Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 14(1): 3546, 2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-37322053

RESUMO

Although adoptive T cell therapy provides the T cell pool needed for immediate tumor debulking, the infused T cells generally have a narrow repertoire for antigen recognition and limited ability for long-term protection. Here, we present a hydrogel that locally delivers adoptively transferred T cells to the tumor site while recruiting and activating host antigen-presenting cells with GMCSF or FLT3L and CpG, respectively. T cells alone loaded into these localized cell depots provided significantly better control of subcutaneous B16-F10 tumors than T cells delivered through direct peritumoral injection or intravenous infusion. T cell delivery combined with biomaterial-driven accumulation and activation of host immune cells prolonged the activation of the delivered T cells, minimized host T cell exhaustion, and enabled long-term tumor control. These findings highlight how this integrated approach provide both immediate tumor debulking and long-term protection against solid tumors, including against tumor antigen escape.


Assuntos
Criogéis , Neoplasias , Humanos , Neoplasias/patologia , Imunoterapia Adotiva , Linfócitos T , Células Apresentadoras de Antígenos
2.
Nat Biomed Eng ; 7(11): 1374-1391, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37365267

RESUMO

The efficacy of adoptive T-cell therapies largely depends on the generation of T-cell populations that provide rapid effector function and long-term protective immunity. Yet it is becoming clearer that the phenotypes and functions of T cells are inherently linked to their localization in tissues. Here we show that functionally distinct T-cell populations can be generated from T cells that received the same stimulation by altering the viscoelasticity of their surrounding extracellular matrix (ECM). By using a model ECM based on a norbornene-modified collagen type I whose viscoelasticity can be adjusted independently from its bulk stiffness by varying the degree of covalent crosslinking via a bioorthogonal click reaction with tetrazine moieties, we show that ECM viscoelasticity regulates T-cell phenotype and function via the activator-protein-1 signalling pathway, a critical regulator of T-cell activation and fate. Our observations are consistent with the tissue-dependent gene-expression profiles of T cells isolated from mechanically distinct tissues from patients with cancer or fibrosis, and suggest that matrix viscoelasticity could be leveraged when generating T-cell products for therapeutic applications.


Assuntos
Matriz Extracelular , Linfócitos T , Humanos , Matriz Extracelular/metabolismo , Colágeno Tipo I/metabolismo , Fibrose , Transdução de Sinais
3.
Nat Commun ; 14(1): 506, 2023 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-36720856

RESUMO

Patient responses to autologous CD19 chimeric antigen receptor (CAR) T-cell therapies are limited by insufficient and inconsistent cellular functionality. Here, we show that controlling the precise level of stimulation during T-cell activation to accommodate individual differences in the donor cells will dictate the functional attributes of CAR-T cell products. The functionality of CAR-T cell products, consisting of a diverse set of blood samples derived from healthy donors, acute lymphoblastic leukemia (ALL), and chronic lymphocytic lymphoma (CLL) patient samples, representing a range of patient health status, is tested upon culturing on artificial antigen-presenting cell scaffolds to deliver T-cell stimulatory ligands (anti-CD3/anti-CD28) at highly defined densities. A clear relationship is observed between the dose of stimulation, the phenotype of the T-cell blood sample prior to T-cell activation, and the functionality of the resulting CAR-T cell products. We present a model, based on this dataset, that predicts the precise stimulation needed to manufacture a desired CAR-T cell product, given the input T-cell attributes in the initial blood sample. These findings demonstrate a simple approach to enhance CAR-T functionality by personalizing the level of stimulation during T-cell activation to enable flexible manufacturing of more consistent and potent CAR-T cells.


Assuntos
Imunoterapia Adotiva , Linfócitos T , Células Apresentadoras de Antígenos , Antígenos CD19 , Humanos , Receptores de Antígenos Quiméricos
4.
Proc Natl Acad Sci U S A ; 120(1): e2213222120, 2023 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-36577059

RESUMO

Adoptive T cell transfer (ACT) therapies suffer from a number of limitations (e.g., poor control of solid tumors), and while combining ACT with cytokine therapy can enhance effectiveness, this also results in significant side effects. Here, we describe a nanotechnology approach to improve the efficacy of ACT therapies by metabolically labeling T cells with unnatural sugar nanoparticles, allowing direct conjugation of antitumor cytokines onto the T cell surface during the manufacturing process. This allows local, concentrated activity of otherwise toxic cytokines. This approach increases T cell infiltration into solid tumors, activates the host immune system toward a Type 1 response, encourages antigen spreading, and improves control of aggressive solid tumors and achieves complete blood cancer regression with otherwise noncurative doses of CAR-T cells. Overall, this method provides an effective and easily integrated approach to the current ACT manufacturing process to increase efficacy in various settings.


Assuntos
Citocinas , Neoplasias , Humanos , Citocinas/metabolismo , Imunoterapia Adotiva/métodos , Receptores de Antígenos de Linfócitos T , Linfócitos T , Neoplasias/patologia , Terapia Baseada em Transplante de Células e Tecidos
5.
Cancer Immunol Immunother ; 71(12): 2957-2968, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35524791

RESUMO

Disruption of the tumor extracellular matrix (ECM) may alter immune cell infiltration into the tumor and antitumor T cell priming in the tumor-draining lymph nodes (tdLNs). Here, we explore how intratumoral enzyme treatment (ET) of B16 melanoma tumors with ECM-depleting enzyme hyaluronidase alters adaptive and innate immune populations, including T cells, DCs, and macrophages, in the tumors and tdLNs. ET increased CD103+ DC abundance in the tdLNs, as well as antigen presentation of a model tumor antigen ovalbumin (OVA), eliciting local OVA-specific CD8+ T cell responses. Delivered in combination with a distant cryogel-based cancer vaccine, ET increased the systemic antigen-specific CD8+ T cell response. By enhancing activity within the tdLN, ET may broadly support immunotherapies in generating tumor-specific immunity.


Assuntos
Vacinas Anticâncer , Melanoma Experimental , Animais , Humanos , Ovalbumina , Células Dendríticas , Hialuronoglucosaminidase , Criogéis , Antígenos de Neoplasias , Linfonodos , Matriz Extracelular
6.
Biomaterials ; 257: 120266, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32763614

RESUMO

Mesenchymal stromal cells (MSCs) hold great therapeutic potential, in part because of their immunomodulatory properties. However, these properties can be transient and depend on multiple factors. Here, we developed a multifunctional hydrogel system to synergistically enhance the immunomodulatory properties of MSCs, using a combination of sustained inflammatory licensing and three-dimensional (3D) encapsulation in hydrogels with tunable mechanical properties. The immunomodulatory extracellular matrix hydrogels (iECM) consist of an interpenetrating network of click functionalized-alginate and fibrillar collagen, in which interferon γ (IFN-γ) loaded heparin-coated beads are incorporated. The 3D microenvironment significantly enhanced the expression of a wide panel of pivotal immunomodulatory genes in bone marrow-derived primary human MSCs (hMSCs), compared to two-dimensional (2D) tissue culture. Moreover, the inclusion of IFN-γ loaded heparin-coated beads prolonged the expression of key regulatory genes upregulated upon licensing, including indoleamine 2,3-dioxygenase 1 (IDO1) and galectin-9 (GAL9). At a protein level, iECM hydrogels enhanced the secretion of the licensing responsive factor Gal-9 by hMSCs. Its presence in hydrogel conditioned media confirmed the correct release and diffusion of the factors secreted by hMSCs from the system. Furthermore, co-culture of iECM-encapsulated hMSCs and activated human T cells resulted in suppressed proliferation, demonstrating direct regulation on immune cells. These data highlight the potential of iECM hydrogels to enhance the immunomodulatory properties of hMSCs in cell therapies.


Assuntos
Células-Tronco Mesenquimais , Biomimética , Meios de Cultivo Condicionados , Humanos , Hidrogéis , Imunomodulação
7.
Nat Mater ; 19(11): 1244-1252, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32424368

RESUMO

Targeted immunomodulation of dendritic cells (DCs) in vivo will enable manipulation of T-cell priming and amplification of anticancer immune responses, but a general strategy has been lacking. Here we show that DCs concentrated by a biomaterial can be metabolically labelled with azido groups in situ, which allows for their subsequent tracking and targeted modulation over time. Azido-labelled DCs were detected in lymph nodes for weeks, and could covalently capture dibenzocyclooctyne (DBCO)-bearing antigens and adjuvants via efficient Click chemistry for improved antigen-specific CD8+ T-cell responses and antitumour efficacy. We also show that azido labelling of DCs allowed for in vitro and in vivo conjugation of DBCO-modified cytokines, including DBCO-IL-15/IL-15Rα, to improve priming of antigen-specific CD8+ T cells. This DC labelling and targeted modulation technology provides an unprecedented strategy for manipulating DCs and regulating DC-T-cell interactions in vivo.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Imunomodulação , Azidas/química , Azidas/metabolismo , Vacinas Anticâncer/imunologia , Linhagem Celular Tumoral , Química Click , Células Dendríticas/citologia , Humanos , Imunoterapia , Coloração e Rotulagem
8.
Nat Protoc ; 15(3): 773-798, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31932772

RESUMO

Synthetic antigen-presenting cells (APCs) are used to mediate scalable ex vivo T-cell expansion for adoptive cell therapy. Recently, we developed APC-mimetic scaffolds (APC-ms), which present signals to T cells in a physiological manner to mediate rapid and controlled T-cell expansion. APC-ms are composed of individual high-aspect-ratio silica microrods loaded with soluble mitogenic cues and coated with liposomes of defined compositions, to form supported lipid bilayers. Membrane-bound ligands for stimulation and co-stimulation of T-cell receptors are presented via the fluid, synthetic membranes, while mitogenic cues are released slowly from the microrods. In culture, interacting T cells assemble the individual APC-ms microrods into a biodegradable 3D matrix. Compared to conventional methods, APC-ms facilitates several-fold greater polyclonal T-cell expansion and improved antigen-specific enrichment of rare T-cell subpopulations. Here we provide a detailed protocol for APC-ms synthesis and use for human T-cell activation, and discuss important considerations for material design and T-cell co-culture. This protocol describes the facile assembly of APC-ms in ~4 h and rapid expansion or enrichment of relevant T-cell clones in <2 weeks, and is applicable for T-cell manufacturing and assay development.


Assuntos
Células Apresentadoras de Antígenos , Linfócitos T , Linhagem da Célula , Clonagem Molecular , Regulação da Expressão Gênica , Humanos
9.
Nat Biotechnol ; 36(2): 160-169, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29334370

RESUMO

Therapeutic ex vivo T-cell expansion is limited by low rates and T-cell products of limited functionality. Here we describe a system that mimics natural antigen-presenting cells (APCs) and consists of a fluid lipid bilayer supported by mesoporous silica micro-rods. The lipid bilayer presents membrane-bound cues for T-cell receptor stimulation and costimulation, while the micro-rods enable sustained release of soluble paracrine cues. Using anti-CD3, anti-CD28, and interleukin-2, we show that the APC-mimetic scaffolds (APC-ms) promote two- to tenfold greater polyclonal expansion of primary mouse and human T cells compared with commercial expansion beads (Dynabeads). The efficiency of expansion depends on the density of stimulatory cues and the amount of material in the starting culture. Following a single stimulation, APC-ms enables antigen-specific expansion of rare cytotoxic T-cell subpopulations at a greater magnitude than autologous monocyte-derived dendritic cells after 2 weeks. APC-ms support over fivefold greater expansion of restimulated CD19 CAR-T cells than Dynabeads, with similar efficacy in a xenograft lymphoma model.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Imunoterapia Adotiva , Linfócitos T Citotóxicos/imunologia , Linfócitos T/imunologia , Animais , Antígenos CD28/antagonistas & inibidores , Antígenos CD28/imunologia , Complexo CD3/antagonistas & inibidores , Complexo CD3/imunologia , Células Dendríticas/imunologia , Humanos , Interleucina-2/imunologia , Bicamadas Lipídicas/imunologia , Ativação Linfocitária/imunologia , Camundongos , Cultura Primária de Células , Receptores de Antígenos de Linfócitos T/imunologia , Dióxido de Silício/química , Alicerces Teciduais , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Acta Biomater ; 65: 36-43, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29128539

RESUMO

Sustained, localized protein delivery can enhance the safety and activity of protein drugs in diverse disease settings. While hydrogel systems are widely studied as vehicles for protein delivery, they often suffer from rapid release of encapsulated cargo, leading to a narrow duration of therapy, and protein cargo can be denatured by incompatibility with the hydrogel crosslinking chemistry. In this work, we describe injectable nanocomposite hydrogels that are capable of sustained, bioactive, release of a variety of encapsulated proteins. Injectable and porous cryogels were formed by bio-orthogonal crosslinking of alginate using tetrazine-norbornene coupling. To provide sustained release from these hydrogels, protein cargo was pre-adsorbed to charged Laponite nanoparticles that were incorporated within the walls of the cryogels. The presence of Laponite particles substantially hindered the release of a number of proteins that otherwise showed burst release from these hydrogels. By modifying the Laponite content within the hydrogels, the kinetics of protein release could be precisely tuned. This versatile strategy to control protein release simplifies the design of hydrogel drug delivery systems. STATEMENT OF SIGNIFICANCE: Here we present an injectable nanocomposite hydrogel for simple and versatile controlled release of therapeutic proteins. Protein release from hydrogels often requires first entrapping the protein in particles and embedding these particles within the hydrogel to allow controlled protein release. This pre-encapsulation process can be cumbersome, can damage the protein's activity, and must be optimized for each protein of interest. The strategy presented in this work simply premixes the protein with charged nanoparticles that bind strongly with the protein. These protein-laden particles are then placed within a hydrogel and slowly release the protein into the surrounding environment. Using this method, tunable release from an injectable hydrogel can be achieved for a variety of proteins. This strategy greatly simplifies the design of hydrogel systems for therapeutic protein release applications.


Assuntos
Criogéis , Sistemas de Liberação de Medicamentos , Nanocompostos , Proteínas/administração & dosagem , Adsorção , Humanos , Cinética
11.
Biomaterials ; 131: 27-35, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28371625

RESUMO

Injectable hydrogels are suitable for local cell delivery to the subcutaneous space, but the lack of vasculature remains a limiting factor. Previously we demonstrated that biomaterials containing methacrylic acid promoted vascularization. Here we report the preparation of a semi-interpenetrating polymer network (SIPN), and its evaluation as an injectable carrier to deliver cells and generate blood vessels in a subcutaneous implantation site. The SIPN was prepared by reacting a blend of vinyl sulfone-terminated polyethylene glycol (PEG-VS) and sodium polymethacrylate (PMAA-Na) with dithiothreitol. The swelling of SIPN was sensitive to the PMAA-Na content but only small differences in gelation time, permeability and stiffness were noted. SIPN containing 20 mol% PMAA-Na generated a vascular network in the surrounding tissues, with 2-3 times as many vessels as was obtained with 10 mol% PMAA-Na or PEG alone. Perfusion studies showed that the generated vessels were perfused and connected to the host vasculature as early as seven days after transplantation. Islets embedded in SIPN were viable and responsive to glucose stimulation in vitro. In a proof of concept study in a streptozotocin-induced diabetic mouse model, a progressive return to normoglycemia was observed and the presence of insulin positive islets was confirmed when islets were embedded in SIPN prior to delivery. Our approach proposes a biomaterial-mediated strategy to deliver cells while enhancing vascularization.


Assuntos
Células Imobilizadas/citologia , Hidrogéis/química , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas/citologia , Polietilenoglicóis/química , Ácidos Polimetacrílicos/química , Alicerces Teciduais/química , Animais , Materiais Biocompatíveis/administração & dosagem , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Contagem de Células , Células Cultivadas , Diabetes Mellitus Experimental/terapia , Ditiotreitol/administração & dosagem , Ditiotreitol/análogos & derivados , Ditiotreitol/farmacologia , Hidrogéis/administração & dosagem , Hidrogéis/farmacologia , Injeções , Transplante das Ilhotas Pancreáticas/métodos , Masculino , Camundongos , Camundongos SCID , Neovascularização Fisiológica/efeitos dos fármacos , Polietilenoglicóis/administração & dosagem , Polietilenoglicóis/farmacologia , Ácidos Polimetacrílicos/administração & dosagem , Ácidos Polimetacrílicos/farmacologia , Ratos
12.
Biomaterials ; 98: 203-14, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27264502

RESUMO

Poly(methacrylic acid-co-methyl methacrylate) (MAA) beads promote a vascular regenerative response when used in diabetic wound healing. Previous studies reported that MAA beads modulated the expression of sonic hedgehog (Shh) and inflammation related genes in diabetic wounds. The aim of this work was to follow up on these observations in a subcutaneous injection model to study the host response in the absence of the confounding factors of diabetic wound healing. In this model, MAA beads improved vascularization in healthy mice of both sexes compared to control poly(methyl methacrylate) (MM) beads, with a stronger effect seen in males than females. MAA-induced vessels were perfusable, as evidenced from the CLARITY-processed images. In Shh-Cre-eGFP/Ptch1-LacZ non-diabetic transgenic mice, the increased vessel formation was accompanied by a higher density of cells expressing GFP (Shh) and ß-Gal (patched 1, Ptch1) suggesting MAA enhanced the activation of the Shh pathway. Ptch1 is the Shh receptor and a target of the pathway. MAA beads also modulated the inflammatory cell infiltrate in CD1 mice: more neutrophils and more macrophages were noted with MAA relative to MM beads at days 1 and 7, respectively. In addition, MAA beads biased macrophages towards a MHCII-CD206+ ("M2") polarization state. This study suggests that the Shh pathway and an altered inflammatory response are two elements of the complex mechanism whereby MAA-based biomaterials effect vascular regeneration.


Assuntos
Polaridade Celular/efeitos dos fármacos , Proteínas Hedgehog/metabolismo , Macrófagos/citologia , Metacrilatos/farmacologia , Microesferas , Transdução de Sinais , Animais , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/crescimento & desenvolvimento , Contagem de Células , Feminino , Proteínas de Fluorescência Verde/metabolismo , Inflamação/patologia , Injeções Subcutâneas , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos , Modelos Animais , Transdução de Sinais/efeitos dos fármacos , beta-Galactosidase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...