Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Bioconjug Chem ; 31(12): 2779-2788, 2020 12 16.
Article in English | MEDLINE | ID: mdl-33284575

ABSTRACT

Diseases of immunity, including autoimmune diseases such as multiple sclerosis, transplantation graft rejection, allergy, and asthma, are prevalent and increasing in prevalence. They contribute to significant morbidity and mortality; however, few if any curative therapies exist, and those that are available lack either potency or specificity. Dendritic cells (DCs) are sentinels of the immune system that connect the innate and adaptive immune system and are critical regulators of both immunity and tolerance. We posited that the tolerogenic potential of DC could be harnessed to develop more specific and potent therapies for diseases of immunity by delivering autoantigen to a sufficient number of tolerogenic DCs in situ that could then inhibit pathogenic effector T cell responses. Specifically, we hypothesized that the steroid dexamethasone covalently coupled to a peptide antigen could be processed by DCs, induce tolerogenic DCs, and attenuate antigen-specific pathogenic T cell responses. To test this hypothesis, we synthesized a series of dexamethasone-peptide immunoconjugates by standard solid-phase peptide synthesis. The antigenic portion of the immunoconjugate could be presented by DCs, and the immunoconjugate induced a tolerogenic phenotype in DCs that then inhibited antigen-specific T cell proliferation in vitro. When the immunoconjugate was administered prophylactically in the murine experimental autoimmune encephalomyelitis model of multiple sclerosis, disease was attenuated compared to dexamethasone and peptide delivered as uncoupled components. Together, this work demonstrates the utility of immunoconjugates for inducing tolerance while establishing the foundation for future studies exploring methods to enrich and target DCs for tolerogenic therapies.


Subject(s)
Dexamethasone/chemistry , Encephalomyelitis, Autoimmune, Experimental/immunology , Immunoconjugates/chemistry , Immunoconjugates/immunology , Multiple Sclerosis/immunology , Peptides/chemistry , T-Lymphocytes/immunology , Adaptive Immunity/immunology , Animals , Encephalomyelitis, Autoimmune, Experimental/therapy , Immunity, Innate/immunology , Mice , Multiple Sclerosis/therapy
2.
Nat Commun ; 11(1): 5696, 2020 11 10.
Article in English | MEDLINE | ID: mdl-33173046

ABSTRACT

Poorly immunogenic tumors, including triple negative breast cancers (TNBCs), remain resistant to current immunotherapies, due in part to the difficulty of reprogramming the highly immunosuppressive tumor microenvironment (TME). Here we show that peritumorally injected, macroporous alginate gels loaded with granulocyte-macrophage colony-stimulating factor (GM-CSF) for concentrating dendritic cells (DCs), CpG oligonucleotides, and a doxorubicin-iRGD conjugate enhance the immunogenic death of tumor cells, increase systemic tumor-specific CD8 + T cells, repolarize tumor-associated macrophages towards an inflammatory M1-like phenotype, and significantly improve antitumor efficacy against poorly immunogenic TNBCs. This system also prevents tumor recurrence after surgical resection and results in 100% metastasis-free survival upon re-challenge. This chemo-immunotherapy that concentrates DCs to present endogenous tumor antigens generated in situ may broadly serve as a facile platform to modulate the suppressive TME, and enable in situ personalized cancer vaccination.


Subject(s)
Biocompatible Materials/therapeutic use , Cancer Vaccines/therapeutic use , Immunotherapy/methods , Triple Negative Breast Neoplasms/therapy , Animals , Antigens, Neoplasm/metabolism , Biotechnology/methods , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Drug Delivery Systems/methods , Female , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Humans , Immunologic Factors/metabolism , Immunologic Factors/therapeutic use , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/prevention & control , Neoplasms/immunology , Neoplasms/therapy , Triple Negative Breast Neoplasms/immunology , Tumor Microenvironment/immunology
3.
J Periodontol ; 91(11): 1475-1485, 2020 11.
Article in English | MEDLINE | ID: mdl-32150760

ABSTRACT

BACKGROUND: Periodontal disease results from the pathogenic interactions between the tissue, immune system, and microbiota; however, standard therapy fails to address the cellular mechanism underlying the chronic inflammation. Dendritic cells (DC) are key regulators of T cell fate, and biomaterials that recruit and program DC locally can direct T cell effector responses. We hypothesized that a biomaterial that recruited and programmed DC toward a tolerogenic phenotype could enrich regulatory T cells within periodontal tissue, with the eventual goal of attenuating T cell mediated pathology. METHODS: The interaction of previously identified factors that could induce tolerance, granulocyte-macrophage colony stimulating factor (GM-CSF) and thymic stromal lymphopoietin (TSLP), with the periodontitis network was confirmed in silico. The effect of the cytokines on DC migration was explored in vitro using time-lapse imaging. Finally, regulatory T cell enrichment in the dermis and periodontal tissue in response to alginate hydrogels delivering TSLP and GM-CSF was examinedin vivo in mice using immunohistochemistry and live-animal imaging. RESULTS: The GM-CSF and TSLP interactome connects to the periodontitis network. GM-CSF enhances DC migration in vitro. An intradermal injection of an alginate hydrogel releasing GM-CSF enhanced DC numbers and the addition of TSLP enriched FOXP3+ regulatory T cells locally. Injection of a hydrogel with GM-CSF and TSLP into the periodontal tissue in mice increased DC and FOXP3+ cell numbers in the tissue, FOXP3+ cells in the lymph node, and IL-10 in the tissue. CONCLUSION: Local biomaterial-mediated delivery of GM-CSF and TSLP can enrich DC and FOXP3+ cells and holds promise for treating the pathologic inflammation of periodontal disease.


Subject(s)
Dendritic Cells , T-Lymphocytes, Regulatory , Animals , Cell Differentiation , Cytokines , Granulocyte-Macrophage Colony-Stimulating Factor , Mice
4.
Adv Healthc Mater ; 6(6)2017 Mar.
Article in English | MEDLINE | ID: mdl-28116870

ABSTRACT

Biomaterial scaffolds that enrich and modulate immune cells in situ can form the basis for potent immunotherapies to elicit immunity or reëstablish tolerance. Here, the authors explore the potential of an injectable, porous hydrogel to induce a regulatory T cell (Treg) response by delivering a peptide antigen to dendritic cells in a noninflammatory context. Two methods are described for delivering the BDC peptide from pore-forming alginate gels in the nonobese diabetic mouse model of type 1 diabetes: encapsulation in poly(lactide-co-glycolide) (PLG) microparticles, or direct conjugation to the alginate polymer. While particle-based delivery leads to antigen-specific T cells responses in vivo, PLG particles alter the phenotype of the cells infiltrating the gels. Following gel-based peptide delivery, transient expansion of endogenous antigen-specific T cells is observed in the draining lymph nodes. Antigen-specific T cells accumulate in the gels, and, strikingly, ≈60% of the antigen-specific CD4+ T cells in the gels are Tregs. Antigen-specific T cells are also enriched in the pancreatic islets, and administration of peptide-loaded gels does not accelerate diabetes. This work demonstrates that a noninflammatory biomaterial system can generate antigen-specific Tregs in vivo, which may enable the development of new therapies for the treatment of transplant rejection or autoimmune diseases.


Subject(s)
Antigens , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 1/therapy , Hydrogels , Immune Tolerance/drug effects , Lactic Acid , Polyglycolic Acid , T-Lymphocytes, Regulatory/immunology , Animals , Antigens/chemistry , Antigens/pharmacology , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Hydrogels/chemistry , Hydrogels/pharmacology , Lactic Acid/chemistry , Lactic Acid/pharmacology , Mice , Mice, Inbred NOD , Polyglycolic Acid/chemistry , Polyglycolic Acid/pharmacology , Polylactic Acid-Polyglycolic Acid Copolymer , T-Lymphocytes, Regulatory/pathology
5.
Adv Healthc Mater ; 4(17): 2677-87, 2015 Dec 09.
Article in English | MEDLINE | ID: mdl-26474318

ABSTRACT

Biomaterials-based vaccines have emerged as a powerful method to evoke potent immune responses directly in vivo, without the need for ex vivo cell manipulation, and modulating dendritic cell (DC) responses in a noninflammatory context could enable the development of tolerogenic vaccines to treat autoimmunity. This study describes the development of a noninflammatory, injectable hydrogel system to locally enrich DCs in vivo without inducing their maturation or activation, as a first step toward this goal. Alginate hydrogels that form pores in situ are characterized and used as a physical scaffold for cell infiltration. These gels are also adapted to control the release of granulocyte-macrophage colony stimulating factor (GM-CSF), a potent inducer of DC recruitment and proliferation. In vivo, sustained release of GM-CSF from the pore-forming gels leads to the accumulation of millions of cells in the material. These cells are highly enriched in CD11b(+) CD11c(+) DCs, and further analysis of cell surface marker expression indicates these DCs are immature. This study demonstrates that a polymeric delivery system can mediate the accumulation of a high number and percentage of immature DCs, and may provide the basis for further development of materials-based, therapeutic vaccines.


Subject(s)
Dendritic Cells/drug effects , Hydrogels/administration & dosage , Animals , Biomarkers/metabolism , Cell Proliferation/drug effects , Dendritic Cells/metabolism , Female , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Injections/methods , Mice , Mice, Inbred C57BL , Vaccines/administration & dosage
6.
Nat Commun ; 6: 7556, 2015 Aug 12.
Article in English | MEDLINE | ID: mdl-26265369

ABSTRACT

A biomaterial-based vaccination system that uses minimal extracorporeal manipulation could provide in situ enhancement of dendritic cell (DC) numbers, a physical space where DCs interface with transplanted tumour cells, and an immunogenic context. Here we encapsulate GM-CSF, serving as a DC enhancement factor, and CpG ODN, serving as a DC activating factor, into sponge-like macroporous cryogels. These cryogels are injected subcutaneously into mice to localize transplanted tumour cells and deliver immunomodulatory factors in a controlled spatio-temporal manner. These vaccines elicit local infiltrates composed of conventional and plasmacytoid DCs, with the subsequent induction of potent, durable and specific anti-tumour T-cell responses in a melanoma model. These cryogels can be delivered in a minimally invasive manner, bypass the need for genetic modification of transplanted cancer cells and provide sustained release of immunomodulators. Altogether, these findings indicate the potential for cryogels to serve as a platform for cancer cell vaccinations.


Subject(s)
Cancer Vaccines/immunology , Cryogels/chemistry , Melanoma/prevention & control , Neoplasms, Experimental/prevention & control , Animals , Dendritic Cells , Female , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes
7.
Nat Biotechnol ; 33(1): 64-72, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25485616

ABSTRACT

Implanting materials in the body to program host immune cells is a promising alternative to transplantation of cells manipulated ex vivo to direct an immune response, but doing so requires a surgical procedure. Here we demonstrate that high-aspect-ratio, mesoporous silica rods (MSRs) injected with a needle spontaneously assemble in vivo to form macroporous structures that provide a 3D cellular microenvironment for host immune cells. In mice, substantial numbers of dendritic cells are recruited to the pores between the scaffold rods. The recruitment of dendritic cells and their subsequent homing to lymph nodes can be modulated by sustained release of inflammatory signals and adjuvants from the scaffold. Moreover, injection of an MSR-based vaccine formulation enhances systemic helper T cells TH1 and TH2 serum antibody and cytotoxic T-cell levels compared to bolus controls. These findings suggest that injectable MSRs may serve as a multifunctional vaccine platform to modulate host immune cell function and provoke adaptive immune responses.


Subject(s)
Immune System/cytology , Tissue Scaffolds , Vaccines/immunology , Animals , Mice
8.
Nat Mater ; 13(10): 970-8, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24930031

ABSTRACT

In vitro models of normal mammary epithelium have correlated increased extracellular matrix (ECM) stiffness with malignant phenotypes. However, the role of increased stiffness in this transformation remains unclear because of difficulties in controlling ECM stiffness, composition and architecture independently. Here we demonstrate that interpenetrating networks of reconstituted basement membrane matrix and alginate can be used to modulate ECM stiffness independently of composition and architecture. We find that, in normal mammary epithelial cells, increasing ECM stiffness alone induces malignant phenotypes but that the effect is completely abrogated when accompanied by an increase in basement-membrane ligands. We also find that the combination of stiffness and composition is sensed through ß4 integrin, Rac1, and the PI3K pathway, and suggest a mechanism in which an increase in ECM stiffness, without an increase in basement membrane ligands, prevents normal α6ß4 integrin clustering into hemidesmosomes.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Extracellular Matrix/physiology , Mammary Glands, Human/pathology , Mammary Glands, Human/physiopathology , Alginates/metabolism , Basement Membrane/physiology , Biocompatible Materials , Biophysical Phenomena , Cell Line , Epithelium/pathology , Epithelium/physiopathology , Female , Glucuronic Acid/metabolism , Hemidesmosomes/physiology , Hexuronic Acids/metabolism , Humans , Integrin alpha6beta4/metabolism , Ligands , Mechanotransduction, Cellular/physiology , Models, Biological , Phenotype , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , rac1 GTP-Binding Protein/metabolism
9.
Proc Natl Acad Sci U S A ; 109(48): 19590-5, 2012 Nov 27.
Article in English | MEDLINE | ID: mdl-23150549

ABSTRACT

Injectable biomaterials are increasingly being explored to minimize risks and complications associated with surgical implantation. We describe a strategy for delivery via conventional needle-syringe injection of large preformed macroporous scaffolds with well-defined properties. Injectable 3D scaffolds, in the form of elastic sponge-like matrices, were prepared by environmentally friendly cryotropic gelation of a naturally sourced polymer. Cryogels with shape-memory properties may be molded to a variety of shapes and sizes, and may be optionally loaded with therapeutic agents or cells. These scaffolds have the capability to withstand reversible deformations at over 90% strain level, and a rapid volumetric recovery allows the structurally defined scaffolds to be injected through a small-bore needle with nearly complete geometric restoration once delivered. These gels demonstrated long-term release of biomolecules in vivo. Furthermore, cryogels impregnated with bioluminescent reporter cells provided enhanced survival, higher local retention, and extended engraftment of transplanted cells at the injection site compared with a standard injection technique. These injectable scaffolds show great promise for various biomedical applications, including cell therapies.


Subject(s)
Biocompatible Materials , Tissue Scaffolds , Animals , Cryogels , Female , Hydrogels , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...