Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros

Bases de datos
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
Mol Med ; 18: 565-76, 2012 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-22354215

RESUMEN

Pharmacological targeting of individual ErbB receptors elicits antitumor activity, but is frequently compromised by resistance leading to therapeutic failure. Here, we describe an immunotherapeutic approach that exploits prevalent and fundamental mechanisms by which aberrant upregulation of the ErbB network drives tumorigenesis. A chimeric antigen receptor named T1E28z was engineered, in which the promiscuous ErbB ligand, T1E, is fused to a CD28 + CD3ζ endodomain. Using a panel of ErbB-engineered 32D hematopoietic cells, we found that human T1E28z⁺ T cells are selectively activated by all ErbB1-based homodimers and heterodimers and by the potently mitogenic ErbB2/3 heterodimer. Owing to this flexible targeting capability, recognition and destruction of several tumor cell lines was achieved by T1E28⁺ T cells in vitro, comprising a wide diversity of ErbB receptor profiles and tumor origins. Furthermore, compelling antitumor activity was observed in mice bearing established xenografts, characterized either by ErbB1/2 or ErbB2/3 overexpression and representative of insidious or rapidly progressive tumor types. Together, these findings support the clinical development of a broadly applicable immunotherapeutic approach in which the propensity of solid tumors to dysregulate the extended ErbB network is targeted for therapeutic gain.


Asunto(s)
Transformación Celular Neoplásica/genética , Multimerización de Proteína/efectos de los fármacos , Receptor ErbB-2/genética , Linfocitos T/metabolismo , Animales , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/terapia , Línea Celular Tumoral , Femenino , Ingeniería Genética , Neoplasias de Cabeza y Cuello/inmunología , Neoplasias de Cabeza y Cuello/terapia , Humanos , Interleucina-4/inmunología , Interleucina-4/metabolismo , Ratones , Ratones SCID , Receptor ErbB-2/química , Receptor ErbB-2/metabolismo , Receptores de Antígenos/genética , Receptores de Antígenos/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Linfocitos T/inmunología , Transducción Genética , Ensayos Antitumor por Modelo de Xenoinjerto
2.
J Clin Immunol ; 32(5): 1059-70, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22526592

RESUMEN

PURPOSE: Chimeric antigen receptor (CAR) engineered T-cells occupy an increasing niche in cancer immunotherapy. In this context, CAR-mediated CD3ζ signaling is sufficient to elicit cytotoxicity and interferon-γ production while the additional provision of CD28-mediated signal 2 promotes T-cell proliferation and interleukin (IL)-2 production. This compartmentalisation of signaling opens the possibility that complementary CARs could be used to focus T-cell activation within the tumor microenvironment. METHODS: Here, we have tested this principle by co-expressing an ErbB2- and MUC1-specific CAR that signal using CD3ζ and CD28 respectively. Stoichiometric co-expression of transgenes was achieved using the SFG retroviral vector containing an intervening Thosea asigna peptide. RESULTS: We found that "dual-targeted" T-cells kill ErbB2(+) tumor cells efficiently and proliferate in a manner that requires co-expression of MUC1 and ErbB2 by target cells. Notably, however, IL-2 production was modest when compared to control CAR-engineered T-cells in which signaling is delivered by a fused CD28 + CD3ζ endodomain. CONCLUSIONS: These findings demonstrate the principle that dual targeting may be achieved using genetically targeted T-cells and pave the way for testing of this strategy in vivo.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Neoplasias de la Mama/inmunología , Inmunoterapia Adoptiva , Mucina-1/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/inmunología , Línea Celular Tumoral , Proliferación Celular , Células Cultivadas , Femenino , Humanos , Interferón gamma/inmunología , Interleucina-2/inmunología , Transducción de Señal/inmunología
3.
J Biol Chem ; 285(33): 25538-44, 2010 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-20562098

RESUMEN

Polyclonal T-cells can be directed against cancer using transmembrane fusion molecules known as chimeric antigen receptors (CARs). Although preclinical studies have provided encouragement, pioneering clinical trials using CAR-based immunotherapy have been disappointing. Key obstacles are the need for robust expansion ex vivo followed by sustained survival of infused T-cells in patients. To address this, we have developed a system to achieve selective proliferation of CAR(+) T-cells using IL-4, a cytokine with several pathophysiologic and therapeutic links to cancer. A chimeric cytokine receptor (4alphabeta) was engineered by fusion of the IL-4 receptor alpha (IL-4Ralpha) ectodomain to the beta(c) subunit, used by IL-2 and IL-15. Addition of IL-4 to T-cells that express 4alphabeta resulted in STAT3/STAT5/ERK phosphorylation and exponential proliferation, mimicking the actions of IL-2. Using receptor-selective IL-4 muteins, partnering of 4alphabeta with gamma(c) was implicated in signal delivery. Next, human T-cells were engineered to co-express 4alphabeta with a CAR specific for tumor-associated MUC1. These T-cells exhibited an unprecedented capacity to elicit repeated destruction of MUC1-expressing tumor cultures and expanded through several logs in vitro. Despite prolonged culture in IL-4, T-cells retained specificity for target antigen, type 1 polarity, and cytokine dependence. Similar findings were observed using CARs directed against two additional tumor-associated targets, demonstrating generality of application. Furthermore, this system allows rapid ex vivo expansion and enrichment of engineered T-cells from small blood volumes, under GMP-compliant conditions. Together, these findings provide proof of principle for the development of IL-4-enhanced T-cell immunotherapy of cancer.


Asunto(s)
Interleucina-4/farmacología , Receptores de Antígenos de Linfocitos T/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Animales , Western Blotting , Línea Celular , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Citometría de Flujo , Humanos , Interleucina-15/farmacología , Interleucina-2/farmacología , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/genética , Ratones , Fosforilación/efectos de los fármacos , Receptores de Antígenos de Linfocitos T/genética , Receptores de Interleucina-4/genética , Receptores de Interleucina-4/metabolismo , Proteínas Recombinantes de Fusión/genética , Factor de Transcripción STAT3/metabolismo
4.
J Clin Immunol ; 31(4): 710-8, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21505816

RESUMEN

Adoptive immunotherapy using chimeric antigen receptor-engrafted T cells is a promising emerging therapy for cancer. Prior to clinical testing, it is mandatory to evaluate human therapeutic cell products in meaningful in vivo pre-clinical models. Here, we describe the use of fused single-photon emission CT-CT imaging to monitor real-time migration of chimeric antigen receptor-engineered T cells in immune compromised (SCID Beige) mice. Following intravenous administration, human T cells migrate in a highly similar manner to that reported in man, but penetrate poorly into established tumors. By contrast, when delivered via intraperitoneal or subcutaneous routes, T cells remain at the site of inoculation with minimal systemic absorption-irrespective of the presence or absence of tumor. Together, these data support the validity of pre-clinical testing of human T-cell immunotherapy in SCID Beige mice. In light of their established efficacy, regional administration of engineered human T cells represents an attractive therapeutic option to minimize toxicity in the treatment of selected malignancies.


Asunto(s)
Movimiento Celular , Inmunoterapia Adoptiva/efectos adversos , Inmunoterapia Adoptiva/métodos , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T/inmunología , Traslado Adoptivo/métodos , Animales , Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/terapia , Línea Celular Tumoral , Humanos , Ratones , Ratones SCID , Mucina-1/inmunología , Proteínas Recombinantes de Fusión , Linfocitos T/citología , Linfocitos T/trasplante , Trasplante Heterólogo , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Cancers (Basel) ; 3(4): 4281-93, 2011 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-24213139

RESUMEN

Prostate cancer is the most common cancer in men, both in the USA and Europe. Although incurable, metastatic disease can often be controlled for years with anti-androgen therapy. Once the disease becomes castrate resistant, the median survival is 18 months. There is growing evidence that the immune system, and in particular cytokines, play an important role in prostate cancer immunosurveillance and progression. Here, we have undertaken a clinical investigation of the role of two closely related cytokines, IL-4 and IL-13 in prostate cancer. In the largest series studied to date, we show that serum IL-4, but not IL-13 is significantly elevated in castrate resistant, compared to androgen sensitive disease. Notably however, serum IL-4 levels are also raised in patients with benign prostatic disease. Analysis of benign and malignant prostate tissue demonstrates that the source of IL-4 is epithelial cells rather than infiltrating leukocytes. Together, our data are consistent with a dual role for IL-4 in prostate cancer development. In benign disease, our data add to the evidence that IL-4 serves a protective role. By contrast, the data support a direct role for IL-4 in the progression of prostate cancer from androgen responsive, to advanced castrate-resistant disease.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA