Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Immunol ; 208(4): 929-940, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35091434

ABSTRACT

CD8+ T cell responses are the foundation of the recent clinical success of immunotherapy in oncologic indications. Although checkpoint inhibitors have enhanced the activity of existing CD8+ T cell responses, therapeutic approaches to generate Ag-specific CD8+ T cell responses have had limited success. Here, we demonstrate that cytosolic delivery of Ag through microfluidic squeezing enables MHC class I presentation to CD8+ T cells by diverse cell types. In murine dendritic cells (DCs), squeezed DCs were ∼1000-fold more potent at eliciting CD8+ T cell responses than DCs cross-presenting the same amount of protein Ag. The approach also enabled engineering of less conventional APCs, such as T cells, for effective priming of CD8+ T cells in vitro and in vivo. Mixtures of immune cells, such as murine splenocytes, also elicited CD8+ T cell responses in vivo when squeezed with Ag. We demonstrate that squeezing enables effective MHC class I presentation by human DCs, T cells, B cells, and PBMCs and that, in clinical scale formats, the system can squeeze up to 2 billion cells per minute. Using the human papillomavirus 16 (HPV16) murine model, TC-1, we demonstrate that squeezed B cells, T cells, and unfractionated splenocytes elicit antitumor immunity and correlate with an influx of HPV-specific CD8+ T cells such that >80% of CD8s in the tumor were HPV specific. Together, these findings demonstrate the potential of cytosolic Ag delivery to drive robust CD8+ T cell responses and illustrate the potential for an autologous cell-based vaccine with minimal turnaround time for patients.


Subject(s)
Antigen Presentation , Antigen-Presenting Cells/immunology , CD8-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class I/immunology , Microfluidics , Neoplasms/immunology , Adoptive Transfer , Animals , Antigen-Presenting Cells/metabolism , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Culture Techniques , Female , Humans , Immunization , Immunophenotyping , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice , Mice, Knockout , Microfluidics/methods , Models, Biological , Neoplasms/metabolism , Neoplasms/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
2.
Biomaterials ; 76: 1-10, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26513216

ABSTRACT

Allogeneic cell therapies have either proven effective or have great potential in numerous applications, though the required systemic, life-long immunosuppression presents significant health risks. Inducing tolerance to allogeneic cells offers the potential to reduce or eliminate chronic immunosuppression. Herein, we investigated antigen-loaded nanoparticles for their ability to promote transplant tolerance in the minor histocompatibility antigen sex-mismatched C57BL/6 model of bone marrow transplantation. In this model, the peptide antigens Dby and Uty mediate rejection of male bone marrow transplants by female CD4+ and CD8+ T cells, respectively, and we investigated the action of nanoparticles on these T cell subsets. Antigens were coupled to or encapsulated within poly(lactide-co-glycolide) (PLG) nanoparticles with an approximate diameter of 500 nm. Delivery of the CD4-encoded Dby epitope either coupled to or encapsulated within PLG particles prevented transplant rejection, promoted donor-host chimerism, and suppressed proliferative and IFN-γ responses in tolerized recipients. Nanoparticles modified with the Uty peptide did not induce tolerance. The dosing regimen was investigated with Dby coupled particles, and a single dose delivered the day after bone marrow transplant was sufficient for tolerance induction. The engraftment of cells was significantly affected by PD-1/PDL-1 costimluation, as blockade of PD-1 reduced engraftment by ∼50%. In contrast, blockade of regulatory T cells did not impact the level of chimerism. The delivery of antigen on PLG nanoparticles promoted long-term engraftment of bone marrow in a model with a minor antigen mismatch in the absence of immunosuppression, and this represents a promising platform for developing a translatable, donor-specific tolerance strategy.


Subject(s)
Bone Marrow Transplantation , H-Y Antigen/administration & dosage , Immune Tolerance , Nanoparticles , Peptides/administration & dosage , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Female , H-Y Antigen/chemistry , Male , Mice , Mice, Inbred C57BL , Polyglactin 910/administration & dosage
3.
Biomaterials ; 80: 11-19, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26701143

ABSTRACT

Biomaterial scaffolds are central to many regenerative strategies as they create a space for infiltration of host tissue and provide a platform to deliver growth factors and progenitor cells. However, biomaterial implantation results in an unavoidable inflammatory response, which can impair tissue regeneration and promote loss or dysfunction of transplanted cells. We investigated localized TGF-ß1 delivery to modulate this immunological environment around scaffolds and transplanted cells. TGF-ß1 was delivered from layered scaffolds, with protein entrapped within an inner layer and outer layers designed for cell seeding and host tissue integration. Scaffolds were implanted into the epididymal fat pad, a site frequently used for cell transplantation. Expression of cytokines TNF-α, IL-12, and MCP-1 were decreased by at least 40% for scaffolds releasing TGF-ß1 relative to control scaffolds. This decrease in inflammatory cytokine production corresponded to a 60% decrease in leukocyte infiltration. Transplantation of islets into diabetic mice on TGF-ß1 scaffolds significantly improved the ability of syngeneic islets to control blood glucose levels within the first week of transplant and delayed rejection of allogeneic islets. Together, these studies emphasize the ability of localized TGF-ß1 delivery to modulate the immune response to biomaterial implants and enhance cell function in cell-based therapies.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/therapeutic use , Diabetes Mellitus, Experimental/therapy , Islets of Langerhans Transplantation/methods , Tissue Scaffolds/chemistry , Transforming Growth Factor beta1/administration & dosage , Transforming Growth Factor beta1/therapeutic use , Animals , Cells, Cultured , Chemokine CCL2/immunology , Diabetes Mellitus, Experimental/immunology , Drug Delivery Systems/methods , Immunomodulation/drug effects , Interleukin-12/immunology , Male , Mice , Mice, Inbred C57BL , Porosity , Tumor Necrosis Factor-alpha/immunology
4.
Clin Immunol ; 160(1): 14-23, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25805659

ABSTRACT

The induction of donor-specific tolerance to transplanted cells and organs, while preserving immune function as a whole, remains a highly sought after and elusive strategy for overcoming transplant rejection. Tolerance necessitates modulating a diverse array of cell types that recognize and respond to alloantigens, including antigen presenting cells and T lymphocytes. Nanotherapeutic strategies that employ cellular and biomaterial engineering represent an emerging technology geared towards the goal of inducing transplant tolerance. Nanocarriers offer a platform for delivering antigens of interest to specific cell types in order to achieve tolerogenic antigen presentation. Furthermore, the technologies also provide an opportunity for local immunomodulation at the graft site. Nanocarriers delivering a combination of antigens and immunomodulating agents, such as rapamycin, provide a unique technology platform with the potential to enhance outcomes for the induction of transplant tolerance.


Subject(s)
Antigen-Presenting Cells/immunology , Antigens/immunology , Graft Rejection/immunology , Molecular Targeted Therapy/methods , Nanomedicine/methods , Transplantation Tolerance/immunology , Antigen Presentation/immunology , Drug Carriers/therapeutic use , Graft Survival/immunology , Humans , Immunomodulation/immunology , Immunosuppressive Agents/therapeutic use , Sirolimus/therapeutic use , Tacrolimus/therapeutic use
5.
Biomaterials ; 35(31): 8887-8894, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25066477

ABSTRACT

Human islet cell transplantation is a promising treatment for type 1 diabetes; however, long-term donor-specific tolerance to islet allografts remains a clinically unmet goal. We have previously shown that recipient infusions of apoptotic donor splenocytes chemically treated with 1-ethyl-3-(3'-dimethylaminopropyl)-carbodiimide (donor ECDI-SP) can mediate long-term acceptance of full major histocompatibility complex (MHC)-mismatched murine islet allografts without the use of immunosuppression. In this report, we investigated the use of poly(lactide-co-glycolide) (PLG) particles in lieu of donor ECDI-SP as a synthetic, cell-free carrier for delivery of donor antigens for the induction of transplant tolerance in full MHC-mismatched murine allogeneic islet transplantation. Infusions of donor antigen-coupled PLG particles (PLG-dAg) mediated tolerance in ∼20% of recipient mice, and the distribution of cellular uptake of PLG-dAg within the spleen was similar to that of donor ECDI-SP. PLG-dAg mediated the contraction of indirectly activated T cells but did not modulate the direct pathway of allorecognition. Combination of PLG-dAg with a short course of low dose immunosuppressant rapamycin at the time of transplant significantly improved the tolerance efficacy to ∼60%. Furthermore, altering the timing of PLG-dAg administration to a schedule that is more feasible for clinical transplantation resulted in equal tolerance efficacy. Thus, the combination therapy of PLG-dAg infusions with peritransplant rapamycin represents a clinically attractive, biomaterials-based and cell-free method for inducing long-term donor-specific tolerance for allogeneic cell transplantation, such as for allogeneic islet transplantation.


Subject(s)
Antigens/administration & dosage , Drug Carriers/chemistry , Immune Tolerance/drug effects , Islets of Langerhans Transplantation/methods , Polyglactin 910/chemistry , Animals , Antigens/therapeutic use , Ethyldimethylaminopropyl Carbodiimide/chemistry , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/therapeutic use , Male , Mice, Inbred BALB C , Nanoparticles/chemistry , Sirolimus/administration & dosage , Sirolimus/therapeutic use , Transplantation, Homologous
SELECTION OF CITATIONS
SEARCH DETAIL
...