Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Semin Immunol ; 56: 101537, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34844835

RESUMEN

Currently, medicine lacks the ability to reprogram selected immune cells so they possess all the functions which, from a clinical standpoint, physicians might wish them to have. To solve this problem, scientists have been marrying concepts from materials science, immunology, and genetic engineering to develop novel nanotherapeutics that directly genetically reprogram immune cells inside the body. These products could address key limitations of existing ex vivo-engineered cell immunotherapies and substantially enhance patient access and outcomes. This review highlights the latest advances in this rapidly emerging biotech field and discusses challenges in translating these preclinical studies into successful clinical nanomedicines.


Asunto(s)
Reprogramación Celular , Nanomedicina , Ingeniería Genética , Humanos , Inmunoterapia , Poder Psicológico
3.
Annu Rev Biomed Eng ; 23: 385-405, 2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-33863239

RESUMEN

Gene therapy makes it possible to engineer chimeric antigen receptors (CARs) to create T cells that target specific diseases. However, current approaches require elaborate and expensive protocols to manufacture engineered T cells ex vivo, putting this therapy beyond the reach of many patients who might benefit. A solution could be to program T cells in vivo. Here, we evaluate the clinical need for in situ CAR T cell programming, compare competing technologies, review current progress, and provide a perspective on the long-term impact of this emerging and rapidly flourishing biotechnology field.


Asunto(s)
Técnicas de Reprogramación Celular , Inmunoterapia Adoptiva , Receptores Quiméricos de Antígenos , Linfocitos T , Terapia Genética , Humanos , Receptores Quiméricos de Antígenos/genética
4.
Nat Biomed Eng ; 4(2): 195-206, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31819155

RESUMEN

Micropatterned nickel titanium (commonly known as nitinol) thin films with complex designs, high structural resolution and excellent biocompatibility can be cheaply fabricated using magnetron sputtering. Here, we show that these benefits can be leveraged to fabricate micromesh implants that are loaded with tumour-specific human chimeric antigen receptor (CAR)-T cells for the treatment of solid tumours. In a mouse model of non-resectable ovarian cancer, the cell-loaded nitinol thin films spatially conformed to the implantation site, fostered the rapid expansion of T cells, delivered a high density of T cells directly to the tumour and significantly improved animal survival. We also show that self-expandable stents that were coated with T-cell-loaded films and implanted into subcutaneous tumours in mice improved the duration of stent patency by delaying tumour ingrowth. By providing direct access to tumours, CAR-T-cell-loaded micropatterned nitinol thin films can improve the effects of cell-based therapies.


Asunto(s)
Aleaciones , Inmunoterapia Adoptiva/instrumentación , Inmunoterapia Adoptiva/métodos , Bombas de Infusión Implantables , Neoplasias Ováricas/terapia , Receptores Quiméricos de Antígenos/uso terapéutico , Animales , Línea Celular Tumoral , Movimiento Celular , Femenino , Humanos , Ratones , Neoplasias Ováricas/inmunología , Linfocitos T/fisiología
5.
Cancer Res ; 78(13): 3718-3730, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29760047

RESUMEN

A major obstacle to the success rate of chimeric antigen receptor (CAR-) T-cell therapy against solid tumors is the microenvironment antagonistic to T cells that solid tumors create. Conventional checkpoint blockade can silence lymphocyte antisurvival pathways activated by tumors, but because they are systemic, these treatments disrupt immune homeostasis and induce autoimmune side effects. Thus, new technologies are required to remodel the tumor milieu without causing systemic toxicities. Here, we demonstrate that targeted nanocarriers that deliver a combination of immune-modulatory agents can remove protumor cell populations and simultaneously stimulate antitumor effector cells. We administered repeated infusions of lipid nanoparticles coated with the tumor-targeting peptide iRGD and loaded with a combination of a PI3K inhibitor to inhibit immune-suppressive tumor cells and an α-GalCer agonist of therapeutic T cells to synergistically sway the tumor microenvironment of solid tumors from suppressive to stimulatory. This treatment created a therapeutic window of 2 weeks, enabling tumor-specific CAR-T cells to home to the lesion, undergo robust expansion, and trigger tumor regression. CAR-T cells administered outside this therapeutic window had no curative effect. The lipid nanoparticles we used are easy to manufacture in substantial amounts, and we demonstrate that repeated infusions of them are safe. Our technology may therefore provide a practical and low-cost strategy to potentiate many cancer immunotherapies used to treat solid tumors, including T-cell therapy, vaccines, and BITE platforms.Significance: A new nanotechnology approach can promote T-cell therapy for solid tumors. Cancer Res; 78(13); 3718-30. ©2018 AACR.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Inmunoterapia Adoptiva/métodos , Neoplasias/terapia , Receptores Quiméricos de Antígenos/inmunología , Microambiente Tumoral/efectos de los fármacos , Animales , Línea Celular Tumoral , Terapia Combinada/métodos , Modelos Animales de Enfermedad , Composición de Medicamentos/métodos , Femenino , Galactosilceramidas/agonistas , Humanos , Liposomas , Ratones , Ratones Endogámicos BALB C , Nanopartículas/química , Neoplasias/inmunología , Neoplasias/patología , Oligopéptidos/química , Inhibidores de las Quinasa Fosfoinosítidos-3 , Resultado del Tratamiento , Microambiente Tumoral/inmunología
6.
J Clin Invest ; 127(6): 2176-2191, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28436934

RESUMEN

Therapies using T cells that are programmed to express chimeric antigen receptors (CAR T cells) consistently produce positive results in patients with hematologic malignancies. However, CAR T cell treatments are less effective in solid tumors for several reasons. First, lymphocytes do not efficiently target CAR T cells; second, solid tumors create an immunosuppressive microenvironment that inactivates T cell responses; and third, solid cancers are typified by phenotypic diversity and thus include cells that do not express proteins targeted by the engineered receptors, enabling the formation of escape variants that elude CAR T cell targeting. Here, we have tested implantable biopolymer devices that deliver CAR T cells directly to the surfaces of solid tumors, thereby exposing them to high concentrations of immune cells for a substantial time period. In immunocompetent orthotopic mouse models of pancreatic cancer and melanoma, we found that CAR T cells can migrate from biopolymer scaffolds and eradicate tumors more effectively than does systemic delivery of the same cells. We have also demonstrated that codelivery of stimulator of IFN genes (STING) agonists stimulates immune responses to eliminate tumor cells that are not recognized by the adoptively transferred lymphocytes. Thus, these devices may improve the effectiveness of CAR T cell therapy in solid tumors and help protect against the emergence of escape variants.


Asunto(s)
Biopolímeros/administración & dosificación , Carcinoma Ductal Pancreático/terapia , Melanoma Experimental/terapia , Neoplasias Pancreáticas/terapia , Traslado Adoptivo , Animales , Células Presentadoras de Antígenos/fisiología , Antineoplásicos/administración & dosificación , Carcinoma Ductal Pancreático/inmunología , Línea Celular Tumoral , GMP Cíclico/administración & dosificación , GMP Cíclico/análogos & derivados , Portadores de Fármacos/administración & dosificación , Femenino , Implantes Experimentales , Melanoma Experimental/inmunología , Proteínas de la Membrana/agonistas , Ratones Endogámicos C57BL , Ratones Transgénicos , Trasplante de Neoplasias , Neoplasias Pancreáticas/inmunología , Linfocitos T/fisiología
7.
Nat Nanotechnol ; 12(8): 813-820, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28416815

RESUMEN

An emerging approach for treating cancer involves programming patient-derived T cells with genes encoding disease-specific chimeric antigen receptors (CARs), so that they can combat tumour cells once they are reinfused. Although trials of this therapy have produced impressive results, the in vitro methods they require to generate large numbers of tumour-specific T cells are too elaborate for widespread application to treat cancer patients. Here, we describe a method to quickly program circulating T cells with tumour-recognizing capabilities, thus avoiding these complications. Specifically, we demonstrate that DNA-carrying nanoparticles can efficiently introduce leukaemia-targeting CAR genes into T-cell nuclei, thereby bringing about long-term disease remission. These polymer nanoparticles are easy to manufacture in a stable form, which simplifies storage and reduces cost. Our technology may therefore provide a practical, broadly applicable treatment that can generate anti-tumour immunity 'on demand' for oncologists in a variety of settings.


Asunto(s)
ADN/química , Portadores de Fármacos , Técnicas de Transferencia de Gen , Inmunidad Celular/efectos de los fármacos , Leucemia/terapia , Nanopartículas/química , Receptores Quiméricos de Antígenos , Linfocitos T/inmunología , Animales , Línea Celular Tumoral , Portadores de Fármacos/química , Portadores de Fármacos/farmacología , Inmunidad Celular/genética , Leucemia/genética , Leucemia/inmunología , Leucemia/patología , Ratones , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/inmunología
8.
Cancer Cell ; 27(4): 489-501, 2015 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-25873172

RESUMEN

Cancer immunotherapies under development have generally focused on either stimulating T cell immunity or driving antibody-directed effector functions of the innate immune system such as antibody-dependent cell-mediated cytotoxicity (ADCC). We find that a combination of an anti-tumor antigen antibody and an untargeted IL-2 fusion protein with delayed systemic clearance induces significant tumor control in aggressive isogenic tumor models via a concerted innate and adaptive response involving neutrophils, NK cells, macrophages, and CD8(+) T cells. This combination therapy induces an intratumoral "cytokine storm" and extensive lymphocyte infiltration. Adoptive transfer of anti-tumor T cells together with this combination therapy leads to robust cures of established tumors and development of immunological memory.


Asunto(s)
Neoplasias/terapia , Inmunidad Adaptativa , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Sinergismo Farmacológico , Semivida , Inmunidad Innata , Inmunoterapia , Interleucina-2/metabolismo , Interleucina-2/farmacocinética , Interleucina-2/farmacología , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Ratones , Ratones Endogámicos C57BL , Neoplasias/inmunología
9.
Nat Biotechnol ; 33(1): 97-101, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25503382

RESUMEN

Although adoptive T-cell therapy holds promise for the treatment of many cancers, its clinical utility has been limited by problems in delivering targeted lymphocytes to tumor sites, and the cells' inefficient expansion in the immunosuppressive tumor microenvironment. Here we describe a bioactive polymer implant capable of delivering, expanding and dispersing tumor-reactive T cells. The approach can be used to treat inoperable or incompletely removed tumors by situating implants near them or at resection sites. Using a mouse breast cancer resection model, we show that the implants effectively support tumor-targeting T cells throughout resection beds and associated lymph nodes, and reduce tumor relapse compared to conventional delivery modalities. In a multifocal ovarian cancer model, we demonstrate that polymer-delivered T cells trigger regression, whereas injected tumor-reactive lymphocytes have little curative effect. Scaffold-based T-cell delivery may provide a viable treatment option for inoperable tumors and reduce the rate of metastatic relapse after surgery.


Asunto(s)
Traslado Adoptivo , Biopolímeros , Prótesis e Implantes , Linfocitos T/inmunología
10.
J Control Release ; 172(2): 426-35, 2013 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-23770010

RESUMEN

In adoptive cell therapy (ACT), autologous tumor-specific T-cells isolated from cancer patients are activated and expanded ex vivo, then infused back into the individual to eliminate metastatic tumors. A major limitation of this promising approach is the rapid loss of ACT T-cell effector function in vivo due to the highly immunosuppressive environment in tumors. Protection of T-cells from immunosuppressive signals can be achieved by systemic administration of supporting adjuvant drugs such as interleukins, chemotherapy, and other immunomodulators, but these adjuvant treatments are often accompanied by serious toxicities and may still fail to optimally stimulate lymphocytes in all tumor and lymphoid compartments. Here we propose a novel strategy to repeatedly stimulate or track ACT T-cells, using cytokines or ACT-cell-specific antibodies as ligands to target PEGylated liposomes to transferred T-cells in vivo. Using F(ab')2 fragments against a unique cell surface antigen on ACT cells (Thy1.1) or an engineered interleukin-2 (IL-2) molecule on an Fc framework as targeting ligands, we demonstrate that >95% of ACT cells can be conjugated with liposomes following a single injection in vivo. Further, we show that IL-2-conjugated liposomes both target ACT cells and are capable of inducing repeated waves of ACT T-cell proliferation in tumor-bearing mice. These results demonstrate the feasibility of repeated functional targeting of T-cells in vivo, which will enable delivery of imaging contrast agents, immunomodulators, or chemotherapy agents in adoptive cell therapy regimens.


Asunto(s)
Inmunoconjugados/administración & dosificación , Inmunoterapia Adoptiva/métodos , Interleucina-2/administración & dosificación , Liposomas/administración & dosificación , Melanoma/terapia , Linfocitos T/inmunología , Linfocitos T/trasplante , Animales , Proliferación Celular , Citocinas/administración & dosificación , Citocinas/química , Citocinas/inmunología , Femenino , Humanos , Inmunoconjugados/química , Inmunoconjugados/inmunología , Fragmentos Fab de Inmunoglobulinas/administración & dosificación , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/inmunología , Interleucina-2/química , Interleucina-2/inmunología , Liposomas/química , Liposomas/inmunología , Activación de Linfocitos , Melanoma/inmunología , Melanoma/patología , Melanoma/secundario , Ratones , Ratones Endogámicos C57BL
11.
Biomaterials ; 33(23): 5776-87, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22594972

RESUMEN

Regulating molecular interactions in the T-cell synapse to prevent autoimmunity or, conversely, to boost anti-tumor immunity has long been a goal in immunotherapy. However, delivering therapeutically meaningful doses of immune-modulating compounds into the synapse represents a major challenge. Here, we report that covalent coupling of maleimide-functionlized nanoparticles (NPs) to free thiol groups on T-cell membrane proteins enables efficient delivery of compounds into the T-cell synapse. We demonstrate that surface-linked NPs are rapidly polarized toward the nascent immunological synapse (IS) at the T-cell/APC contact zone during antigen recognition. To translate these findings into a therapeutic application we tested the NP delivery of NSC-87877, a dual inhibitor of Shp1 and Shp2, key phosphatases that downregulate T-cell receptor activation in the synapse, in the context of adoptive T cell therapy of cancer. Conjugating NSC-87877-loaded NPs to the surface of tumor-specific T cells just prior to adoptive transfer into mice with advanced prostate cancer promoted a much greater T-cell expansion at the tumor site, relative to co-infusing the same drug dose systemically, leading to enhanced survival of treated animals. In summary, our studies support the application of T-cell-linked synthetic NPs as efficient drug delivery vehicles into the IS, as well as the broad applicability of this new paradigm for therapeutically modulating signaling events at the T-cell/APC interface.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Factores Inmunológicos/administración & dosificación , Sinapsis Inmunológicas/metabolismo , Nanopartículas/química , Linfocitos T/citología , Animales , Células Cultivadas , Factores Inmunológicos/uso terapéutico , Inmunoterapia , Masculino , Maleimidas/química , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Próstata/patología , Neoplasias de la Próstata/terapia , Compuestos de Sulfhidrilo/química
12.
Nano Today ; 6(3): 309-325, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21826117

RESUMEN

Therapeutic treatments based on the injection of living cells are in clinical use and preclinical development for diseases ranging from cancer to cardiovascular disease to diabetes. To enhance the function of therapeutic cells, a variety of chemical and materials science strategies are being developed that engineer the surface of therapeutic cells with new molecules, artificial receptors, and multifunctional nanomaterials, synthetically endowing donor cells with new properties and functions. These approaches offer a powerful complement to traditional genetic engineering strategies for enhancing the function of living cells.

13.
Nat Mater ; 10(3): 243-51, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21336265

RESUMEN

Vaccines based on recombinant proteins avoid the toxicity and antivector immunity associated with live vaccine (for example, viral) vectors, but their immunogenicity is poor, particularly for CD8(+) T-cell responses. Synthetic particles carrying antigens and adjuvant molecules have been developed to enhance subunit vaccines, but in general these materials have failed to elicit CD8(+) T-cell responses comparable to those for live vectors in preclinical animal models. Here, we describe interbilayer-crosslinked multilamellar vesicles formed by crosslinking headgroups of adjacent lipid bilayers within multilamellar vesicles. Interbilayer-crosslinked vesicles stably entrapped protein antigens in the vesicle core and lipid-based immunostimulatory molecules in the vesicle walls under extracellular conditions, but exhibited rapid release in the presence of endolysosomal lipases. We found that these antigen/adjuvant-carrying vesicles form an extremely potent whole-protein vaccine, eliciting endogenous T-cell and antibody responses comparable to those for the strongest vaccine vectors. These materials should enable a range of subunit vaccines and provide new possibilities for therapeutic protein delivery.


Asunto(s)
Inmunidad Celular , Inmunidad Humoral , Liposomas/química , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología , Adyuvantes Inmunológicos/química , Animales , Portadores de Fármacos , Diseño de Fármacos , Memoria Inmunológica , Membrana Dobles de Lípidos/química , Liposomas/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/inmunología , Vacunas Sintéticas/química , Vacunas Virales/química , Vacunas Virales/inmunología
14.
Nat Med ; 16(9): 1035-41, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20711198

RESUMEN

A major limitation of cell therapies is the rapid decline in viability and function of the transplanted cells. Here we describe a strategy to enhance cell therapy via the conjugation of adjuvant drug-loaded nanoparticles to the surfaces of therapeutic cells. With this method of providing sustained pseudoautocrine stimulation to donor cells, we elicited marked enhancements in tumor elimination in a model of adoptive T cell therapy for cancer. We also increased the in vivo repopulation rate of hematopoietic stem cell grafts with very low doses of adjuvant drugs that were ineffective when given systemically. This approach is a simple and generalizable strategy to augment cytoreagents while minimizing the systemic side effects of adjuvant drugs. In addition, these results suggest therapeutic cells are promising vectors for actively targeted drug delivery.


Asunto(s)
Ingeniería Biomédica/métodos , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Nanopartículas/uso terapéutico , Linfocitos T/inmunología , Animales , Portadores de Fármacos/uso terapéutico , Luciérnagas/enzimología , Humanos , Cinética , Luciferasas/genética , Luminiscencia , Ratones , Neoplasias/inmunología , Neoplasias/terapia , Organismos Modificados Genéticamente/inmunología , Bazo/citología , Bazo/inmunología , Bazo/fisiología , Trasplante de Células Madre/métodos , Propiedades de Superficie
15.
Blood ; 116(11): e18-25, 2010 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-20511541

RESUMEN

We have developed a dual bioluminescent reporter system allowing noninvasive, concomitant imaging of T-cell trafficking, expansion, and activation of nuclear factor of activated T cells (NFAT) in vivo. NFAT activation plays an important role in T-cell activation and T-cell development. Therefore we used this system to determine spatial-temporal activation patterns of (1) proliferating T lymphocytes during graft-versus-host disease (GVHD) and (2) T-cell precursors during T-cell development after allogeneic hematopoietic stem cell transplantation (HSCT). In the first days after HSCT, donor T cells migrated to the peripheral lymph nodes and the intestines, whereas the NFAT activation was dominant in the intestines, suggesting an important role for the intestines in the early stages of alloactivation during development of GVHD. After adoptive transfer of in vitro-derived T-cell receptor (TCR) H-Y transgenic T-cell precursors into B6 (H-2(b)) hosts of both sexes, NFAT signaling and development into CD4(+) or CD8(+) single-positive cells could only be detected in the thymus of female recipients indicating either absence of positive selection or prompt depletion of double-positive thymocytes in the male recipients. Because NFAT plays an important role in a wide range of cell types, our system could provide new insights into a variety of biologic processes.


Asunto(s)
Movimiento Celular , Proliferación Celular , Células Precursoras de Linfocitos T/citología , Linfocitos T/citología , Células 3T3 , Traslado Adoptivo , Animales , Línea Celular , Línea Celular Tumoral , Femenino , Enfermedad Injerto contra Huésped/etiología , Enfermedad Injerto contra Huésped/metabolismo , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Células Jurkat , Lentivirus/genética , Luciferasas/genética , Luciferasas/metabolismo , Luminiscencia , Mediciones Luminiscentes/métodos , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Factores de Transcripción NFATC/genética , Células Precursoras de Linfocitos T/metabolismo , Células Precursoras de Linfocitos T/trasplante , Regiones Promotoras Genéticas/genética , Linfocitos T/metabolismo
16.
J Immunol ; 183(1): 631-41, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19542472

RESUMEN

Aspergillus fumigatus, a common environmental fungus, can cause lethal invasive infections in immunocompromised hosts. In immunocompetent individuals, however, inhaled A. fumigatus spores prime CD4(+) T cells and activate immune responses that prevent invasive infection. Calibration of inflammatory responses to levels that prevent fungal invasion without inducing collateral tissue damage is essential for host survival, but the underlying regulatory mechanisms remain undefined. Although IL-10 is a validated regulatory cytokine that suppresses immune responses, and IL-10 deficiency or blockade generally enhances immune responses, we find that A. fumigatus-specific T cell frequencies are markedly reduced in airways of IL-10-deficient mice. T cell priming, proliferation, and survival were unaffected by IL-10 deficiency and did not account for decreased frequencies of A. fumigatus-specific T cells in the airways of IL-10-deficient mice. Instead, IL-10 deficiency results in redistribution of A. fumigatus-specific T cells from infected lungs to the gut, a process that is reversed by antibiotic-mediated depletion of intestinal microbes. Our studies demonstrate that disregulated immune responses in the gut can result in dramatic redistribution of pathogen-specific T cells within the host.


Asunto(s)
Aspergilosis/inmunología , Aspergilosis/patología , Aspergillus fumigatus/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/microbiología , Epítopos de Linfocito T/inmunología , Interleucina-10/deficiencia , Animales , Aspergilosis/genética , Linfocitos T CD4-Positivos/patología , Movimiento Celular/inmunología , Células Cultivadas , Interleucina-10/genética , Intestinos/inmunología , Intestinos/microbiología , Intestinos/patología , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/microbiología , Ganglios Linfáticos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos
17.
Mol Ther ; 16(10): 1745-52, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18682698

RESUMEN

We have developed a lentiviral vector system for human factor IX (hFIX) gene transfer in hematopoietic stem cells (HSCs) that provides erythroid cell-derived systemic protein delivery following nonmyeloablative conditioning and in vivo methylguanine methyltransferase (MGMT) drug selection. After bone marrow transplantation under moderate Busulfan conditioning, the initial hFIX expression in the chimeras was minimally detectable. However, the hFIX levels rose sharply following in vivo MGMT-drug selection and eventually reached a level that is considered curative in hemophilia B therapy (>500 ng/ml). The rise of hFIX levels was proportional to the increase in vector copy (VC) number in peripheral blood cells. High levels of hFIX expression were maintained in serially engrafted mice chimeras for 18 months. Importantly, high-level hFIX expression by erythroid cells did not result in anemia or adversely affect red blood cell counts. The prospect of combining reduced intensity conditioning, a presumably lowered risk of insertional mutagenesis due to low VC number requirement and erythroid-restricted transgene expression, as well as long-term protein expression at high level, strongly supports the potential applicability of adult stem cell-based gene therapy in nonlethal blood or metabolic disorders, as demonstrated here for hemophilia.


Asunto(s)
Factor IX/administración & dosificación , Células Madre Hematopoyéticas/metabolismo , Hemofilia B/terapia , Animales , Metilasas de Modificación del ADN/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Factor IX/genética , Factor IX/farmacología , Vectores Genéticos , Células Madre Hematopoyéticas/citología , Homeostasis/efectos de los fármacos , Humanos , Lentivirus/genética , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Proteínas Supresoras de Tumor/metabolismo
18.
Nat Med ; 13(12): 1440-9, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18026115

RESUMEN

To reject tumors, T cells must overcome poor tumor immunogenicity and an adverse tumor microenvironment. Providing agonistic costimulatory signals to tumor-infiltrating T cells to augment T cell function remains a challenge for the implementation of safe and effective immunotherapy. We hypothesized that T cells overexpressing selected costimulatory ligands could serve as cellular vehicles mediating powerful, yet constrained, anatomically targeted costimulation. Here, we show that primary human T cells expressing CD80 and 4-1BB ligand (4-1BBL) vigorously respond to tumor cells lacking costimulatory ligands and provoke potent rejection of large, systemic tumors in immunodeficient mice. In addition to showing costimulation of bystander T cells (transcostimulation), we show the effect of CD80 and 4-1BBL binding to their respective receptors in the immunological synapse of isolated single cells (autocostimulation). This new strategy of endowing T cells with constitutively expressed costimulatory ligands could be extended to other ligand-receptor pairs and used to enhance any targeted adoptive transfer therapy.


Asunto(s)
Ligando 4-1BB/metabolismo , Antígeno B7-1/metabolismo , Neoplasias/inmunología , Traslado Adoptivo , Animales , Células Presentadoras de Antígenos , Línea Celular Tumoral , Humanos , Sistema Inmunológico , Ratones , Ratones SCID , Microscopía Confocal , Neoplasias/metabolismo , Unión Proteica , Retroviridae/metabolismo , Linfocitos T/metabolismo
19.
Nat Biotechnol ; 24(8): 1017-21, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16845377

RESUMEN

We demonstrate here the capacity of erythroid cells to mediate long-term, systemic and therapeutic protein delivery in vivo. By targeting human factor IX (hFIX) expression to late-stage erythropoiesis, we achieve long-term hFIX secretion at levels significantly higher (>tenfold) than those obtained with an archetypal ubiquitous promoter in a mouse model of hemophilia B. Erythroid cell-derived hFIX is biologically active, resulting in phenotypic correction of the bleeding disorder. In addition to achieving high expression levels and resistance to transcriptional silencing, red cell-mediated protein delivery offers multiple advantages including immune tolerance induction, reduction of the risk of insertional oncogenesis and relative ease of application by either engrafting transduced hematopoietic stem cells or transfusing ex vivo-generated, stem cell-derived erythroid cells.


Asunto(s)
Células Eritroides/metabolismo , Factor IX/biosíntesis , Células Madre Hematopoyéticas/metabolismo , Ingeniería de Proteínas/métodos , Proteínas Recombinantes/biosíntesis , Animales , Células Cultivadas , Células Eritroides/citología , Factor IX/genética , Mejoramiento Genético/métodos , Células Madre Hematopoyéticas/citología , Humanos , Ratones , Ratones Endogámicos C57BL
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...