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

RESUMO

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.


Assuntos
Sistemas CRISPR-Cas/genética , Cerebelo/patologia , Córtex Cerebral/patologia , Edição de Genes/métodos , Histona Acetiltransferases/genética , Fatores Associados à Proteína de Ligação a TATA/genética , Fator de Transcrição TFIID/genética , Animais , Animais Recém-Nascidos , Cerebelo/anormalidades , Cerebelo/fisiologia , Córtex Cerebral/anormalidades , Córtex Cerebral/fisiologia , Feminino , Injeções Intraventriculares , Locomoção/fisiologia , Técnicas de Cultura de Órgãos , Gravidez , Ratos , Ratos Sprague-Dawley
2.
Immunology ; 143(1): 96-108, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24678989

RESUMO

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.


Assuntos
Diferenciação Celular/imunologia , Células Dendríticas/imunologia , Interferon gama/imunologia , Monócitos/citologia , Monócitos/imunologia , Linhagem Celular , Citocinas/biossíntese , Células Dendríticas/citologia , Citometria de Fluxo , Humanos , Transcriptoma
3.
J Immunol ; 184(11): 5988-98, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20427771

RESUMO

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.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Terapia Genética/métodos , Melanoma Experimental/imunologia , Monofenol Mono-Oxigenase/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Transferência Adotiva , Animais , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Separação Celular , Citometria de Fluxo , Vetores Genéticos , Humanos , Linfócitos do Interstício Tumoral/imunologia , Melanoma Experimental/terapia , Camundongos , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos T/genética , Retroviridae/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução Genética
4.
Methods Mol Biol ; 515: 137-50, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19378113

RESUMO

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.


Assuntos
Técnicas de Transferência de Genes , Vetores Genéticos/genética , HIV-1/genética , Lentivirus/genética , Linhagem Celular , Eletroforese em Gel de Poliacrilamida , Citometria de Fluxo/instrumentação , Citometria de Fluxo/métodos , Genes Reporter/genética , Humanos , Técnicas de Cultura de Tecidos
5.
Virol J ; 3: 14, 2006 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-16539700

RESUMO

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.


Assuntos
Vírus da Febre Aftosa/genética , Vetores Genéticos/genética , Lentivirus/genética , Vírus da Encefalomiocardite/genética , Terapia Genética , Proteínas de Fluorescência Verde/genética , Células HeLa , Proteínas de Homeodomínio/genética , Humanos , Células K562 , O(6)-Metilguanina-DNA Metiltransferase/genética , Ribossomos/metabolismo , Fatores de Transcrição/genética , Transgenes
6.
Hum Gene Ther ; 15(8): 758-69, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15319033

RESUMO

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.


Assuntos
Dacarbazina/análogos & derivados , Regulação Enzimológica da Expressão Gênica , Terapia Genética/métodos , Vetores Genéticos/genética , Células-Tronco Hematopoéticas/enzimologia , Lentivirus/genética , O(6)-Metilguanina-DNA Metiltransferase/genética , Antígenos CD34/metabolismo , Western Blotting , Carmustina/toxicidade , Linhagem Celular , Ensaio de Unidades Formadoras de Colônias , Primers do DNA , Dacarbazina/toxicidade , Citometria de Fluxo , Vetores Genéticos/metabolismo , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Concentração Inibidora 50 , Reação em Cadeia da Polimerase , Temozolomida , Transdução Genética , Transgenes/genética , Células Tumorais Cultivadas
7.
Cancer Res ; 73(11): 3371-80, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23633494

RESUMO

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.


Assuntos
Antígenos de Neoplasias/imunologia , Imunoterapia Adotiva/métodos , Melanoma Experimental/imunologia , Melanoma Experimental/terapia , Linfócitos T/imunologia , Animais , Modelos Animais de Doenças , Ativação Linfocitária/imunologia , Melanoma Experimental/irrigação sanguínea , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica/imunologia , Neovascularização Patológica/terapia , Receptores de Antígenos/genética , Receptores de Antígenos/imunologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/imunologia
8.
J Exp Med ; 210(6): 1125-35, 2013 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-23712432

RESUMO

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.


Assuntos
Caquexia/imunologia , Fibroblastos/imunologia , Gelatinases/imunologia , Proteínas de Membrana/imunologia , Células-Tronco Mesenquimais/imunologia , Serina Endopeptidases/imunologia , Animais , Antígenos Ly/imunologia , Antígenos de Neoplasias/imunologia , Caquexia/patologia , Linhagem Celular Tumoral , Endopeptidases , Feminino , Humanos , Masculino , Melanoma Experimental/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/imunologia , Receptores de Antígenos/imunologia , Linfócitos T/imunologia
9.
Clin Cancer Res ; 18(6): 1672-83, 2012 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-22291136

RESUMO

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.


Assuntos
Imunoterapia Adotiva/métodos , Interleucina-12/administração & dosagem , Interleucina-12/genética , Neoplasias/terapia , Linfócitos T/imunologia , Transdução Genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/imunologia , Animais , Estudos de Viabilidade , Feminino , Humanos , Interleucina-12/metabolismo , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Neoplasias/irrigação sanguínea , Receptores de Antígenos de Linfócitos T/imunologia , Proteínas Recombinantes/metabolismo
10.
J Clin Invest ; 121(12): 4746-57, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22056381

RESUMO

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.


Assuntos
Linfócitos T CD8-Positivos/transplante , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Imunoterapia Adotiva , Interleucina-12/fisiologia , Melanoma Experimental/terapia , Células Mieloides/fisiologia , Evasão Tumoral/imunologia , Microambiente Tumoral/imunologia , Animais , Apresentação de Antígeno , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/patologia , Antígenos de Neoplasias/imunologia , Células da Medula Óssea/imunologia , Células da Medula Óssea/patologia , Linfócitos T CD8-Positivos/metabolismo , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/imunologia , Inflamação/genética , Interferon gama/fisiologia , Interleucina-12/genética , Interleucina-12/metabolismo , Subpopulações de Linfócitos/imunologia , Subpopulações de Linfócitos/patologia , Macrófagos/fisiologia , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/imunologia , Quimera por Radiação , Proteínas Recombinantes de Fusão/fisiologia , Células Estromais/patologia , Células Estromais/fisiologia
11.
J Immunother ; 34(4): 343-52, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21499127

RESUMO

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.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Engenharia Genética , Imunoterapia Adotiva , Animais , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Células HEK293 , Humanos , Células Jurkat , Melanoma Experimental/imunologia , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células NIH 3T3 , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Retroviridae/genética , Transdução Genética , Antígeno gp100 de Melanoma/genética , Antígeno gp100 de Melanoma/imunologia
12.
J Immunother ; 33(7): 672-83, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20664359

RESUMO

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.


Assuntos
Imunoterapia Adotiva , Melanoma/imunologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Retroviridae/genética , Linfócitos T/efeitos dos fármacos , Transferência Adotiva , Animais , Antígenos de Diferenciação/metabolismo , Apoptose/efeitos dos fármacos , Apoptose/genética , Vacinas Anticâncer , Sobrevivência Celular/genética , Engenharia Genética , Humanos , Interferon gama/metabolismo , Interleucina-2/farmacologia , Antígeno MART-1/imunologia , Melanoma/terapia , Melanoma Experimental , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/imunologia , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/patologia
13.
Cancer Res ; 70(15): 6171-80, 2010 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-20631075

RESUMO

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.


Assuntos
Inibidores da Angiogênese/farmacologia , Imunoterapia Adotiva/métodos , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfócitos do Interstício Tumoral/imunologia , Melanoma Experimental/terapia , Neoplasias Cutâneas/terapia , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Bevacizumab , Terapia Combinada , Epitopos de Linfócito T/imunologia , Melanoma Experimental/imunologia , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neoplasias Cutâneas/imunologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/imunologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/imunologia , Irradiação Corporal Total , Antígeno gp100 de Melanoma
14.
J Clin Invest ; 120(11): 3953-68, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20978347

RESUMO

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.


Assuntos
Terapia Genética/métodos , Linfócitos/fisiologia , Neoplasias Experimentais , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Transferência Adotiva , Animais , Linhagem Celular , Feminino , Vetores Genéticos , Humanos , Interleucina-2/genética , Interleucina-2/imunologia , Linfócitos/citologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Linfócitos T/imunologia , Linfócitos T/fisiologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
15.
Cancer Immunol Immunother ; 56(6): 885-95, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17102977

RESUMO

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.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/imunologia , Neoplasias da Próstata/terapia , Proteínas Tirosina Fosfatases/imunologia , Vacinas de DNA/imunologia , Fosfatase Ácida , Animais , Autoantígenos/imunologia , Vacinas Anticâncer/administração & dosagem , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Vetores Genéticos/imunologia , Humanos , Masculino , Plasmídeos , Neoplasias da Próstata/imunologia , Ratos , Ratos Endogâmicos Lew , Transdução Genética , Vacinas de DNA/administração & dosagem , Vaccinia virus/genética
16.
Cancer Immunol Immunother ; 55(12): 1504-14, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16612600

RESUMO

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.


Assuntos
Antígenos CD/genética , Antígenos de Diferenciação/genética , Antígenos/imunologia , Ativação Linfocitária , Proteínas Recombinantes de Fusão/imunologia , Animais , Formação de Anticorpos , Antígenos/genética , Antígenos CD/química , Antígenos de Diferenciação/química , Antígeno B7-1/imunologia , Antígeno B7-2/imunologia , Antígeno CTLA-4 , Células Dendríticas/efeitos dos fármacos , Humanos , Imunoglobulina G/imunologia , Ligantes , Camundongos , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/genética , Linfócitos T/imunologia , Linfócitos T Citotóxicos/imunologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa