Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
PLoS One ; 15(2): e0228112, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32040512

RESUMEN

Neoantigens can be predicted and in some cases identified using the data obtained from the whole exome sequencing and transcriptome sequencing of tumor cells. These sequencing data can be coupled with single-cell RNA sequencing for the direct interrogation of the transcriptome, surfaceome, and pairing of αß T-cell receptors (TCRαß) from hundreds of single T cells. Using these 2 large datasets, we established a platform for identifying antigens recognized by TCRαßs obtained from single T cells. Our approach is based on the rapid expression of cloned TCRαß genes as Sleeping Beauty transposons and the determination of the introduced TCRαßs' antigen specificity and avidity using a reporter cell line. The platform enables the very rapid identification of tumor-reactive TCRs for the bioengineering of T cells with redirected specificity.


Asunto(s)
Ingeniería Celular/métodos , Clonación Molecular/métodos , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T/citología , Linfocitos T/metabolismo , Expresión Génica , Biblioteca de Genes , Genes MHC Clase I/genética , Genes MHC Clase II/genética , Células HEK293 , Humanos , Cinética , Receptores de Antígenos de Linfocitos T alfa-beta/genética
2.
Oncogene ; 37(27): 3686-3697, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29622795

RESUMEN

The CD56 antigen (NCAM-1) is highly expressed on several malignancies with neuronal or neuroendocrine differentiation, including small-cell lung cancer and neuroblastoma, tumor types for which new therapeutic options are needed. We hypothesized that CD56-specific chimeric antigen receptor (CAR) T cells could target and eliminate CD56-positive malignancies. Sleeping Beauty transposon-generated CD56R-CAR T cells exhibited αßT-cell receptors, released antitumor cytokines upon co-culture with CD56+ tumor targets, demonstrated a lack of fratricide, and expression of cytolytic function in the presence of CD56+ stimulation. The CD56R-CAR+ T cells are capable of killing CD56+ neuroblastoma, glioma, and SCLC tumor cells in in vitro co-cultures and when tested against CD56+ human xenograft neuroblastoma models and SCLC models, CD56R-CAR+ T cells were able to inhibit tumor growth in vivo. These results indicate that CD56-CARs merit further investigation as a potential treatment for CD56+ malignancies.


Asunto(s)
Antígeno CD56/metabolismo , Glioma/terapia , Neoplasias Pulmonares/terapia , Neuroblastoma/terapia , Receptores Quiméricos de Antígenos/metabolismo , Carcinoma Pulmonar de Células Pequeñas/terapia , Linfocitos T/inmunología , Linfocitos T/trasplante , Animales , Antígeno CD56/genética , Línea Celular Tumoral , Glioma/patología , Humanos , Neoplasias Pulmonares/patología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos NOD , Ratones Desnudos , Neuroblastoma/patología , Carcinoma Pulmonar de Células Pequeñas/patología , Transposasas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Proc Natl Acad Sci U S A ; 113(48): E7788-E7797, 2016 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-27849617

RESUMEN

Adoptive immunotherapy retargeting T cells to CD19 via a chimeric antigen receptor (CAR) is an investigational treatment capable of inducing complete tumor regression of B-cell malignancies when there is sustained survival of infused cells. T-memory stem cells (TSCM) retain superior potential for long-lived persistence, but challenges exist in manufacturing this T-cell subset because they are rare among circulating lymphocytes. We report a clinically relevant approach to generating CAR+ T cells with preserved TSCM potential using the Sleeping Beauty platform. Because IL-15 is fundamental to T-cell memory, we incorporated its costimulatory properties by coexpressing CAR with a membrane-bound chimeric IL-15 (mbIL15). The mbIL15-CAR T cells signaled through signal transducer and activator of transcription 5 to yield improved T-cell persistence independent of CAR signaling, without apparent autonomous growth or transformation, and achieved potent rejection of CD19+ leukemia. Long-lived T cells were CD45ROnegCCR7+CD95+, phenotypically most similar to TSCM, and possessed a memory-like transcriptional profile. Overall, these results demonstrate that CAR+ T cells can develop long-term persistence with a memory stem-cell phenotype sustained by signaling through mbIL15. This observation warrants evaluation in clinical trials.


Asunto(s)
Interleucina-15/metabolismo , Neoplasias Experimentales/terapia , Receptores de Antígenos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/fisiología , Animales , Antígenos CD19/metabolismo , Humanos , Inmunoterapia Adoptiva , Activación de Linfocitos , Ratones , Células Precursoras de Linfocitos T/fisiología , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal
4.
PLoS One ; 11(8): e0159477, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27548616

RESUMEN

Adoptive immunotherapy infusing T cells with engineered specificity for CD19 expressed on B- cell malignancies is generating enthusiasm to extend this approach to other hematological malignancies, such as acute myelogenous leukemia (AML). CD123, or interleukin 3 receptor alpha, is overexpressed on most AML and some lymphoid malignancies, such as acute lymphocytic leukemia (ALL), and has been an effective target for T cells expressing chimeric antigen receptors (CARs). The prototypical CAR encodes a VH and VL from one monoclonal antibody (mAb), coupled to a transmembrane domain and one or more cytoplasmic signaling domains. Previous studies showed that treatment of an experimental AML model with CD123-specific CAR T cells was therapeutic, but at the cost of impaired myelopoiesis, highlighting the need for systems to define the antigen threshold for CAR recognition. Here, we show that CARs can be engineered using VH and VL chains derived from different CD123-specific mAbs to generate a panel of CAR+ T cells. While all CARs exhibited specificity to CD123, one VH and VL combination had reduced lysis of normal hematopoietic stem cells. This CAR's in vivo anti-tumor activity was similar whether signaling occurred via chimeric CD28 or CD137, prolonging survival in both AML and ALL models. Co-expression of inducible caspase 9 eliminated CAR+ T cells. These data help support the use of CD123-specific CARs for treatment of CD123+ hematologic malignancies.


Asunto(s)
Ingeniería Genética/métodos , Inmunoterapia Adoptiva/métodos , Subunidad alfa del Receptor de Interleucina-3/inmunología , Leucemia Mieloide Aguda/terapia , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Proteínas Recombinantes de Fusión/inmunología , Linfocitos T/trasplante , Animales , Linfocitos B/inmunología , Linfocitos B/patología , Antígenos CD28/genética , Antígenos CD28/inmunología , Caspasa 9/genética , Caspasa 9/inmunología , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Expresión Génica , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/patología , Humanos , Subunidad alfa del Receptor de Interleucina-3/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Terapia Molecular Dirigida , Plásmidos , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Proteínas Recombinantes de Fusión/genética , Anticuerpos de Dominio Único/genética , Linfocitos T/citología , Linfocitos T/inmunología , Transfección , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología
5.
Mol Imaging Biol ; 18(6): 838-848, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27246312

RESUMEN

PURPOSE: We have incorporated a positron emission tomography (PET) functionality in T cells expressing a CD19-specific chimeric antigen receptor (CAR) to non-invasively monitor the adoptively transferred cells. PROCEDURES: We engineered T cells to express CD19-specific CAR, firefly luciferase (ffLuc), and herpes simplex virus type-1 thymidine kinase (TK) using the non-viral-based Sleeping Beauty (SB) transposon/transposase system adapted for human application. Electroporated primary T cells were propagated on CD19+ artificial antigen-presenting cells. RESULTS: After 4 weeks, 90 % of cultured cells exhibited specific killing of CD19+ targets in vitro, could be ablated by ganciclovir, and were detected in vivo by bioluminescent imaging and PET following injection of 2'-deoxy-2'-[18F]fluoro-5-ethyl-1-ß-D-arabinofuranosyl-uracil ([18F]FEAU). CONCLUSION: This is the first report demonstrating the use of SB transposition to generate T cells which may be detected using PET laying the foundation for imaging the distribution and trafficking of T cells in patients treated for B cell malignancies.


Asunto(s)
Herpesvirus Humano 1/enzimología , Tomografía de Emisión de Positrones/métodos , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/metabolismo , Timidina Quinasa/metabolismo , Transposasas/metabolismo , Animales , Arabinofuranosil Uracilo/análogos & derivados , Arabinofuranosil Uracilo/química , Línea Celular , Ganciclovir/farmacología , Técnicas de Transferencia de Gen , Humanos , Luciferasas/metabolismo , Ratones , Radiofármacos/química , Transgenes , Xenopus
6.
Sci Rep ; 6: 21757, 2016 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-26902653

RESUMEN

Mismatch of human leukocyte antigens (HLA) adversely impacts the outcome of patients after allogeneic hematopoietic stem-cell transplantation (alloHSCT). This translates into the clinical requirement to timely identify suitable HLA-matched donors which in turn curtails the chances of recipients, especially those from a racial minority, to successfully undergo alloHSCT. We thus sought to broaden the existing pool of registered unrelated donors based on analysis that eliminating the expression of the HLA-A increases the chance for finding a donor matched at HLA-B, -C, and -DRB1 regardless of a patient's race. Elimination of HLA-A expression in HSC was achieved using artificial zinc finger nucleases designed to target HLA-A alleles. Significantly, these engineered HSCs maintain their ability to engraft and reconstitute hematopoiesis in immunocompromised mice. This introduced loss of HLA-A expression decreases the need to recruit large number of donors to match with potential recipients and has particular importance for patients whose HLA repertoire is under-represented in the current donor pool. Furthermore, the genetic engineering of stem cells provides a translational approach to HLA-match a limited number of third-party donors with a wide number of recipients.


Asunto(s)
Desoxirribonucleasas/genética , Eliminación de Gen , Antígenos HLA-A/genética , Trasplante de Células Madre Hematopoyéticas/etnología , Células Madre Hematopoyéticas/inmunología , Alelos , Animales , Desoxirribonucleasas/metabolismo , Selección de Donante/ética , Expresión Génica , Ingeniería Genética/métodos , Antígenos HLA-A/inmunología , Antígenos HLA-B/genética , Antígenos HLA-B/inmunología , Antígenos HLA-C/genética , Antígenos HLA-C/inmunología , Cadenas HLA-DRB1/genética , Cadenas HLA-DRB1/inmunología , Accesibilidad a los Servicios de Salud/ética , Trasplante de Células Madre Hematopoyéticas/ética , Células Madre Hematopoyéticas/citología , Prueba de Histocompatibilidad , Humanos , Ratones , Grupos Raciales , Trasplante Heterólogo , Trasplante Homólogo , Donante no Emparentado , Dedos de Zinc
7.
Cancer Res ; 75(17): 3505-18, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26330164

RESUMEN

Many tumors overexpress tumor-associated antigens relative to normal tissue, such as EGFR. This limits targeting by human T cells modified to express chimeric antigen receptors (CAR) due to potential for deleterious recognition of normal cells. We sought to generate CAR(+) T cells capable of distinguishing malignant from normal cells based on the disparate density of EGFR expression by generating two CARs from monoclonal antibodies that differ in affinity. T cells with low-affinity nimotuzumab-CAR selectively targeted cells overexpressing EGFR, but exhibited diminished effector function as the density of EGFR decreased. In contrast, the activation of T cells bearing high-affinity cetuximab-CAR was not affected by the density of EGFR. In summary, we describe the generation of CARs able to tune T-cell activity to the level of EGFR expression in which a CAR with reduced affinity enabled T cells to distinguish malignant from nonmalignant cells.


Asunto(s)
Antígenos de Neoplasias/inmunología , Receptores ErbB/inmunología , Neoplasias/inmunología , Receptores de Antígenos/inmunología , Linfocitos T/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales Humanizados/administración & dosificación , Línea Celular Tumoral , Cetuximab/administración & dosificación , Epítopos/inmunología , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/biosíntesis , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunoterapia Adoptiva , Ratones , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/patología , Receptores de Antígenos/uso terapéutico , Transducción de Señal , Linfocitos T/patología , Ensayos Antitumor por Modelo de Xenoinjerto
8.
PLoS One ; 10(6): e0128151, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26030772

RESUMEN

T cells modified with chimeric antigen receptors (CARs) targeting CD19 demonstrated clinical activity against some B-cell malignancies. However, this is often accompanied by a loss of normal CD19+ B cells and humoral immunity. Receptor tyrosine kinase-like orphan receptor-1 (ROR1) is expressed on sub-populations of B-cell malignancies and solid tumors, but not by healthy B cells or normal post-partum tissues. Thus, adoptive transfer of T cells specific for ROR1 has potential to eliminate tumor cells and spare healthy tissues. To test this hypothesis, we developed CARs targeting ROR1 in order to generate T cells specific for malignant cells. Two Sleeping Beauty transposons were constructed with 2nd generation ROR1-specific CARs signaling through CD3ζ and either CD28 (designated ROR1RCD28) or CD137 (designated ROR1RCD137) and were introduced into T cells. We selected for T cells expressing CAR through co-culture with γ-irradiated activating and propagating cells (AaPC), which co-expressed ROR1 and co-stimulatory molecules. Numeric expansion over one month of co-culture on AaPC in presence of soluble interleukin (IL)-2 and IL-21 occurred and resulted in a diverse memory phenotype of CAR+ T cells as measured by non-enzymatic digital array (NanoString) and multi-panel flow cytometry. Such T cells produced interferon-γ and had specific cytotoxic activity against ROR1+ tumors. Moreover, such cells could eliminate ROR1+ tumor xenografts, especially T cells expressing ROR1RCD137. Clinical trials will investigate the ability of ROR1-specific CAR+ T cells to specifically eliminate tumor cells while maintaining normal B-cell repertoire.


Asunto(s)
Elementos Transponibles de ADN/genética , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/metabolismo , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Linfocitos T/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Técnicas de Cocultivo , Citocinas/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Interferón gamma/biosíntesis , Leucemia/inmunología , Ratones , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Fenotipo , Linfocitos T/citología , Linfocitos T/inmunología , Transcripción Genética
9.
Clin Cancer Res ; 21(14): 3241-51, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25829402

RESUMEN

PURPOSE: The human endogenous retrovirus (HERV-K) envelope (env) protein is a tumor-associated antigen (TAA) expressed on melanoma but not normal cells. This study was designed to engineer a chimeric antigen receptor (CAR) on T-cell surface, such that they target tumors in advanced stages of melanoma. EXPERIMENTAL DESIGN: Expression of HERV-K protein was analyzed in 220 melanoma samples (with various stages of disease) and 139 normal organ donor tissues using immunohistochemical (IHC) analysis. HERV-K env-specific CAR derived from mouse monoclonal antibody was introduced into T cells using the transposon-based Sleeping Beauty (SB) system. HERV-K env-specific CAR(+) T cells were expanded ex vivo on activating and propagating cells (AaPC) and characterized for CAR expression and specificity. This includes evaluating the HERV-K-specific CAR(+) T cells for their ability to kill A375-SM metastasized tumors in a mouse xenograft model. RESULTS: We detected HERV-K env protein on melanoma but not in normal tissues. After electroporation of T cells and selection on HERV-K(+) AaPC, more than 95% of genetically modified T cells expressed the CAR with an effector memory phenotype and lysed HERV-K env(+) tumor targets in an antigen-specific manner. Even though there is apparent shedding of this TAA from tumor cells that can be recognized by HERV-K env-specific CAR(+) T cells, we observed a significant antitumor effect. CONCLUSIONS: Adoptive cellular immunotherapy with HERV-K env-specific CAR(+) T cells represents a clinically appealing treatment strategy for advanced-stage melanoma and provides an approach for targeting this TAA on other solid tumors.


Asunto(s)
Terapia Genética/métodos , Inmunoterapia Adoptiva/métodos , Melanoma/virología , Linfocitos T/trasplante , Proteínas Virales/inmunología , Animales , Ingeniería Genética/métodos , Humanos , Inmunohistoquímica , Melanoma/inmunología , Ratones , Ratones Endogámicos NOD , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/genética , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Proc Natl Acad Sci U S A ; 111(29): 10660-5, 2014 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-25002471

RESUMEN

Clinical-grade T cells are genetically modified ex vivo to express chimeric antigen receptors (CARs) to redirect their specificity to target tumor-associated antigens in vivo. We now have developed this molecular strategy to render cytotoxic T cells specific for fungi. We adapted the pattern-recognition receptor Dectin-1 to activate T cells via chimeric CD28 and CD3-ζ (designated "D-CAR") upon binding with carbohydrate in the cell wall of Aspergillus germlings. T cells genetically modified with the Sleeping Beauty system to express D-CAR stably were propagated selectively on artificial activating and propagating cells using an approach similar to that approved by the Food and Drug Administration for manufacturing CD19-specific CAR(+) T cells for clinical trials. The D-CAR(+) T cells exhibited specificity for ß-glucan which led to damage and inhibition of hyphal growth of Aspergillus in vitro and in vivo. Treatment of D-CAR(+) T cells with steroids did not compromise antifungal activity significantly. These data support the targeting of carbohydrate antigens by CAR(+) T cells and provide a clinically appealing strategy to enhance immunity for opportunistic fungal infections using T-cell gene therapy.


Asunto(s)
Aspergilosis/inmunología , Aspergilosis/terapia , Bioingeniería/métodos , Carbohidratos/antagonistas & inhibidores , Infecciones Oportunistas/inmunología , Infecciones Oportunistas/terapia , Linfocitos T/inmunología , Animales , Antígenos CD19/metabolismo , Aspergilosis/microbiología , Aspergilosis/patología , Aspergillus/efectos de los fármacos , Aspergillus/fisiología , Dexametasona/farmacología , Humanos , Hifa/efectos de los fármacos , Hifa/fisiología , Inmunofenotipificación , Lectinas Tipo C/metabolismo , Activación de Linfocitos/efectos de los fármacos , Ratones , Infecciones Oportunistas/patología , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/efectos de los fármacos
11.
Clin Cancer Res ; 20(22): 5708-19, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24833662

RESUMEN

PURPOSE: To activate and propagate populations of γδ T cells expressing polyclonal repertoire of γ and δ T-cell receptor (TCR) chains for adoptive immunotherapy of cancer, which has yet to be achieved. EXPERIMENTAL DESIGN: Clinical-grade artificial antigen-presenting cells (aAPC) derived from K562 tumor cells were used as irradiated feeders to activate and expand human γδ T cells to clinical scale. These cells were tested for proliferation, TCR expression, memory phenotype, cytokine secretion, and tumor killing. RESULTS: γδ T-cell proliferation was dependent upon CD137L expression on aAPC and addition of exogenous IL2 and IL21. Propagated γδ T cells were polyclonal as they expressed TRDV1, TRDV2-2, TRDV3, TRDV5, TRDV7, and TRDV8 with TRGV2, TRGV3F, TRGV7, TRGV8, TRGV9*A1, TRGV10*A1, and TRGV11 TCR chains. IFNγ production by Vδ1, Vδ2, and Vδ1(neg)Vδ2(neg) subsets was inhibited by pan-TCRγδ antibody when added to cocultures of polyclonal γδ T cells and tumor cell lines. Polyclonal γδ T cells killed acute and chronic leukemia, colon, pancreatic, and ovarian cancer cell lines, but not healthy autologous or allogeneic normal B cells. Blocking antibodies demonstrated that polyclonal γδ T cells mediated tumor cell lysis through combination of DNAM1, NKG2D, and TCRγδ. The adoptive transfer of activated and propagated γδ T cells expressing polyclonal versus defined Vδ TCR chains imparted a hierarchy (polyclonal>Vδ1>Vδ1(neg)Vδ2(neg)>Vδ2) of survival of mice with ovarian cancer xenografts. CONCLUSIONS: Polyclonal γδ T cells can be activated and propagated with clinical-grade aAPCs and demonstrate broad antitumor activities, which will facilitate the implementation of γδ T-cell cancer immunotherapies in humans.


Asunto(s)
Activación de Linfocitos/inmunología , Neoplasias/inmunología , Neoplasias/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Especificidad del Receptor de Antígeno de Linfocitos T/inmunología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Traslado Adoptivo , Animales , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Antígenos de Diferenciación de Linfocitos T/metabolismo , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Citocinas/biosíntesis , Citocinas/farmacología , Modelos Animales de Enfermedad , Expresión Génica , Humanos , Interferón gamma/biosíntesis , Ratones , Ratones Transgénicos , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Neoplasias/mortalidad , Neoplasias/terapia , ARN Mensajero , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Subgrupos de Linfocitos T/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Mol Ther ; 21(3): 638-47, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23295945

RESUMEN

Even though other γδ T-cell subsets exhibit antitumor activity, adoptive transfer of γδ Tcells is currently limited to one subset (expressing Vγ9Vδ2 T-cell receptor (TCR)) due to dependence on aminobisphosphonates as the only clinically appealing reagent for propagating γδ T cells. Therefore, we developed an approach to propagate polyclonal γδ T cells and rendered them bispecific through expression of a CD19-specific chimeric antigen receptor (CAR). Peripheral blood mononuclear cells (PBMC) were electroporated with Sleeping Beauty (SB) transposon and transposase to enforce expression of CAR in multiple γδ T-cell subsets. CAR(+)γδ T cells were expanded on CD19(+) artificial antigen-presenting cells (aAPC), which resulted in >10(9) CAR(+)γδ T cells from <10(6) total cells. Digital multiplex assay detected TCR mRNA coding for Vδ1, Vδ2, and Vδ3 with Vγ2, Vγ7, Vγ8, Vγ9, and Vγ10 alleles. Polyclonal CAR(+)γδ T cells were functional when TCRγδ and CAR were stimulated and displayed enhanced killing of CD19(+) tumor cell lines compared with CAR(neg)γδ T cells. CD19(+) leukemia xenografts in mice were reduced with CAR(+)γδ T cells compared with control mice. Since CAR, SB, and aAPC have been adapted for human application, clinical trials can now focus on the therapeutic potential of polyclonal γδ T cells.


Asunto(s)
Antígenos CD19/inmunología , Inmunoterapia Adoptiva/métodos , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Linfocitos T/inmunología , Animales , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Antígenos CD19/metabolismo , Línea Celular Tumoral , Electroporación , Humanos , Leucemia/terapia , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados , Transposasas/genética , Transposasas/metabolismo
13.
PLoS One ; 7(2): e29236, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22363401

RESUMEN

BACKGROUND: Inadequate placental development is associated with a high incidence of early embryonic lethality and serious pregnancy disorders in both humans and mice. However, the lack of well-defined trophoblast-specific gene regulatory elements has hampered investigations regarding the role of specific genes in placental development and fetal growth. PRINCIPAL FINDINGS: By random assembly of placental enhancers from two previously characterized genes, trophoblast specific protein α (Tpbpa) and adenosine deaminase (Ada), we identified a chimeric Tpbpa/Ada enhancer that when combined with the basal Ada promoter provided the highest luciferase activity in cultured human trophoblast cells, in comparison with non-trophoblast cell lines. We used this chimeric enhancer arrangement to drive the expression of a Cre recombinase transgene in the placentas of transgenic mice. Cre transgene expression occurred throughout the placenta but not in maternal organs examined or in the fetus. SIGNIFICANCE: In conclusion, we have provided both in vitro and in vivo evidence for a novel genetic system to achieve placental transgene expression by the use of a chimeric Tpbpa/Ada enhancer driven transgene. The availability of this expression vector provides transgenic opportunities to direct the production of desired proteins to the placenta.


Asunto(s)
ADN/genética , Marcación de Gen , Integrasas/metabolismo , Placenta/metabolismo , Recombinación Genética/genética , Adenosina Desaminasa/genética , Animales , Línea Celular , Células Cultivadas , Elementos de Facilitación Genéticos/genética , Femenino , Regulación de la Expresión Génica , Genes Reporteros/genética , Humanos , Luciferasas/genética , Masculino , Ratones , Ratones Transgénicos , Especificidad de Órganos/genética , Embarazo , Proteínas Gestacionales/genética , Regiones Promotoras Genéticas/genética , Transcripción Genética , Transgenes/genética , Trofoblastos/metabolismo
14.
Cancer Res ; 71(10): 3516-27, 2011 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-21558388

RESUMEN

Improving the therapeutic efficacy of T cells expressing a chimeric antigen receptor (CAR) represents an important goal in efforts to control B-cell malignancies. Recently an intrinsic strategy has been developed to modify the CAR itself to improve T-cell signaling. Here we report a second extrinsic approach based on altering the culture milieu to numerically expand CAR(+) T cells with a desired phenotype, for the addition of interleukin (IL)-21 to tissue culture improves CAR-dependent T-cell effector functions. We used electrotransfer of Sleeping Beauty system to introduce a CAR transposon and selectively propagate CAR(+) T cells on CD19(+) artificial antigen-presenting cells (aAPC). When IL-21 was present, there was preferential numeric expansion of CD19-specific T cells which lysed and produced IFN-γ in response to CD19. Populations of these numerically expanded CAR(+) T cells displayed an early memory surface phenotype characterized as CD62L(+)CD28(+) and a transcriptional profile of naïve T cells. In contrast, T cells propagated with only exogenous IL-2 tended to result in an overgrowth of CD19-specific CD4(+) T cells. Furthermore, adoptive transfer of CAR(+) T cells cultured with IL-21 exhibited improved control of CD19(+) B-cell malignancy in mice. To provide coordinated signaling to propagate CAR(+) T cells, we developed a novel mutein of IL-21 bound to the cell surface of aAPC that replaced the need for soluble IL-21. Our findings show that IL-21 can provide an extrinsic reprogramming signal to generate desired CAR(+) T cells for effective immunotherapy.


Asunto(s)
Antígenos CD19/biosíntesis , Linfocitos B/citología , Neoplasias Hematológicas/metabolismo , Inmunoterapia Adoptiva/métodos , Interleucina-12/metabolismo , Linfocitos T/inmunología , Animales , Antígenos CD19/metabolismo , Antígenos CD28/metabolismo , Linaje de la Célula , Técnicas de Cocultivo , Humanos , Interferón gamma/metabolismo , Interleucinas/metabolismo , Células K562 , Selectina L/metabolismo , Ratones , Ratones Endogámicos NOD , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Linfocitos T/metabolismo
15.
Circulation ; 121(3): 436-44, 2010 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-20065159

RESUMEN

BACKGROUND: Preeclampsia is a prevalent life-threatening hypertensive disorder of pregnancy. The circulating antiangiogenic factor, soluble endoglin (sEng), is elevated in the blood circulation of women with preeclampsia and contributes to disease pathology; however, the underlying mechanisms responsible for its induction in preeclampsia are unknown. METHODS AND RESULTS: Here, we discovered that a circulating autoantibody, the angiotensin receptor agonistic autoantibody (AT(1)-AA), stimulates sEng production via AT(1) angiotensin receptor activation in pregnant mice but not in nonpregnant mice. We subsequently demonstrated that the placenta is a major source contributing to sEng induction in vivo and that AT(1)-AA-injected pregnant mice display impaired placental angiogenesis. Using drug screening, we identified tumor necrosis factor-alpha as a circulating factor increased in the serum of autoantibody-injected pregnant mice contributing to AT(1)-AA-mediated sEng induction in human umbilical vascular endothelial cells. Subsequently, among all the drugs screened, we found that hemin, an inducer of heme oxygenase, functions as a break to control AT(1)-AA-mediated sEng induction by suppressing tumor necrosis factor-alpha signaling in human umbilical vascular endothelial cells. Finally, we demonstrated that the AT(1)-AA-mediated decreased angiogenesis seen in human placenta villous explants was attenuated by tumor necrosis factor-alpha-neutralizing antibodies, soluble tumor necrosis factor-alpha receptors, and hemin by abolishing both sEng and soluble fms-like tyrosine kinase-1 induction. CONCLUSIONS: Our findings demonstrate that AT(1)-AA-mediated tumor necrosis factor-alpha induction, by overcoming its negative regulator, heme oxygenase-1, is a key underlying mechanism responsible for impaired placental angiogenesis by inducing both sEng and soluble fms-like tyrosine kinase-1 secretion from human villous explants. Our results provide important new targets for diagnosis and therapeutic intervention in the management of preeclampsia.


Asunto(s)
Autoanticuerpos/sangre , Péptidos y Proteínas de Señalización Intracelular/sangre , Preeclampsia/sangre , Preeclampsia/inmunología , Receptor de Angiotensina Tipo 1/inmunología , Factor de Necrosis Tumoral alfa/sangre , Animales , Autoanticuerpos/farmacología , Vellosidades Coriónicas/irrigación sanguínea , Vellosidades Coriónicas/inmunología , Vellosidades Coriónicas/metabolismo , Endoglina , Células Endoteliales/metabolismo , Femenino , Hemo-Oxigenasa 1/metabolismo , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina G/farmacología , Ratones , Neovascularización Patológica/sangre , Neovascularización Patológica/inmunología , Placenta/irrigación sanguínea , Placenta/inmunología , Placenta/metabolismo , Embarazo , Receptor de Angiotensina Tipo 1/metabolismo , Transducción de Señal/fisiología , Solubilidad
16.
Hum Gene Ther ; 21(4): 427-37, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19905893

RESUMEN

Nonviral integrating vectors can be used for expression of therapeutic genes. piggyBac (PB), a transposon/transposase system, has been used to efficiently generate induced pluripotent stems cells from somatic cells, without genetic alteration. In this paper, we apply PB transposition to express a chimeric antigen receptor (CAR) in primary human T cells. We demonstrate that T cells electroporated to introduce the PB transposon and transposase stably express CD19-specific CAR and when cultured on CD19(+) artificial antigen-presenting cells, numerically expand in a CAR-dependent manner, display a phenotype associated with both memory and effector T cell populations, and exhibit CD19-dependent killing of tumor targets. Integration of the PB transposon expressing CAR was not associated with genotoxicity, based on chromosome analysis. PB transposition for generating human T cells with redirected specificity to a desired target such as CD19 is a new genetic approach with therapeutic implications.


Asunto(s)
Antígenos CD19/metabolismo , Elementos Transponibles de ADN , Linfoma de Células B/terapia , Receptores de Antígenos/genética , Linfocitos T/inmunología , Transposasas , Células Presentadoras de Antígenos/inmunología , Línea Celular Tumoral , Células Cultivadas , Técnicas de Cocultivo , Elementos Transponibles de ADN/genética , Elementos Transponibles de ADN/inmunología , Electroporación , Terapia Genética/métodos , Vectores Genéticos , Glioblastoma , Humanos , Células K562 , Plásmidos , Receptores de Antígenos/metabolismo , Linfocitos T/metabolismo , Transgenes , Transposasas/genética , Transposasas/inmunología
17.
FASEB J ; 24(3): 740-9, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19858092

RESUMEN

Priapism is a condition of persistent penile erection in the absence of sexual excitation. Of men with sickle cell disease (SCD), 40% display priapism. The disorder is a dangerous and urgent condition, given its association with penile fibrosis and eventual erectile dysfunction. Current strategies to prevent its progression are poor because of a lack of fundamental understanding of the molecular mechanisms for penile fibrosis in priapism. Here we demonstrate that increased adenosine is a novel causative factor contributing to penile fibrosis in two independent animal models of priapism, adenosine deaminase (ADA)-deficient mice and SCD transgenic mice. An important finding is that chronic reduction of adenosine by ADA enzyme therapy successfully attenuated penile fibrosis in both mouse models, indicating an essential role of increased adenosine in penile fibrosis and a novel therapeutic possibility for this serious complication. Subsequently, we identified that both mice models share a similar fibrotic gene expression profile in penile tissue (including procollagen I, TGF-beta(1), and plasminogen activator inhibitor-1 mRNA), suggesting that they share similar signaling pathways for progression to penile fibrosis. Thus, in an effort to decipher specific cell types and underlying mechanism responsible for adenosine-mediated penile fibrosis, we purified corpus cavernosal fibroblast cells (CCFCs), the major cell type involved in this process, from wild-type mice. Quantitative RT-PCR showed that the major receptor expressed in these cells is the adenosine receptor A(2B)R. Based on this fact, we further purified CCFCs from A(2B)R-deficient mice and demonstrated that A(2B)R is essential for excess adenosine-mediated penile fibrosis. Finally, we revealed that TGF-beta functions downstream of the A(2B)R to increase CCFC collagen secretion and proliferation. Overall, our studies identify an essential role of increased adenosine in the pathogenesis of penile fibrosis via A(2B)R signaling and offer a potential target for prevention and treatment of penile fibrosis, a dangerous complication seen in priapism.-Wen, J., Jiang, X., Dai, Y., Zhang, Y., Tang, Y., Sun, H., Mi, T., Phatarpekar, P. V., Kellems, R. E., Blackburn, M. R., Xia, Y. Increased adenosine contributes to penile fibrosis, a dangerous feature of priapism, via A(2B) adenosine receptor signaling.


Asunto(s)
Adenosina/metabolismo , Fibrosis/metabolismo , Fibrosis/patología , Pene/metabolismo , Pene/patología , Priapismo/patología , Receptor de Adenosina A2B/metabolismo , Actinas/metabolismo , Animales , Células Cultivadas , Cromatografía Líquida de Alta Presión , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Ratones Transgénicos , Priapismo/metabolismo , Receptor de Adenosina A2B/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Crecimiento Transformador beta1/metabolismo
18.
J Sex Med ; 7(9): 3011-22, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19845544

RESUMEN

INTRODUCTION: Priapism featured with painful prolonged penile erection is dangerous and commonly seen in sickle cell disease (SCD). The preventive approaches or effective treatment options for the disorder are limited because of poor understanding of its pathogenesis. Recent studies have revealed a novel role of excess adenosine in priapism caused by heightened cavernosal relaxation, and therefore present an intriguing mechanism-based therapeutic possibility. AIM: The aim of this study was to determine the therapeutic effects of adenosine deaminase (ADA) enzyme therapy to lower adenosine in priapism. METHODS: Both ADA-deficient mice and SCD transgenic (Tg) mice display priapism caused by excessive adenosine. Thus, we used these two distinct lines of mouse models of priapism as our investigative tools. Specifically, we treated both of these mice with different dosages of polyethylene glycol-modified ADA (PEG-ADA) to reduce adenosine levels in vivo. At the end points of the experiments, we evaluated the therapeutic effects of PEG-ADA treatment by measuring adenosine levels and monitoring the cavernosal relaxation. MAIN OUTCOME MEASURES: Adenosine levels in penile tissues were measured by high-performance liquid chromatography, and cavernosal relaxation was quantified by electrical field stimulation (EFS)-induced corporal cavernosal strip (CCS) assays. RESULTS: We found that lowering adenosine levels in penile tissues by PEG-ADA treatment from birth in ADA-deficient mice prevented the increased EFS-induced CCS relaxation associated with priapism. Intriguingly, in both ADA-deficient mice and SCD Tg mice with established priapism, we found that normalization of adenosine levels in penile tissues by PEG-ADA treatment relieved the heightened EFS-induced cavernosal relaxation in priapism. CONCLUSIONS: Our studies have identified that PEG-ADA is a novel, safe, and mechanism-based drug to prevent and correct excess adenosine-mediated increased cavernosal relaxation seen in two independent priapic animal models, and suggested its therapeutic possibility in men suffering from priapism.


Asunto(s)
Adenosina Desaminasa/farmacología , Relajación Muscular/efectos de los fármacos , Priapismo/tratamiento farmacológico , Priapismo/enzimología , Adenosina Desaminasa/deficiencia , Adenosina Desaminasa/metabolismo , Animales , Cromatografía Líquida de Alta Presión , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Estimulación Eléctrica , Masculino , Ratones , Ratones Noqueados , Músculo Liso/efectos de los fármacos , Pene/enzimología , Pene/inervación , Polietilenglicoles
19.
J Sex Med ; 6 Suppl 3: 292-301, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19267852

RESUMEN

INTRODUCTION: Priapism is defined as abnormal prolonged penile erection lasting at least for 4 hours occurring without sexual interest. Forty percent of sickle cell disease (SCD) patients display priapism. The disorder is dangerous and urgent given its association with ischemia-mediated erectile tissue damage and subsequent erectile dysfunction. Current strategies to manage the disorder are poor due to lack of fundamental understanding of the molecular mechanisms of priapism. Adenosine is a signaling nucleoside that elicits many pathophysiological effects by engaging membrane receptors. Recent evidence shows that adenosine may play an important role in priapism via adenosine receptor. AIM: To summarize the recent findings on the importance of adenosine signaling in the pathogenesis of priapism. MAIN OUTCOME MEASURES: Evidence in the literature on the association between adenosine signaling and the development of priapism. METHODS: This article reviews the literature that relates to the contributory role of adenosine signaling in priapism in multiple animal models and humans. RESULTS: Excessive adenosine accumulation in the penis, coupled with increased A(2B)R signaling, contributes to priapism in two independent lines of mutant mice. One is adenosine deaminase (ADA)-deficient mice, the only animal displaying spontaneously prolonged penile erection, and the other is SCD transgenic mice, a well-accepted priapic animal model. Both polyethylene glycol-modified ADA (PEG-ADA) enzyme therapy and A(2B)R antagonists are capable of inhibiting potent corpus cavernosal vascular relaxation associated with priapic-like activity seen in both ADA-deficient mice and SCD transgenic mice, indicating that PEG-ADA enzyme therapy is likely to be a novel therapy for such a dangerous urological disorder. CONCLUSION: Overall, the research reviewed here raises the intriguing possibility that elevated adenosine signaling contributes to priapism in general and that this signaling pathway represents a potentially important therapeutic target for the treatment of priapism.


Asunto(s)
Adenosina Desaminasa/metabolismo , Adenosina/fisiología , Inhibidores de Fosfodiesterasa/uso terapéutico , Priapismo , Transducción de Señal/fisiología , Adenosina/metabolismo , Animales , Endotelio Vascular/enzimología , Endotelio Vascular/patología , Endotelio Vascular/fisiopatología , Fibrosis/patología , Fibrosis/fisiopatología , Masculino , Ratones , Músculo Liso/metabolismo , Músculo Liso/patología , Músculo Liso/fisiopatología , Pene/patología , Pene/fisiopatología , Inhibidores de Fosfodiesterasa 5 , Priapismo/tratamiento farmacológico , Priapismo/enzimología , Priapismo/fisiopatología
20.
Nat Med ; 14(8): 855-62, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18660815

RESUMEN

Pre-eclampsia affects approximately 5% of pregnancies and remains a leading cause of maternal and neonatal mortality and morbidity in the United States and the world. The clinical hallmarks of this maternal disorder include hypertension, proteinuria, endothelial dysfunction and placental defects. Advanced-stage clinical symptoms include cerebral hemorrhage, renal failure and the HELLP (hemolysis, elevated liver enzymes and low platelets) syndrome. An effective treatment of pre-eclampsia is unavailable owing to the poor understanding of the pathogenesis of the disease. Numerous recent studies have shown that women with pre-eclampsia possess autoantibodies, termed AT(1)-AAs, that bind and activate the angiotensin II receptor type 1a (AT(1) receptor). We show here that key features of pre-eclampsia, including hypertension, proteinuria, glomerular endotheliosis (a classical renal lesion of pre-eclampsia), placental abnormalities and small fetus size appeared in pregnant mice after injection with either total IgG or affinity-purified AT(1)-AAs from women with pre-eclampsia. These features were prevented by co-injection with losartan, an AT(1) receptor antagonist, or by an antibody neutralizing seven-amino-acid epitope peptide. Thus, our studies indicate that pre-eclampsia may be a pregnancy-induced autoimmune disease in which key features of the disease result from autoantibody-induced angiotensin receptor activation. This hypothesis has obvious implications regarding pre-eclampsia screening, diagnosis and therapy.


Asunto(s)
Autoanticuerpos/química , Preeclampsia/inmunología , Receptores de Angiotensina/química , Animales , Enfermedades Autoinmunes/metabolismo , Epítopos/química , Femenino , Humanos , Hipertensión/complicaciones , Hipertensión/inmunología , Inmunoglobulina G/química , Riñón/inmunología , Ratones , Preeclampsia/metabolismo , Embarazo , Complicaciones Cardiovasculares del Embarazo/inmunología , Proteinuria/complicaciones , Proteinuria/inmunología , Receptores de Angiotensina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA