Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Neurobiol Dis ; 132: 104539, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31344492

RESUMEN

TAF1/MRSX33 intellectual disability syndrome is an X-linked disorder caused by loss-of-function mutations in the TAF1 gene. How these mutations cause dysmorphology, hypotonia, intellectual and motor defects is unknown. Mouse models which have embryonically targeted TAF1 have failed, possibly due to TAF1 being essential for viability, preferentially expressed in early brain development, and intolerant of mutation. Novel animal models are valuable tools for understanding neuronal pathology. Here, we report the development and characterization of a novel animal model for TAF1 ID syndrome in which the TAF1 gene is deleted in embryonic rats using clustered regularly interspaced short palindromic repeats (CRISPR) associated protein 9 (Cas9) technology and somatic brain transgenesis mediated by lentiviral transduction. Rat pups, post-natal day 3, were subjected to intracerebroventricular (ICV) injection of either gRNA-control or gRNA-TAF1 vectors. Rats were subjected to a battery of behavioral tests followed by histopathological analyses of brains at post-natal day 14 and day 35. TAF1-edited rats exhibited behavioral deficits at both the neonatal and juvenile stages of development. Deletion of TAF1 lead to a hypoplasia and loss of the Purkinje cells. We also observed a decreased in GFAP positive astrocytes and an increase in Iba1 positive microglia within the granular layer of the cerebellum in TAF1-edited animals. Immunostaining revealed a reduction in the expression of the CaV3.1 T-type calcium channel. Abnormal motor symptoms in TAF1-edited rats were associated with irregular cerebellar output caused by changes in the intrinsic activity of the Purkinje cells due to loss of pre-synaptic CaV3.1. This animal model provides a powerful new tool for studies of neuronal dysfunction in conditions associated with TAF1 abnormalities and should prove useful for developing therapeutic strategies to treat TAF1 ID syndrome.


Asunto(s)
Sistemas CRISPR-Cas/genética , Cerebelo/patología , Corteza Cerebral/patología , Edición Génica/métodos , Histona Acetiltransferasas/genética , Factores Asociados con la Proteína de Unión a TATA/genética , Factor de Transcripción TFIID/genética , Animales , Animales Recién Nacidos , Cerebelo/anomalías , Cerebelo/fisiología , Corteza Cerebral/anomalías , Corteza Cerebral/fisiología , Femenino , Inyecciones Intraventriculares , Locomoción/fisiología , Técnicas de Cultivo de Órganos , Embarazo , Ratas , Ratas Sprague-Dawley
2.
Immunology ; 143(1): 96-108, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24678989

RESUMEN

A growing body of evidence suggests that inflammatory cytokines have a dualistic role in immunity. In this study, we sought to determine the direct effects of interferon-γ (IFN-γ) on the differentiation and maturation of human peripheral blood monocyte-derived dendritic cells (moDC). Here, we report that following differentiation of monocytes into moDC with granulocyte-macrophage colony-stimulating factor and interleukin-4, IFN-γ induces moDC maturation and up-regulates the co-stimulatory markers CD80/CD86/CD95 and MHC Class I, enabling moDC to effectively generate antigen-specific CD4(+) and CD8(+) T-cell responses for multiple viral and tumour antigens. Early exposure of monocytes to high concentrations of IFN-γ during differentiation promotes the formation of macrophages. However, under low concentrations of IFN-γ, monocytes continue to differentiate into dendritic cells possessing a unique gene-expression profile, resulting in impairments in subsequent maturation by IFN-γ or lipopolysaccharide and an inability to generate effective antigen-specific CD4(+) and CD8(+) T-cell responses. These findings demonstrate that IFN-γ imparts differential programmes on moDC that shape the antigen-specific T-cell responses they induce. Timing and intensity of exposure to IFN-γ can therefore determine the functional capacity of moDC.


Asunto(s)
Diferenciación Celular/inmunología , Células Dendríticas/inmunología , Interferón gamma/inmunología , Monocitos/citología , Monocitos/inmunología , Línea Celular , Citocinas/biosíntesis , Células Dendríticas/citología , Citometría de Flujo , Humanos , Transcriptoma
3.
J Immunol ; 184(11): 5988-98, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20427771

RESUMEN

Tyrosinase, an enzyme involved in melanin synthesis, is expressed in nearly all primary and metastatic melanoma lesions and thus is an attractive target for TCR-based gene therapy using adoptive cell transfer. The TCR alpha- and beta-chain genes from a tumor-infiltrating lymphocyte, which recognized the tyrosinase 368-376 peptide in the context of HLA-A2, were cloned into a gamma-retroviral vector. Following transduction of PBL, specific reactivity was confirmed by cytokine production following coculture with tumor targets. Experiments using Ab blockade and CD4/CD8 sorting of the transduced PBLs demonstrated that this antityrosinase TCR was CD4/CD8 independent. The introduction of a second disulfide bond between the TCR constant regions and/or creation of a chimeric protein in which the human constant regions were replaced by murine homologs resulted in enhanced TCR expression as demonstrated by tetramer staining and improved tumor reactivity that was comparable to PBL transduced with either anti-melanoma Ag recognized by T cells-1 or anti-gp100 TCR vectors currently used in clinical trials. The chimeric TCR also allowed us to test antitumor function of in HLA-A2/K(b)-transgenic mice. Transfer of the antityrosinase TCR into mouse splenocytes conferred CD4/CD8-independent, HLA-A2-restricted Ag reactivity against B16/A2K(b) murine melanoma in vitro. Furthermore, adoptive transfer of transduced splenocytes mediated B16/A2K(b) melanoma tumor regression in lymphodepleted mice, and, surprisingly, both CD8 and CD4 T cells were equally effective in mediating tumor regression. These results suggest that this highly active tyrosinase-specific TCR could be of value in adoptive cell transfer for melanoma.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Terapia Genética/métodos , Melanoma Experimental/inmunología , Monofenol Monooxigenasa/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Traslado Adoptivo , Animales , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Separación Celular , Citometría de Flujo , Vectores Genéticos , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma Experimental/terapia , Ratones , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/genética , Retroviridae/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción Genética
4.
Methods Mol Biol ; 515: 137-50, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19378113

RESUMEN

In the last decade lentiviral gene transfer vectors have gained significant place both in basic science and gene therapy applications. A number of gene transfer applications would benefit from vectors capable of expressing multiple genes. This chapter focuses on production of bicistronic and tricistronic lentiviral vectors based on the internal ribosomal entry site (IRES) sequence of encephalomyocarditis virus (EMCV) and/or foot-and-mouth disease virus (FMDV) cleavage factor 2A. Multigene vectors produced high titer viral particles and were able to simultaneously express two or three transgenes in transduced cells. The level of expression of individual transgenes varied depending on the transgene itself, its position within the construct, the total number of transgenes expressed, the strategy used for multigene expression, and the number of copies of proviral insertions.


Asunto(s)
Técnicas de Transferencia de Gen , Vectores Genéticos/genética , VIH-1/genética , Lentivirus/genética , Línea Celular , Electroforesis en Gel de Poliacrilamida , Citometría de Flujo/instrumentación , Citometría de Flujo/métodos , Genes Reporteros/genética , Humanos , Técnicas de Cultivo de Tejidos
5.
Virol J ; 3: 14, 2006 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-16539700

RESUMEN

BACKGROUND: A number of gene therapy applications would benefit from vectors capable of expressing multiple genes. In this study we explored the feasibility and efficiency of expressing two or three transgenes in HIV-1 based lentiviral vector. Bicistronic and tricistronic self-inactivating lentiviral vectors were constructed employing the internal ribosomal entry site (IRES) sequence of encephalomyocarditis virus (EMCV) and/or foot-and-mouth disease virus (FMDV) cleavage factor 2A. We employed enhanced green fluorescent protein (eGFP), O6-methylguanine-DNA-methyltransferase (MGMT), and homeobox transcription factor HOXB4 as model genes and their expression was detected by appropriate methods including fluorescence microscopy, flow cytometry, immunocytochemistry, biochemical assay, and western blotting. RESULTS: All the multigene vectors produced high titer virus and were able to simultaneously express two or three transgenes in transduced cells. However, the level of expression of individual transgenes varied depending on: the transgene itself; its position within the construct; the total number of transgenes expressed; the strategy used for multigene expression and the average copy number of pro-viral insertions. Notably, at limiting MOI, the expression of eGFP in a bicistronic vector based on 2A was approximately 4 times greater than that of an IRES based vector. CONCLUSION: The small and efficient 2A sequence can be used alone or in combination with an IRES for the construction of multicistronic lentiviral vectors which can express encoded transgenes at functionally relevant levels in cells containing an average of one pro-viral insert.


Asunto(s)
Virus de la Fiebre Aftosa/genética , Vectores Genéticos/genética , Lentivirus/genética , Virus de la Encefalomiocarditis/genética , Terapia Genética , Proteínas Fluorescentes Verdes/genética , Células HeLa , Proteínas de Homeodominio/genética , Humanos , Células K562 , O(6)-Metilguanina-ADN Metiltransferasa/genética , Ribosomas/metabolismo , Factores de Transcripción/genética , Transgenes
6.
Hum Gene Ther ; 15(8): 758-69, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15319033

RESUMEN

Lentiviral vectors are capable of efficiently transducing nondividing and slowly dividing cells, including hematopoietic stem cells, resulting in stable integration and sustained transgene expression. We constructed human immunodeficiency virus type 1-based self-inactivating lentiviral vectors to express either wild-type or an O6-benzylguanine (O6-beG)-resistant mutant form of the human O6-alkylguanine-DNA methyltransferase (MGMT; DNA-O6-methylguanine:[protein]-L-cysteine S-methyltransferase, EC 2.1.1.63) and transduced K562 and granulocyte colony-stimulating factor-mobilized human peripheral blood CD34+ cells. After transduction, K562 cells expressed high levels of MGMT as determined by Western blot, immunocytochemistry, and biochemical assay. A colony-forming survival assay showed significant protection against O6-beG plus 1,3-bis(2-chloroethyl)-nitrosourea (BCNU) or temozolomide (TMZ) toxicity. Similarly, a single transduction of CD34+ cells resulted in a 13- to 14-fold increase in the level of MGMT expression. In comparison with non-transduced cells, mutant MGMTP140K-transduced CD34+ cells showed significant resistance against the combined toxicity of O6-beG with either TMZ or BCNU: there was an approximately 9-fold increase in the survival of colony-forming cells as indicated by the IC50 values after O6-beG plus TMZ treatment and an approximately 5-fold increase in the case of O6-beG plus BCNU treatment. These results show that lentivirus-mediated expression of MGMTP140K can efficiently protect the hematopoietic compartment against the combined toxicity of O6-beG plus TMZ or BCNU.


Asunto(s)
Dacarbazina/análogos & derivados , Regulación Enzimológica de la Expresión Génica , Terapia Genética/métodos , Vectores Genéticos/genética , Células Madre Hematopoyéticas/enzimología , Lentivirus/genética , O(6)-Metilguanina-ADN Metiltransferasa/genética , Antígenos CD34/metabolismo , Western Blotting , Carmustina/toxicidad , Línea Celular , Ensayo de Unidades Formadoras de Colonias , Cartilla de ADN , Dacarbazina/toxicidad , Citometría de Flujo , Vectores Genéticos/metabolismo , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Inmunohistoquímica , Concentración 50 Inhibidora , Reacción en Cadena de la Polimerasa , Temozolomida , Transducción Genética , Transgenes/genética , Células Tumorales Cultivadas
7.
Cancer Res ; 73(11): 3371-80, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23633494

RESUMEN

Most systemic cancer therapies target tumor cells directly, although there is increasing interest in targeting the tumor stroma that can comprise a substantial portion of the tumor mass. We report here a synergy between two T-cell therapies, one directed against the stromal tumor vasculature and the other directed against antigens expressed on the tumor cell. Simultaneous transfer of genetically engineered syngeneic T cells expressing a chimeric antigen receptor targeting the VEGF receptor-2 (VEGFR2; KDR) that is overexpressed on tumor vasculature and T-cells specific for the tumor antigens gp100 (PMEL), TRP-1 (TYRP1), or TRP-2 (DCT) synergistically eradicated established B16 melanoma tumors in mice and dramatically increased the tumor-free survival of mice compared with treatment with either cell type alone or T cells coexpressing these two targeting molecules. Host lymphodepletion before cell transfer was required to mediate the antitumor effect. The synergistic antitumor response was accompanied by a significant increase in the infiltration and expansion and/or persistence of the adoptively transferred tumor antigen-specific T cells in the tumor microenvironment and thus enhanced their antitumor potency. The data presented here emphasize the possible beneficial effects of combining antiangiogenic with tumor-specific immunotherapeutic approaches for the treatment of patients with cancer.


Asunto(s)
Antígenos de Neoplasias/inmunología , Inmunoterapia Adoptiva/métodos , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Linfocitos T/inmunología , Animales , Modelos Animales de Enfermedad , Activación de Linfocitos/inmunología , Melanoma Experimental/irrigación sanguínea , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/inmunología , Neovascularización Patológica/terapia , Receptores de Antígenos/genética , Receptores de Antígenos/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/inmunología
8.
J Exp Med ; 210(6): 1125-35, 2013 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-23712432

RESUMEN

Fibroblast activation protein (FAP) is a candidate universal target antigen because it has been reported to be selectively expressed in nearly all solid tumors by a subset of immunosuppressive tumor stromal fibroblasts. We verified that 18/18 human tumors of various histologies contained pronounced stromal elements staining strongly for FAP, and hypothesized that targeting tumor stroma with FAP-reactive T cells would inhibit tumor growth in cancer-bearing hosts. T cells genetically engineered with FAP-reactive chimeric antigen receptors (CARs) specifically degranulated and produced effector cytokines upon stimulation with FAP or FAP-expressing cell lines. However, adoptive transfer of FAP-reactive T cells into mice bearing a variety of subcutaneous tumors mediated limited antitumor effects and induced significant cachexia and lethal bone toxicities in two mouse strains. We found that FAP was robustly expressed on PDGFR-α(+), Sca-1(+) multipotent bone marrow stromal cells (BMSCs) in mice, as well as on well-characterized, clinical-grade multipotent human BMSCs. Accordingly, both mouse and human multipotent BMSCs were recognized by FAP-reactive T cells. The lethal bone toxicity and cachexia observed after cell-based immunotherapy targeting FAP cautions against its use as a universal target. Moreover, the expression of FAP by multipotent BMSCs may point toward the cellular origins of tumor stromal fibroblasts.


Asunto(s)
Caquexia/inmunología , Fibroblastos/inmunología , Gelatinasas/inmunología , Proteínas de la Membrana/inmunología , Células Madre Mesenquimatosas/inmunología , Serina Endopeptidasas/inmunología , Animales , Antígenos Ly/inmunología , Antígenos de Neoplasias/inmunología , Caquexia/patología , Línea Celular Tumoral , Endopeptidasas , Femenino , Humanos , Masculino , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos BALB C , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/inmunología , Receptores de Antígenos/inmunología , Linfocitos T/inmunología
9.
Clin Cancer Res ; 18(6): 1672-83, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22291136

RESUMEN

PURPOSE: We investigated the feasibility of delivering the proinflammatory cytokine interleukin (IL)-12 into tumor using T cells genetically engineered to express a chimeric antigen receptor (CAR) against the VEGF receptor-2 (VEGFR-2). EXPERIMENTAL DESIGN: Two different strains of mice bearing five different established subcutaneous tumors were treated with syngeneic T cells cotransduced with an anti-VEGFR-2 CAR and a constitutively expressed single-chain murine IL-12 or an inducible IL-12 gene after host lymphodepletion. Tumor regression, survival of mice, and persistence of the transferred cells were evaluated. RESULTS: Adoptive transfer of syngeneic T cells cotransduced with an anti-VEGFR-2 CAR and a constitutively expressing single-chain IL-12 resulted in the regression of five different established tumors of different histologies without the need for IL-2 administration. T cells transduced with either anti-VEGFR-2 CAR or single-chain IL-12 alone did not alter the tumor growth indicating that both of them had to be expressed in the same cell to mediate tumor regression. Anti-VEGFR-2 CAR and IL-12-cotransduced T cells infiltrated the tumors, expanded, and persisted for prolonged periods. The antitumor effect did not require the presence of host T and B cells but was dependent on host IL-12R-expressing cells. The anti-VEGFR-2 CAR changed the immunosuppressive tumor environment by altering/reducing both the systemic and the intratumoral CD11b(+)Gr1(+) myeloid suppressor cell subsets that expressed VEGFR-2. CONCLUSIONS: These results suggest that targeted delivery of IL-12 into the tumor environment with T cells redirected against VEGFR-2 is a promising approach for treating patients with a variety of solid tumor types.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Interleucina-12/administración & dosificación , Interleucina-12/genética , Neoplasias/terapia , Linfocitos T/inmunología , Transducción Genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/inmunología , Animales , Estudios de Factibilidad , Femenino , Humanos , Interleucina-12/metabolismo , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neoplasias/irrigación sanguínea , Receptores de Antígenos de Linfocitos T/inmunología , Proteínas Recombinantes/metabolismo
10.
J Clin Invest ; 121(12): 4746-57, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22056381

RESUMEN

Solid tumors are complex masses with a local microenvironment, or stroma, that supports tumor growth and progression. Among the diverse tumor-supporting stromal cells is a heterogeneous population of myeloid-derived cells. These cells are alternatively activated and contribute to the immunosuppressive environment of the tumor; overcoming their immunosuppressive effects may improve the efficacy of cancer immunotherapies. We recently found that engineering tumor-specific CD8(+) T cells to secrete the inflammatory cytokine IL-12 improved their therapeutic efficacy in the B16 mouse model of established melanoma. Here, we report the mechanism underlying this finding. Surprisingly, direct binding of IL-12 to receptors on lymphocytes or NK cells was not required. Instead, IL-12 sensitized bone marrow-derived tumor stromal cells, including CD11b(+)F4/80(hi) macrophages, CD11b(+)MHCII(hi)CD11c(hi) dendritic cells, and CD11b(+)Gr-1(hi) myeloid-derived suppressor cells, causing them to enhance the effects of adoptively transferred CD8(+) T cells. This reprogramming of myeloid-derived cells occurred partly through IFN-γ. Surprisingly, direct presentation of antigen to the transferred CD8(+) T cells by tumor was not necessary; however, MHCI expression on host cells was essential for IL-12-mediated antitumor enhancements. These results are consistent with a model in which IL-12 enhances the ability of CD8(+) T cells to collapse large vascularized tumors by triggering programmatic changes in otherwise suppressive antigen-presenting cells within tumors and support the use of IL-12 as part of immunotherapy for the treatment of solid tumors.


Asunto(s)
Linfocitos T CD8-positivos/trasplante , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Inmunoterapia Adoptiva , Interleucina-12/fisiología , Melanoma Experimental/terapia , Células Mieloides/fisiología , Escape del Tumor/inmunología , Microambiente Tumoral/inmunología , Animales , Presentación de Antígeno , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/patología , Antígenos de Neoplasias/inmunología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/patología , Linfocitos T CD8-positivos/metabolismo , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/inmunología , Inflamación/genética , Interferón gamma/fisiología , Interleucina-12/genética , Interleucina-12/metabolismo , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/patología , Macrófagos/fisiología , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Quimera por Radiación , Proteínas Recombinantes de Fusión/fisiología , Células del Estroma/patología , Células del Estroma/fisiología
11.
J Immunother ; 34(4): 343-52, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21499127

RESUMEN

T-cell receptor (TCR) gene therapy enables for the rapid creation of antigen-specific T cells from mice of any strain and represents a valuable tool for preclinical immunotherapy studies. Here, we describe the superiority of γ-retroviral vectors compared with lentiviral vectors for transduction of murine T cells and surprisingly illustrate robust gene-transfer into phenotypically naive/memory-stem cell like (TN/TSCM; CD62L(hi)/CD44(low)) and central memory (TCM; CD62L(hi)/CD44(hi)) CD8+ T cells using murine stem cell-based γ-retroviral vectors (MSGV1). We created MSGV1 vectors for a major histocompatibility complex-class I-restricted TCR specific for the melanocyte-differentiation antigen, glycoprotein 100 (MSGV1-pmel-1), and a major histocompatibility complex-class II-restricted TCR specific for tyrosinase-related protein-1 (MSGV1-TRP-1), and found that robust gene expression required codon optimization of TCR sequences for the pmel-1 TCR. To test for functionality, we adoptively transferred TCR-engineered T cells into mice bearing B16 melanomas and observed delayed growth of established tumors with pmel-1 TCR engineered CD8+ T cells and significant tumor regression with TRP-1 TCR transduced CD4 T cells. We simultaneously created lentiviral vectors encoding the pmel-1 TCR, but found that these vectors mediated low TCR expression in murine T cells, but robust gene expression in other murine and human cell lines. These results indicate that preclinical murine models of adoptive immunotherapies are more practical using γ-retroviral rather than lentiviral vectors.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Ingeniería Genética , Inmunoterapia Adoptiva , Animales , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Células HEK293 , Humanos , Células Jurkat , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células 3T3 NIH , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Retroviridae/genética , Transducción Genética , Antígeno gp100 del Melanoma/genética , Antígeno gp100 del Melanoma/inmunología
12.
J Immunother ; 33(7): 672-83, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20664359

RESUMEN

Adoptive cell transfer using autologous tumor infiltrating lymphocytes or lymphocytes transduced with antitumor T-cell receptor (TCR) is an effective therapy for patients with metastatic melanoma. A limiting factor in the effectiveness of this treatment is the apoptosis of the transferred cells when Interleukin-2 (IL-2) administration is withdrawn. In an attempt to improve persistence of the transferred lymphocytes, we cotransduced human peripheral blood lymphocytes with retroviruses encoding Bcl-2 or Bcl-xL, antiapoptotic genes of the BCL2 family, and the MART-1 melanoma tumor antigen-specific TCR, DMF5. Lymphocytes were cotransduced with 38% to 64% cotransduction efficiency, and exhibited a marked delay in apoptosis after IL-2 withdrawal. Cotransduction with Bcl-2 or Bcl-xL did not affect cytokine secretion or lytic ability of the DMF5-transduced lymphocytes. After 5 days of IL-2 withdrawal, cotransduced lymphocytes produced similar levels of IFN-γ per cell as DMF5-alone transduced lymphocytes in response to tumor cells. Cotransduction did not alter the phenotype of lymphocytes with respect to a panel of T-cell differentiation markers. In a mouse model of melanoma, adoptively transferred T cells transduced with Bcl-2 persisted better in vivo at the site of tumor, 13 and 21 days after adoptive transfer (P=0.0064 and 0.041, respectively), with evidence of enrichment of the Bcl-2-transduced population over time (P<0.0001). Thus, by coexpressing Bcl-2 or Bcl-xL with a tumor-specific TCR, we have engineered a lymphocyte that resists apoptosis owing to IL-2 withdrawal without altering its tumor-specific function or phenotype, and thus may show improved antitumor effectiveness in vivo after cell transfer.


Asunto(s)
Inmunoterapia Adoptiva , Melanoma/inmunología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Retroviridae/genética , Linfocitos T/efectos de los fármacos , Traslado Adoptivo , Animales , Antígenos de Diferenciación/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/genética , Vacunas contra el Cáncer , Supervivencia Celular/genética , Ingeniería Genética , Humanos , Interferón gamma/metabolismo , Interleucina-2/farmacología , Antígeno MART-1/inmunología , Melanoma/terapia , Melanoma Experimental , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/inmunología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/patología
13.
Cancer Res ; 70(15): 6171-80, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20631075

RESUMEN

Adoptive cell transfer (ACT)-based immunotherapies can mediate objective cancer regression in animal models and in up to 70% of patients with metastatic melanoma; however, it remains unclear whether the tumor vasculature impedes the egress of tumor-specific T cells, thus hindering this immunotherapy. Disruption of the proangiogenic interaction of vascular endothelial growth factor (VEGF) with its receptor (VEGFR-2) has been reported to "normalize" tumor vasculature, enhancing the efficacy of chemotherapeutic agents by increasing their delivery to the tumor intersitium. We thus sought to determine whether disrupting VEGF/VEGFR-2 signaling could enhance the effectiveness of ACT in a murine cancer model. The administration of an antibody against mouse VEGF synergized with ACT to enhance inhibition of established, vascularized, B16 melanoma (P = 0.009) and improve survival (P = 0.003). Additive effects of an antibody against VEGFR-2 in conjunction with ACT were seen in this model (P = 0.013). Anti-VEGF, but not anti-VEGFR-2, antibody significantly increased infiltration of transferred cells into the tumor. Thus, normalization of tumor vasculature through disruption of the VEGF/VEGFR-2 axis can increase extravasation of adoptively transferred T cells into the tumor and improve ACT-based immunotherapy. These studies provide a rationale for the exploration of combining antiangiogenic agents with ACT for the treatment of patients with cancer.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Inmunoterapia Adoptiva/métodos , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma Experimental/terapia , Neoplasias Cutáneas/terapia , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Bevacizumab , Terapia Combinada , Epítopos de Linfocito T/inmunología , Melanoma Experimental/inmunología , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias Cutáneas/inmunología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/inmunología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/inmunología , Irradiación Corporal Total , Antígeno gp100 del Melanoma
14.
J Clin Invest ; 120(11): 3953-68, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20978347

RESUMEN

Immunotherapies based on adoptive cell transfer are highly effective in the treatment of metastatic melanoma, but the use of this approach in other cancer histologies has been hampered by the identification of appropriate target molecules. Immunologic approaches targeting tumor vasculature provide a means for the therapy of multiple solid tumor types. We developed a method to target tumor vasculature, using genetically redirected syngeneic or autologous T cells. Mouse and human T cells were engineered to express a chimeric antigen receptor (CAR) targeted against VEGFR-2, which is overexpressed in tumor vasculature and is responsible for VEGF-mediated tumor progression and metastasis. Mouse and human T cells expressing the relevant VEGFR-2 CARs mediated specific immune responses against VEGFR-2 protein as well as VEGFR-2-expressing cells in vitro. A single dose of VEGFR-2 CAR-engineered mouse T cells plus exogenous IL-2 significantly inhibited the growth of 5 different types of established, vascularized syngeneic tumors in 2 different strains of mice and prolonged the survival of mice. T cells transduced with VEGFR-2 CAR showed durable and increased tumor infiltration, correlating with their antitumor effect. This approach provides a potential method for the gene therapy of a variety of human cancers.


Asunto(s)
Terapia Genética/métodos , Linfocitos/fisiología , Neoplasias Experimentales , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Traslado Adoptivo , Animales , Línea Celular , Femenino , Vectores Genéticos , Humanos , Interleucina-2/genética , Interleucina-2/inmunología , Linfocitos/citología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Linfocitos T/inmunología , Linfocitos T/fisiología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
15.
Cancer Immunol Immunother ; 56(6): 885-95, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17102977

RESUMEN

Prostatic acid phosphatase (PAP) is a prostate cancer tumor antigen and a prostate-specific protein shared by rats and humans. Previous studies indicated that Copenhagen rats immunized with a recombinant vaccinia virus expressing human PAP (hPAP) developed PAP-specific cytotoxic T cells (CTL) with cross reactivity to rat PAP (rPAP) and evidence of prostate inflammation. Viral delivery of vaccine antigens is an active area of clinical investigation. However, a potential difficulty with viral-based immunizations is that immune responses elicited to the viral vector might limit the possibility of multiple immunizations. In this paper, we investigate the ability of another genetic immunization method, a DNA vaccine encoding PAP, to elicit antigen-specific CD8+ T cell immune responses. Specifically, Lewis rats were immunized with either a plasmid DNA-based (pTVG-HP) or vaccinia-based (VV-HP) vaccine each encoding hPAP. We determined that rats immunized with a DNA vaccine encoding hPAP developed a Th1-biased immune response as indicated by proliferating PAP-specific CD4+ and CD8+ cells and IFNgamma production. Rats immunized with vaccinia virus encoding PAP did not develop a PAP-specific response unless boosted with a heterologous vaccination scheme. Most importantly, multiple immunizations with a DNA vaccine encoding the rat PAP homologue (pTVG-RP) could overcome peripheral self-tolerance against rPAP and generate a Th1-biased antigen-specific CD4+ and CD8+ T cell response. Overall, DNA vaccines provide a safe and effective method of generating prostate antigen-specific T cell responses. These findings support the investigation of PAP-specific DNA vaccines in human clinical trials.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Neoplasias de la Próstata/terapia , Proteínas Tirosina Fosfatasas/inmunología , Vacunas de ADN/inmunología , Fosfatasa Ácida , Animales , Autoantígenos/inmunología , Vacunas contra el Cáncer/administración & dosificación , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Vectores Genéticos/inmunología , Humanos , Masculino , Plásmidos , Neoplasias de la Próstata/inmunología , Ratas , Ratas Endogámicas Lew , Transducción Genética , Vacunas de ADN/administración & dosificación , Virus Vaccinia/genética
16.
Cancer Immunol Immunother ; 55(12): 1504-14, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16612600

RESUMEN

Fusion proteins consisting of the ligand-binding domain of CTLA4 covalently attached to an antigen (Ag) are potent immunogens. This fusion strategy effectively induces Ag-specific immunity both when introduced as a DNA-based vaccine and as a recombinant protein. CTLA4 is a ligand for B7 molecules expressed on the surface of antigen-presenting cells (APCs), and this interaction is critical for the fusion protein to stimulate Ag-specific immunity. We show that interaction of the fusion protein with either B7-1 or B7-2 is sufficient to stimulate immune activity, and that T cells are essential for the development of IgG responses. In addition, we demonstrate that human dendritic cells (DCs) pulsed with CTLA4-Ag fusion proteins can efficiently present Ag to T cells and induce an Ag-specific immune response in vitro. These studies provide further mechanistic understanding of the process by which CTLA4-Ag fusion proteins stimulate the immune system, and represent an efficient means of generating Ag-specific T cells for immunotherapy.


Asunto(s)
Antígenos CD/genética , Antígenos de Diferenciación/genética , Antígenos/inmunología , Activación de Linfocitos , Proteínas Recombinantes de Fusión/inmunología , Animales , Formación de Anticuerpos , Antígenos/genética , Antígenos CD/química , Antígenos de Diferenciación/química , Antígeno B7-1/inmunología , Antígeno B7-2/inmunología , Antígeno CTLA-4 , Células Dendríticas/efectos de los fármacos , Humanos , Inmunoglobulina G/inmunología , Ligandos , Ratones , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/genética , Linfocitos T/inmunología , Linfocitos T Citotóxicos/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA