DOI: 10.1016/j.addr.2017.06.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Type 1 diabetes is an autoimmune disease marked by the destruction of insulin-producing beta cells in the pancreatic islets. Strategies to delay onset or prevent the autoimmune recognition of beta cell antigens or T cell-mediated killing of beta cells have mainly focused on systemic immunomodulation and antigen-specific immunotherapy. To bridge the fields of type 1 diabetes immunology and biomaterials engineering, this article will review recent trends in the etiology of type 1 diabetes immunopathology and will focus on the contributions of emerging bioengineered strategies in the fight against beta cell autoimmunity in type 1 diabetes.
Collapse
Affiliation(s)
- Steinunn Baekkeskov
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Building SV 3826, Station 19, CH-1015 Lausanne, Switzerland; Departments of Medicine and Microbiology/Immunology, Diabetes Center, 513 Parnassus Ave, 20159, Box 0534, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Jeffrey A Hubbell
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Building SV 3826, Station 19, CH-1015 Lausanne, Switzerland; Institute for Molecular Engineering, University of Chicago, 5640 S Ellis Avenue, Chicago, IL 60615, USA
| | - Edward A Phelps
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Building SV 3826, Station 19, CH-1015 Lausanne, Switzerland; J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Drive, P.O. Box 116131, Gainesville, FL 32611, USA.
| |
Collapse
|
121
|
Combinatorial drug delivery approaches for immunomodulation. Adv Drug Deliv Rev 2017; 114:161-174. [PMID: 28532690 DOI: 10.1016/j.addr.2017.05.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 05/05/2017] [Accepted: 05/17/2017] [Indexed: 12/19/2022]
Abstract
Immunotherapy has been widely explored for applications to both augment and suppress intrinsic host immunity. Clinical achievements have seen a number of immunotherapeutic drugs displace established strategies like chemotherapy in treating immune-associated diseases. However, single drug approaches modulating an individual arm of the immune system are often incompletely effective. Imperfect mechanistic understanding and heterogeneity within disease pathology have seen monotherapies inadequately equipped to mediate complete disease remission. Recent success in applications of combinatorial immunotherapy has suggested that targeting multiple biological pathways simultaneously may be critical in treating complex immune pathologies. Drug delivery approaches through engineered biomaterials offer the potential to augment desired immune responses while mitigating toxic side-effects by localizing immunotherapy. This review discusses recent advances in immunotherapy and highlights newly explored combinatorial drug delivery approaches. Furthermore, prospective future directions for immunomodulatory drug delivery to exploit are provided.
Collapse
|
122
|
Gosselin EA, Tostanoski LH, Jewell CM. Controlled Release of Second Generation mTOR Inhibitors to Restrain Inflammation in Primary Immune Cells. AAPS JOURNAL 2017; 19:1175-1185. [PMID: 28484962 DOI: 10.1208/s12248-017-0089-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Accepted: 04/14/2017] [Indexed: 12/20/2022]
Abstract
Autoimmune disease occurs when the immune system incorrectly targets the body's own tissue. Inflammatory CD4+ T cell phenotypes, such as TH1 and TH17, are key drivers of this attack. Recent studies demonstrate treatment with rapamycin-a key inhibitor of the mTOR pathway-can skew T cell development, moving T cell responses away from inflammatory phenotypes and toward regulatory T cells (TREGS). TREGS are important in inducing and maintaining tolerance to self-antigens, creating new potential to treat autoimmune diseases more effectively and specifically. Next generation analogs of rapamycin, such as everolimus and temsirolimus, confer increased potency with reduced toxicity, but are understudied in the context of autoimmunity. Further, these drugs are still broadly-acting and require frequent treatment due to short half-lives. Thus, there is strong interest in harnessing the unique properties of biomaterials-controlled drug release and targeting, for example, to improve autoimmune therapies. Using second generation mTOR inhibitors and rapamycin, we prepared sets of degradable polymer particles from poly(lactide-co-glycolide). We then used these materials to assess physicochemical properties and the ability to control autoimmune inflammation in a primary cell co-culture model. Treatment with particle formulations resulted in significant dose-dependent decreases in dendritic cell activation, T cell proliferation, inflammatory cytokines, and frequencies of inflammatory TH1 phenotypes. Considering the current limitations of rapamycin, and the potential of next-generation analogs, this work provides a screening platform for biomaterials and sets the stage for in vivo evaluation, where delivery kinetics, stability, and targeting could improve autoimmune therapies through biomaterial-enabled delivery.
Collapse
Affiliation(s)
- Emily A Gosselin
- Fischell Department of Bioengineering, University of Maryland, 2212 Jeong H. Kim Engineering Building, 8228 Paint Branch Drive, College Park, Maryland, 20742, USA
| | - Lisa H Tostanoski
- Fischell Department of Bioengineering, University of Maryland, 2212 Jeong H. Kim Engineering Building, 8228 Paint Branch Drive, College Park, Maryland, 20742, USA
| | - Christopher M Jewell
- Fischell Department of Bioengineering, University of Maryland, 2212 Jeong H. Kim Engineering Building, 8228 Paint Branch Drive, College Park, Maryland, 20742, USA. .,Department of Microbiology and Immunology, University of Maryland Medical School, Baltimore, Maryland, USA. .,Marlene and Stewart Greenebaum Cancer Center, Baltimore, Maryland, USA. .,United States Department of Veterans Affairs, Baltimore, Maryland, USA.
| |
Collapse
|
123
|
Nanoparticles for immune system targeting. Drug Discov Today 2017; 22:1295-1301. [PMID: 28390214 DOI: 10.1016/j.drudis.2017.03.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 02/18/2017] [Accepted: 03/29/2017] [Indexed: 02/02/2023]
Abstract
Nanoparticles (NPs) are found in numerous applications used to modulate the immune system. They serve as drug delivery carriers or vaccine adjuvants and are utilized as therapeutics against a variety of diseases. NPs can be engineered to target distinct cellular components representing multiple pathways of immunity. The combination of NPs with immune system-targeting moieties has paved the way for improved targeted immune therapies. Here we provide an update of recent progress in this field.
Collapse
|
124
|
Chen N, Peine KJ, Collier MA, Gautam S, Jablonski KA, Guerau-de-Arellano M, Ainslie KM, Bachelder EM. Co-Delivery of Disease Associated Peptide and Rapamycin via Acetalated Dextran Microparticles for Treatment of Multiple Sclerosis. ACTA ACUST UNITED AC 2017. [DOI: 10.1002/adbi.201700022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Naihan Chen
- Division of Pharmacoengineering and Molecular Pharmaceutics; Eshelman School of Pharmacy; The University of North Carolina at Chapel Hill; Chapel Hill NC 27599 USA
| | - Kevin J. Peine
- Division of Pharmacoengineering and Molecular Pharmaceutics; Eshelman School of Pharmacy; The University of North Carolina at Chapel Hill; Chapel Hill NC 27599 USA
| | - Michael A. Collier
- Division of Pharmacoengineering and Molecular Pharmaceutics; Eshelman School of Pharmacy; The University of North Carolina at Chapel Hill; Chapel Hill NC 27599 USA
| | - Shalini Gautam
- Division of Pharmaceutics; College of Pharmacy; The Ohio State University; Columbus OH 43210 USA
| | - Kyle A. Jablonski
- Medical Laboratory Science Division; School of Health and Rehabilitation Sciences; The Ohio State University; Columbus OH 43210 USA
| | - Mireia Guerau-de-Arellano
- Medical Laboratory Science Division; School of Health and Rehabilitation Sciences; The Ohio State University; Columbus OH 43210 USA
| | - Kristy M. Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics; Eshelman School of Pharmacy; The University of North Carolina at Chapel Hill; Chapel Hill NC 27599 USA
| | - Eric M. Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics; Eshelman School of Pharmacy; The University of North Carolina at Chapel Hill; Chapel Hill NC 27599 USA
| |
Collapse
|
125
|
Gammon JM, Gosselin EA, Tostanoski LH, Chiu YC, Zeng X, Zeng Q, Jewell CM. Low-dose controlled release of mTOR inhibitors maintains T cell plasticity and promotes central memory T cells. J Control Release 2017; 263:151-161. [PMID: 28257991 DOI: 10.1016/j.jconrel.2017.02.034] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 02/25/2017] [Accepted: 02/27/2017] [Indexed: 12/12/2022]
Abstract
An important goal for improving vaccine and immunotherapy technologies is the ability to provide further control over the specific phenotypes of T cells arising from these agents. Along these lines, frequent administration of rapamycin (Rapa), a small molecule inhibitor of the mammalian target of rapamycin (mTOR), exhibits a striking ability to polarize T cells toward central memory phenotypes (TCM), or to suppress immune function, depending on the concentrations and other signals present during administration. TCM exhibit greater plasticity and proliferative capacity than effector memory T cells (TEFF) and, therefore, polarizing vaccine-induced T cells toward TCM is an intriguing strategy to enhance T cell expansion and function against pathogens or tumors. Here we combined biodegradable microparticles encapsulating Rapa (Rapa MPs) with vaccines composed of soluble peptide antigens and molecular adjuvants to test if this approach allows polarization of differentiating T cells toward TCM. We show Rapa MPs modulate DC function, enhancing secretion of inflammatory cytokines at very low doses, and suppressing function at high doses. While Rapa MP treatment reduced - but did not stop - T cell proliferation in both CD4+ and CD8+ transgenic T cell co-cultures, the expanding CD8+ T cells differentiated to higher frequencies of TCM at low doses of MP Rapa MPs. Lastly, we show in mice that local delivery of Rapa MPs to lymph nodes during vaccination either suppresses or enhances T cell function in response to melanoma antigens, depending on the dose of drug in the depots. In particular, at low Rapa MP doses, vaccines increased antigen-specific TCM, resulting in enhanced T cell expansion measured during subsequent booster injections over at least 100days.
Collapse
Affiliation(s)
- Joshua M Gammon
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Emily A Gosselin
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Lisa H Tostanoski
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Yu-Chieh Chiu
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Xiangbin Zeng
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Qin Zeng
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Christopher M Jewell
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States; Department of Microbiology and Immunology, University of Maryland Medical School, Baltimore, MD, United States; Marlene and Stewart Greenebaum Cancer Center, Baltimore, MD, United States; United States Department of Veterans Affairs, Baltimore, MD, United States.
| |
Collapse
|
126
|
Yang T, Wang S, Yang X, Zheng Q, Wang L, Li Q, Wei M, Du Z, Fan Y. Upregulation of Bcl-2 and Its Promoter Signals in CD4+ T Cells during Neuromyelitis Optica Remission. Front Neurosci 2017; 11:11. [PMID: 28174515 PMCID: PMC5258721 DOI: 10.3389/fnins.2017.00011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 01/09/2017] [Indexed: 12/27/2022] Open
Abstract
The homeostatic balance between production and elimination of CD4+ T cells in peripheral blood plays an important role in patients with neuromyelitis optica (NMO). The objective of the present study was to evaluate the anti-apoptosis genes Bcl-2 and its promoter signal (nuclear factor kappa-light-chain-enhancer of activated B cells, NFκB) in CD4+ T cells. Healthy subjects (HS, n = 25) and patients with multiple sclerosis (MS) (n = 25) and NMO (n = 30) in remission were consecutively enrolled in this prospective study between May and December 2015. CD4+ T cells were isolated using magnetic beads coated with anti-CD4 monoclonal antibodies, and gene expression of Bcl-2, NFκB, phosphatidylinositol-4, 5-bisphosphate 3-kinase/protein kinase B (PI3K/Akt), and MAP kinase kinase kinase 7 (MAP3K7) was measured by real-time reverse transcription-polymerase chain reaction (rt-PCR). Cytokines of tumor necrosis factor (TNF)-α and interleukin (IL)-1β were detected using human cytokine multiplex assay. Bcl-2 and NFκB gene expressions were elevated in NMO patients (1.63 ± 0.25; 2.35 ± 0.25) compared with those of HS (0.90 ± 0.11; 1.42 ± 0.22) and/or MS patients (1.03 ± 0.18; 1.55 ± 0.20) (P < 0.05). MAP3K7, but not Akt, was increased in NMO patients (1.23 ± 0.18; 1.56 ± 0.22) (P < 0.01) and was a significant factor related to elevated NFκB gene expressions (P < 0.001). On the other hand, IL-1β and TNF-α were also detected in the study and the results showed that both were elevated in NMO patients (23.84 ± 1.81; 56.40 ± 2.45) (P < 0.01; P < 0.05, respectively). We propose that MAP3K7 induced by IL-1β and TNF-α but not Akt promotes NFκB expression and, in turn, prolongs Bcl-2-mediated survival of CD4+ T cells in NMO patients.
Collapse
Affiliation(s)
- Tao Yang
- Department of Traditional Chinese Medicine, Beijing Tiantan Hospital, Capital Medical University Beijing, China
| | - Su Wang
- Department of Tumor Radiotherapy, Hiser Medical Center of Qingdao Qingdao, China
| | - Xiao Yang
- School of Management Science and Engineering, Shandong University of Finance and Economics Jinan, China
| | - Qi Zheng
- Department of Oncology, Guang An Men Hospital of China Academy of Chinese Medical SciencesBeijing, China; School of Traditional Chinese Medicine, Capital Medical UniversityBeijing, China
| | - Lei Wang
- School of Traditional Chinese Medicine, Capital Medical University Beijing, China
| | - Qian Li
- Department of Traditional Chinese Medicine, Beijing Tiantan Hospital, Capital Medical University Beijing, China
| | - Mingyan Wei
- Department of Traditional Chinese Medicine, Beijing Tiantan Hospital, Capital Medical University Beijing, China
| | - Zongpan Du
- Department of Traditional Chinese Medicine, Beijing Tiantan Hospital, Capital Medical University Beijing, China
| | - Yongping Fan
- Department of Traditional Chinese Medicine, Beijing Tiantan Hospital, Capital Medical University Beijing, China
| |
Collapse
|
127
|
Zhang P, Bookstaver ML, Jewell CM. Engineering Cell Surfaces with Polyelectrolyte Materials for Translational Applications. Polymers (Basel) 2017; 9:E40. [PMID: 30970718 PMCID: PMC6431965 DOI: 10.3390/polym9020040] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Revised: 01/18/2017] [Accepted: 01/19/2017] [Indexed: 11/16/2022] Open
Abstract
Engineering cell surfaces with natural or synthetic materials is a unique and powerful strategy for biomedical applications. Cells exhibit more sophisticated migration, control, and functional capabilities compared to nanoparticles, scaffolds, viruses, and other engineered materials or agents commonly used in the biomedical field. Over the past decade, modification of cell surfaces with natural or synthetic materials has been studied to exploit this complexity for both fundamental and translational goals. In this review we present the existing biomedical technologies for engineering cell surfaces with one important class of materials, polyelectrolytes. We begin by introducing the challenges facing the cell surface engineering field. We then discuss the features of polyelectrolytes and how these properties can be harnessed to solve challenges in cell therapy, tissue engineering, cell-based drug delivery, sensing and tracking, and immune modulation. Throughout the review, we highlight opportunities to drive the field forward by bridging new knowledge of polyelectrolytes with existing translational challenges.
Collapse
Affiliation(s)
- Peipei Zhang
- Fischell Department of Bioengineering, University of Maryland, College Park, MA 20742, USA.
| | - Michelle L Bookstaver
- Fischell Department of Bioengineering, University of Maryland, College Park, MA 20742, USA.
| | - Christopher M Jewell
- Fischell Department of Bioengineering, University of Maryland, College Park, MA 20742, USA.
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MA 21201, USA.
- Marlene and Stewart Greenebaum Cancer Center, Baltimore, MA 21201, USA.
- United States Department of Veterans Affairs, Baltimore, MA 21201, USA.
| |
Collapse
|
128
|
Northrup L, Sullivan BP, Hartwell BL, Garza A, Berkland C. Screening Immunomodulators To Skew the Antigen-Specific Autoimmune Response. Mol Pharm 2016; 14:66-80. [PMID: 28043135 DOI: 10.1021/acs.molpharmaceut.6b00725] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Current therapies to treat autoimmune diseases often result in side effects such as nonspecific immunosuppression. Therapies that can induce antigen-specific immune tolerance provide an opportunity to reverse autoimmunity and mitigate the risks associated with global immunosuppression. In an effort to induce antigen-specific immune tolerance, co-administration of immunomodulators with autoantigens has been investigated in an effort to reprogram autoimmunity. To date, identifying immunomodulators that may skew the antigen-specific immune response has been ad hoc at best. To address this need, we utilized splenocytes obtained from mice with experimental autoimmune encephalomyelitis (EAE) in order to determine if certain immunomodulators may induce markers of immune tolerance following antigen rechallenge. Of the immunomodulatory compounds investigated, only dexamethasone modified the antigen-specific immune response by skewing the cytokine response and decreasing T-cell populations at a concentration corresponding to a relevant in vivo dose. Thus, antigen-educated EAE splenocytes provide an ex vivo screen for investigating compounds capable of skewing the antigen-specific immune response, and this approach could be extrapolated to antigen-educated cells from other diseases or human tissues.
Collapse
Affiliation(s)
- Laura Northrup
- Department of Pharmaceutical Chemistry, University of Kansas , Lawrence, Kansas 66047, United States
| | - Bradley P Sullivan
- Department of Pharmaceutical Chemistry, University of Kansas , Lawrence, Kansas 66047, United States
| | - Brittany L Hartwell
- Bioengineering Graduate Program, University of Kansas , Lawrence, Kansas 66045, United States
| | - Aaron Garza
- Department of Chemical and Petroleum Engineering, University of Kansas , Lawrence, Kansas 66045, United States
| | - Cory Berkland
- Department of Pharmaceutical Chemistry, University of Kansas , Lawrence, Kansas 66047, United States.,Bioengineering Graduate Program, University of Kansas , Lawrence, Kansas 66045, United States.,Department of Chemical and Petroleum Engineering, University of Kansas , Lawrence, Kansas 66045, United States
| |
Collapse
|
129
|
Hess KL, Andorko JI, Tostanoski LH, Jewell CM. Polyplexes assembled from self-peptides and regulatory nucleic acids blunt toll-like receptor signaling to combat autoimmunity. Biomaterials 2016; 118:51-62. [PMID: 27940382 DOI: 10.1016/j.biomaterials.2016.11.052] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 11/27/2016] [Indexed: 01/09/2023]
Abstract
Autoimmune diseases occur when the immune system incorrectly recognizes self-molecules as foreign; in the case of multiple sclerosis (MS), myelin is attacked. Intriguingly, new studies reveal toll-like receptors (TLRs), pathways usually involved in generating immune responses against pathogens, play a significant role in driving autoimmune disease in both humans and animal models. We reasoned polyplexes formed from myelin self-antigen and regulatory TLR antagonists might limit TLR signaling during differentiation of myelin-specific T cells, inducing tolerance by biasing T cells away from inflammatory phenotypes. Complexes were formed by modifying myelin peptide with cationic amino acids to create peptides able to condense the anionic nucleic-acid based TLR antagonist. These immunological polyplexes eliminate synthetic polymers commonly used to condense polyplexes and do not rely on gene expression; however, the complexes mimic key features of traditional polyplexes such as tunable loading and co-delivery. Using these materials and classic polyplex analysis techniques, we demonstrate condensation of both immune signals, protection from enzymatic degradation, and tunable physicochemical properties. We show polyplexes reduce TLR signaling, and in primary dendritic cell and T cell co-culture, reduce myelin-driven inflammation. During mouse models of MS, these tolerogenic polyplexes improve the progression, severity, and incidence of disease.
Collapse
Affiliation(s)
- Krystina L Hess
- Fischell Department of Bioengineering, University of Maryland, 8228 Paint Branch Drive, College Park, MD 20742, USA
| | - James I Andorko
- Fischell Department of Bioengineering, University of Maryland, 8228 Paint Branch Drive, College Park, MD 20742, USA
| | - Lisa H Tostanoski
- Fischell Department of Bioengineering, University of Maryland, 8228 Paint Branch Drive, College Park, MD 20742, USA
| | - Christopher M Jewell
- Fischell Department of Bioengineering, University of Maryland, 8228 Paint Branch Drive, College Park, MD 20742, USA; Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201, USA; Marlene and Stewart Greenebaum Cancer Center, 22 S. Greene Street, Baltimore, MD 21201, USA; United States Department of Veterans Affairs, 10 North Greene Street, Baltimore, Maryland 21201, USA.
| |
Collapse
|