Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(19): e2123483119, 2022 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-35507878

RESUMEN

Immunotherapy approaches focusing on T cells have provided breakthroughs in treating solid tumors. However, there remains an opportunity to drive anticancer immune responses via other cell types, particularly myeloid cells. ATRC-101 was identified via a target-agnostic process evaluating antibodies produced by the plasmablast population of B cells in a patient with non-small cell lung cancer experiencing an antitumor immune response during treatment with checkpoint inhibitor therapy. Here, we describe the target, antitumor activity in preclinical models, and data supporting a mechanism of action of ATRC-101. Immunohistochemistry studies demonstrated tumor-selective binding of ATRC-101 to multiple nonautologous tumor tissues. In biochemical analyses, ATRC-101 appears to target an extracellular, tumor-specific ribonucleoprotein (RNP) complex. In syngeneic murine models, ATRC-101 demonstrated robust antitumor activity and evidence of immune memory following rechallenge of cured mice with fresh tumor cells. ATRC-101 increased the relative abundance of conventional dendritic cell (cDC) type 1 cells in the blood within 24 h of dosing, increased CD8+ T cells and natural killer cells in blood and tumor over time, decreased cDC type 2 cells in the blood, and decreased monocytic myeloid-derived suppressor cells in the tumor. Cellular stress, including that induced by chemotherapy, increased the amount of ATRC-101 target in tumor cells, and ATRC-101 combined with doxorubicin enhanced efficacy compared with either agent alone. Taken together, these data demonstrate that ATRC-101 drives tumor destruction in preclinical models by targeting a tumor-specific RNP complex leading to activation of innate and adaptive immune responses.


Asunto(s)
Antineoplásicos , Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Neoplasias , Inmunidad Adaptativa , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Humanos , Inmunidad Innata , Ratones , Neoplasias/patología
2.
Clin Immunol ; 187: 37-45, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29031828

RESUMEN

There is significant debate regarding whether B cells and their antibodies contribute to effective anti-cancer immune responses. Here we show that patients with metastatic but non-progressing melanoma, lung adenocarcinoma, or renal cell carcinoma exhibited increased levels of blood plasmablasts. We used a cell-barcoding technology to sequence their plasmablast antibody repertoires, revealing clonal families of affinity matured B cells that exhibit progressive class switching and persistence over time. Anti-CTLA4 and other treatments were associated with further increases in somatic hypermutation and clonal family size. Recombinant antibodies from clonal families bound non-autologous tumor tissue and cell lines, and families possessing immunoglobulin paratope sequence motifs shared across patients exhibited increased rates of binding. We identified antibodies that caused regression of, and durable immunity toward, heterologous syngeneic tumors in mice. Our findings demonstrate convergent functional anti-tumor antibody responses targeting public tumor antigens, and provide an approach to identify antibodies with diagnostic or therapeutic utility.


Asunto(s)
Antígenos de Neoplasias/inmunología , Linfocitos B/inmunología , Neoplasias/inmunología , Adenocarcinoma del Pulmón/inmunología , Adenocarcinoma del Pulmón/secundario , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos , Sitios de Unión de Anticuerpos/inmunología , Carcinoma de Células Renales/inmunología , Carcinoma de Células Renales/secundario , Progresión de la Enfermedad , Femenino , Humanos , Neoplasias Renales/inmunología , Neoplasias Renales/patología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Masculino , Melanoma/inmunología , Melanoma/secundario , Persona de Mediana Edad , Metástasis de la Neoplasia , Células Plasmáticas/inmunología , Células Precursoras de Linfocitos B , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología
3.
Oncotarget ; 7(20): 29228-44, 2016 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-27081082

RESUMEN

Isolation of prostate stem cells (PSCs) is crucial for understanding their biology during normal development and tumorigenesis. In this aim, we used a transgenic mouse model expressing GFP from the stem cell-specific s-SHIP promoter to mark putative stem cells during postnatal prostate development. Here we show that cells identified by GFP expression are present transiently during early prostate development and localize to the basal cell layer of the epithelium. These prostate GFP+ cells are a subpopulation of the Lin- CD24+ Sca-1+ CD49f+ cells and are capable of self-renewal together with enhanced growth potential in sphere-forming assay in vitro, a phenotype consistent with that of a PSC population. Transplantation assays of prostate GFP+ cells demonstrate reconstitution of prostate ducts containing both basal and luminal cells in renal grafts. Altogether, these results demonstrate that s-SHIP promoter expression is a new marker for neonatal basal prostate cells exhibiting stem cell properties that enables PSCs in situ identification and isolation via a single consistent parameter. Transcriptional profiling of these GFP+ neonatal stem cells showed an increased expression of several components of the Wnt signaling pathway. It also identified stem cell regulators with potential applications for further analyses of normal and cancer stem cells.


Asunto(s)
Células Epiteliales/citología , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas/metabolismo , Próstata/citología , Células Madre/citología , Animales , Biomarcadores , Células Epiteliales/metabolismo , Masculino , Ratones , Ratones SCID , Ratones Transgénicos , Próstata/metabolismo , Ratas , Ratas Sprague-Dawley , Células Madre/metabolismo
4.
J Clin Invest ; 123(10): 4410-22, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24018560

RESUMEN

The activating receptor NK cell group 2 member D (NKG2D) mediates antitumor immunity in experimental animal models. However, whether NKG2D ligands contribute to tumor suppression or progression clinically remains controversial. Here, we have described 2 novel lines of "humanized" bi-transgenic (bi-Tg) mice in which native human NKG2D ligand MHC class I polypeptide-related sequence B (MICB) or the engineered membrane-restricted MICB (MICB.A2) was expressed in the prostate of the transgenic adenocarcinoma of the mouse prostate (TRAMP) model of spontaneous carcinogenesis. Bi-Tg TRAMP/MICB mice exhibited a markedly increased incidence of progressed carcinomas and metastasis, whereas TRAMP/MICB.A2 mice enjoyed long-term tumor-free survival conferred by sustained NKG2D-mediated antitumor immunity. Mechanistically, we found that cancer progression in TRAMP/MICB mice was associated with loss of the peripheral NK cell pool owing to high serum levels of tumor-derived soluble MICB (sMICB). Prostate cancer patients also displayed reduction of peripheral NK cells and high sMIC levels. Our study has not only provided direct evidence in "humanized" mouse models that soluble and membrane-restricted NKG2D ligands pose opposite impacts on cancer progression, but also uncovered a mechanism of sMIC-induced impairment of NK cell antitumor immunity. Our findings suggest that the impact of soluble NKG2D ligands should be considered in NK cell-based cancer immunotherapy and that our unique mouse models should be valuable for therapy optimization.


Asunto(s)
Neoplasias Óseas/sangre , Carcinoma/sangre , Antígenos de Histocompatibilidad Clase I/sangre , Células Asesinas Naturales/fisiología , Neoplasias Hepáticas/sangre , Neoplasias Pulmonares/sangre , Neoplasias de la Próstata/sangre , Anciano , Animales , Neoplasias Óseas/inmunología , Neoplasias Óseas/secundario , Carcinoma/inmunología , Carcinoma/secundario , Línea Celular Tumoral , Proliferación Celular , Antígenos de Histocompatibilidad Clase I/fisiología , Homeostasis , Humanos , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Metástasis Linfática , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Trasplante de Neoplasias , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/patología
5.
J Clin Invest ; 123(2): 682-99, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23348745

RESUMEN

The contextual signals that regulate the expansion of prostate tumor progenitor cells are poorly defined. We found that a significant fraction of advanced human prostate cancers and castration-resistant metastases express high levels of the ß4 integrin, which binds to laminin-5. Targeted deletion of the signaling domain of ß4 inhibited prostate tumor growth and progression in response to loss of p53 and Rb function in a mouse model of prostate cancer (PB-TAg mice). Additionally, it suppressed Pten loss-driven prostate tumorigenesis in tissue recombination experiments. We traced this defect back to an inability of signaling-defective ß4 to sustain self-renewal of putative cancer stem cells in vitro and proliferation of transit-amplifying cells in vivo. Mechanistic studies indicated that mutant ß4 fails to promote transactivation of ErbB2 and c-Met in prostate tumor progenitor cells and human cancer cell lines. Pharmacological inhibition of ErbB2 and c-Met reduced the ability of prostate tumor progenitor cells to undergo self-renewal in vitro. Finally, we found that ß4 is often coexpressed with c-Met and ErbB2 in human prostate cancers and that combined pharmacological inhibition of these receptor tyrosine kinases exerts antitumor activity in a mouse xenograft model. These findings indicate that the ß4 integrin promotes prostate tumorigenesis by amplifying ErbB2 and c-Met signaling in tumor progenitor cells.


Asunto(s)
Integrina beta4/metabolismo , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Expresión Génica , Marcación de Gen , Humanos , Integrina beta4/química , Integrina beta4/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Ratones Transgénicos , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Neoplasia Intraepitelial Prostática/genética , Neoplasia Intraepitelial Prostática/metabolismo , Neoplasia Intraepitelial Prostática/patología , Neoplasias de la Próstata/genética , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Transducción de Señal
6.
J Immunol ; 189(8): 3936-46, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22984076

RESUMEN

Adoptive T cell therapy (ACT) for the treatment of established cancers is actively being pursued in clinical trials. However, poor in vivo persistence and maintenance of antitumor activity of transferred T cells remain major problems. TGF-ß is a potent immunosuppressive cytokine that is often expressed at high levels within the tumor microenvironment, potentially limiting T cell-mediated antitumor activity. In this study, we used a model of autochthonous murine prostate cancer to evaluate the effect of cell-intrinsic abrogation of TGF-ß signaling in self/tumor-specific CD8 T cells used in ACT to target the tumor in situ. We found that persistence and antitumor activity of adoptively transferred effector T cells deficient in TGF-ß signaling were significantly improved in the cancerous prostate. However, over time, despite persistence in peripheral lymphoid organs, the numbers of transferred cells in the prostate decreased and the residual prostate-infiltrating T cells were no longer functional. These findings reveal that TGF-ß negatively regulates the accumulation and effector function of transferred self/tumor-specific CD8 T cells and highlight that, when targeting a tumor Ag that is also expressed as a self-protein, additional substantive obstacles are operative within the tumor microenvironment, potentially hampering the success of ACT for solid tumors.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Epítopos de Linfocito T/inmunología , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/patología , Transducción de Señal/inmunología , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/fisiología , Traslado Adoptivo/métodos , Animales , Linfocitos T CD8-positivos/trasplante , Femenino , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Neoplasias de la Próstata/terapia , Miembro 25 de Receptores de Factores de Necrosis Tumoral/genética , Transducción de Señal/genética
7.
Int J Cancer ; 131(3): 662-72, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22275114

RESUMEN

Mutations in the androgen receptor (AR) have been detected in experimental and clinical prostate tumors. Mice with enforced prostate-specific expression of one such receptor variant, AR-E231G, invariably develop prostatic intraepithelial neoplasia by 12 weeks and metastatic prostate cancer by 52 weeks. The aim of this study was to identify genes with altered expression in the prostates of AR-E231G mice at an early stage of disease that may act as drivers of AR-mediated tumorigenesis. The gene expression profile of AR-E231G prostate tissue from 12-week-old mice was compared to an equivalent profile from mice expressing the AR-T857A receptor variant (analogous to the AR-T877A variant in LNCaP cells), which do not develop prostate tumors. One hundred and thirty-two genes were differentially expressed in AR-E231G prostates. Classification of these genes revealed enrichment for cellular pathways known to be involved in prostate cancer, including cell cycle and lipid metabolism. Suppression of two genes upregulated in the AR-E231G model, ADM and CITED1, increased cell death and reduced proliferation of human prostate cancer cells. Many genes differentially expressed in AR-E231G prostates are also deregulated in human tumors. Three of these genes, ID4, NR2F1 and PTGDS, which were expressed at consistently lower levels in clinical prostate cancer compared to nonmalignant tissues, formed a signature that predicted biochemical relapse (hazard ratio 2.2, p = 0.038). We believe that our findings support the value of this novel mouse model of prostate cancer to identify candidate therapeutic targets and/or biomarkers of human disease.


Asunto(s)
Factor de Transcripción COUP I/genética , Proteínas Inhibidoras de la Diferenciación/genética , Oxidorreductasas Intramoleculares/genética , Lipocalinas/genética , Neoplasias de la Próstata/genética , Receptores Androgénicos/genética , Adrenomedulina/genética , Animales , Proteínas Reguladoras de la Apoptosis , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Transgénicos , Mutación , Proteínas Nucleares/genética , Pronóstico , Neoplasia Intraepitelial Prostática/genética , Neoplasias de la Próstata/patología , Interferencia de ARN , ARN Interferente Pequeño , Transactivadores/genética
8.
Mol Cancer Ther ; 10(7): 1229-40, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21593385

RESUMEN

A novel TRIM family member, TRIM59 gene was characterized to be upregulated in SV40 Tag oncogene-directed transgenic and knockout mouse prostate cancer models as a signaling pathway effector. We identified two phosphorylated forms of TRIM59 (p53 and p55) and characterized them using purified TRIM59 proteins from mouse prostate cancer models at different stages with wild-type mice and NIH3T3 cells as controls. p53/p55-TRIM59 proteins possibly represent Ser/Thr and Tyr phosphorylation modifications, respectively. Quantitative measurements by ELISA showed that the p-Ser/Thr TRIM59 correlated with tumorigenesis, whereas the p-Tyr-TRIM59 protein correlated with advanced cancer of the prostate (CaP). The function of TRIM59 was elucidated using short hairpin RNA (shRNA)-mediated knockdown of the gene in human CaP cells, which caused S-phase cell-cycle arrest and cell growth retardation. A hit-and-run effect of TRIM59 shRNA knockdown was observed 24 hours posttransfection. Differential cDNA microarrray analysis was conducted, which showed that the initial and rapid knockdown occurred early in the Ras signaling pathway. To confirm the proto-oncogenic function of TRIM59 in the Ras signaling pathway, we generated a transgenic mouse model using a prostate tissue-specific gene (PSP94) to direct the upregulation of the TRIM59 gene. Restricted TRIM59 gene upregulation in the prostate revealed the full potential for inducing tumorigenesis, similar to the expression of SV40 Tag, and coincided with the upregulation of genes specific to the Ras signaling pathway and bridging genes for SV40 Tag-mediated oncogenesis. The finding of a possible novel oncogene in animal models will implicate a novel strategy for diagnosis, prognosis, and therapy for cancer.


Asunto(s)
Transformación Celular Neoplásica/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Metaloproteínas/genética , Metaloproteínas/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Animales , Antígenos Transformadores de Poliomavirus/genética , Antígenos Transformadores de Poliomavirus/metabolismo , Ciclo Celular/genética , Línea Celular Transformada , Línea Celular Tumoral , Proliferación Celular , Progresión de la Enfermedad , Técnicas de Silenciamiento del Gen , Orden Génico , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Transgénicos , Células 3T3 NIH , Neoplasias/patología , Fosforilación/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , ARN Mensajero/genética , Transducción de Señal/genética , Proteínas de Motivos Tripartitos
9.
Clin Cancer Res ; 16(22): 5390-401, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-21062931

RESUMEN

PURPOSE: Cell adhesion molecules (CADM) comprise a newly identified protein family whose functions include cell polarity maintenance and tumor suppression. CADM-1, CADM-3, and CADM-4 have been shown to act as tumor suppressor genes in multiple cancers including prostate cancer. However, CADM-2 expression has not been determined in prostate cancer. EXPERIMENTAL DESIGN: The CADM-2 gene was cloned and characterized and its expression in human prostatic cell lines and cancer specimens was analyzed by reverse transcription-PCR and an immunohistochemical tissue array, respectively. The effects of adenovirus-mediated CADM-2 expression on prostate cancer cells were also investigated. CADM-2 promoter methylation was evaluated by bisulfite sequencing and methylation-specific PCR. RESULTS: We report the initial characterization of CADM-2 isoforms: CADM-2a and CADM-2b, each with separate promoters, in human chromosome 3p12.1. Prostate cancer cell lines, LNCaP and DU145, expressed negligible CADM-2a relative to primary prostate tissue and cell lines, RWPE-1 and PPC-1, whereas expression of CADM-2b was maintained. Using immunohistochemistry, tissue array results from clinical specimens showed statistically significant decreased expression in prostate carcinoma compared with normal donor prostate, benign prostatic hyperplasia, prostatic intraepithelial neoplasia, and normal tissue adjacent to tumor (P < 0.001). Adenovirus-mediated CADM-2a expression suppressed DU145 cell proliferation in vitro and colony formation in soft agar. The decrease in CADM-2a mRNA in cancer cell lines correlated with promoter region hypermethylation as determined by bisulfite sequencing and methylation-specific PCR. Accordingly, treatment of cells with the demethylating agent 5-aza-2'-deoxycytidine alone or in combination with the histone deacetylase inhibitor trichostatin A resulted in the reactivation of CADM-2a expression. CONCLUSIONS: CADM-2a protein expression is significantly reduced in prostate cancer. Its expression is regulated in part by promoter methylation and implicates CADM-2 as a previously unrecognized tumor suppressor gene in a proportion of human prostate cancers.


Asunto(s)
Moléculas de Adhesión Celular/genética , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Próstata/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Moléculas de Adhesión Celular/biosíntesis , Proliferación Celular , Clonación Molecular , Metilación de ADN/genética , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Regiones Promotoras Genéticas/genética , Neoplasias de la Próstata/patología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Adulto Joven
10.
Mol Cell Endocrinol ; 323(2): 239-45, 2010 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-20417685

RESUMEN

The progression of prostate cancer from an organ-confined, androgen-sensitive disease to a metastatic one is associated with dysregulation of androgen receptor (AR)-regulated target genes and with a decrease in insulin-like growth factor-I receptor (IGF-IR) expression. To investigate the differential effects of wild type (wt) and mutant AR on IGF-IR levels we employed a series of isogenic prostate-derived cell lines and human xenografts. We show that basal and phosphorylated IGF-IR levels progressively decreased as prostate cancer cells became more tumorigenic and metastatic. In addition, we show that wt, but not mutant, AR along with dihydrotestosterone treatment increased IGF-IR promoter activity and endogenous IGF-IR levels. ChIP analysis show enhanced AR binding to the IGF-IR promoter in AR-overexpressing cells. Finally, wt AR-overexpressing cells display an enhanced proliferation rate. In summary, we provide evidence that activated wt AR enhances IGF-IR transcription in prostate cancer cells via a mechanism that involves AR binding to the IGF-IR promoter. AR mutations alter the ability of the mutated protein to regulate IGF-IR expression. Our results suggest that prostate cancer progression is associated with a decrease in IGF-IR expression that could be the result of impaired ability of AR to stimulate IGF-IR gene expression.


Asunto(s)
Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Próstata/patología , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Animales , Castración , Proliferación Celular , Humanos , Masculino , Ratones , Ratones SCID , Regiones Promotoras Genéticas , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Trasplante Heterólogo
11.
J Immunol ; 183(8): 4848-52, 2009 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-19801511

RESUMEN

We previously reported that naive, tumor-specific CD8(+) (TcR-I) T cells transferred into prostate tumor-bearing mice traffic to the prostate where they become tolerized. We now report that TcR-I cells suppress the proliferation of naive T cells. This suppression is mediated at least in part by secreted factors, and the suppressive activity can be blocked by Abs directed against TGF-beta. We further report that TcR-I cells must infiltrate the prostate to acquire suppressive activity. Delivery of tumor-specific CD4(+) T cells prevents the conversion of TcR-I cells into suppressor cells. Taken together, our findings may have critical implications for sustaining T cell responsiveness during immunotherapy, as the development of suppressor cells in the tumor microenvironment may eliminate the potency of T cells primed in the periphery or delivered during adoptive immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias de la Próstata/inmunología , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta/inmunología , Animales , Anticuerpos/inmunología , Linfocitos T CD8-positivos/trasplante , Tolerancia Inmunológica , Inmunoterapia Adoptiva , Linfocitos Infiltrantes de Tumor/trasplante , Masculino , Ratones , Ratones Transgénicos , Neoplasias de la Próstata/terapia , Linfocitos T Reguladores/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
12.
Mol Imaging ; 8(4): 209-20, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19728975

RESUMEN

Molecular imaging techniques used to detect the initiation of disease have the potential to provide the best opportunity for early treatment and cure. This report aimed at testing the possibility that Flk1+ (vascular endothelial growth factor receptor 2), a crucial angiogenesis factor of most tumor cells, could be a molecular targeted imaging marker for the diagnosis and prognosis of cancer. We performed Flk1-targeted microbubble-enhanced ultrasound (US) imaging of prostate cancer in a genetically engineered mouse model with normal-appearing intact US (negative) prostates and with three different tumor sizes (small, medium, and large). Higher levels of Flk1+ molecular signals were identified in the intact US (negative) prostate group by US-targeted imaging and immunohistochemical analysis. The increase in Flk1+ expression occurred prior to the angiogenesis switch-on phase and vascularity peak. After this peak accumulation stage of Flk1+ molecules, lower and stabilized levels of Flk1+ signals were maintained together with tumor growth from small, to medium, to large size. In a longitudinal observation in a subset (n = 5) of mice with established tumors, elevated Flk1+ signals were observed in tissues surrounding the prostate cancer, for example, the ipsilateral boundary zones between two developing tumor lobes, new tumor blood vessel recruits, the urethra border, and the pelvic node basin. The potential of Flk1-targeted US imaging as a predictive imaging tool was confirmed by correlation studies of three-dimensional US B-mode imaging, gross pathology, and histology analyses. The results of the application in a genetically engineered mouse model with prostate cancer of molecular Flk1-targeted US imaging support the contention that Flk1 can be used as a molecular imaging marker for small tumors undetectable by microimaging and as a molecular diagnostic and prognosis marker for tumor metastasis and progression.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Medios de Contraste/administración & dosificación , Neoplasias de la Próstata/diagnóstico por imagen , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Biomarcadores de Tumor/análisis , Medios de Contraste/análisis , Medios de Contraste/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Detección Precoz del Cáncer , Procesamiento de Imagen Asistido por Computador/métodos , Imagenología Tridimensional/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Metástasis de la Neoplasia , Neovascularización Patológica/diagnóstico por imagen , Pronóstico , Neoplasias de la Próstata/irrigación sanguínea , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Ultrasonografía , Receptor 2 de Factores de Crecimiento Endotelial Vascular/análisis
13.
Cancer Res ; 69(15): 6256-64, 2009 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-19622771

RESUMEN

We reported previously that tumor-specific CD8(+) T cells (TcR-I) become tolerant in the transgenic adenocarcinoma of the mouse prostate (TRAMP) model. In this study, we show that CD4(+) TcR transgenic (TcR-II) T cells transferred into TRAMP mice became activated in lymph nodes, trafficked to the prostate, and initially functioned as T(H)1 cells. Although a single cotransfer of TcR-II cells delayed TcR-I cell tolerization, repeated transfer of TcR-II cells was required to prevent TcR-I cell tolerization and significantly slowed progression of TRAMP prostate tumors. After transfer of TcR-II cells, dendritic cells within the tumor expressed higher levels of costimulatory molecules and displayed an enhanced ability to stimulate proliferation of naive T cells. Blockade of CD40-CD40L interactions during TcR-II transfer resulted in a profound reduction in dendritic cell stimulatory capacity and a partial loss of TcR-I effector functions and tumor immunity. These data show that sustained provision of activated tumor-specific CD4(+) T cells alters the immunosuppressive tumor microenvironment, ultimately leading to the control of tumor growth. These findings will assist in the design of more effective immunotherapeutic approaches for cancer.


Asunto(s)
Adenocarcinoma/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Neoplasias de la Próstata/inmunología , Adenocarcinoma/terapia , Secuencia de Aminoácidos , Animales , Células Presentadoras de Antígenos/inmunología , Antígenos CD40/antagonistas & inhibidores , Antígenos CD40/inmunología , Ligando de CD40/antagonistas & inhibidores , Ligando de CD40/inmunología , Tolerancia Inmunológica , Inmunoterapia Adoptiva , Ganglios Linfáticos/inmunología , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Datos de Secuencia Molecular , Neoplasias de la Próstata/terapia
14.
Eur J Cancer Prev ; 18(4): 331-41, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19444125

RESUMEN

Scutellaria barbata (SB) has been used in Chinese medicine to treat various cancers. This study investigated the effects of SB on prostate cancer prevention. Male TRansgenic Adenocarcinoma Mouse Prostate (TRAMP) mice at 9 weeks were randomly divided into four groups and given daily oral feedings of 8, 16, or 32 mg SB or sterilized water. In the control group, palpable tumors initially appeared at 19 weeks of age and were present in all mice by 32 weeks. In the respective treatment groups, palpable tumor development was delayed by 2, 4, and 7 weeks and 22, 30, and 38% of the mice were free of palpable tumors. Palpable tumor development in 50% of the mice occurred at 25 weeks in the placebo group, 29 weeks in the low-dose and mid-dose treatment groups, and 33 weeks in the high-dose group (log rank, P = 0.0211). Histological assessment further showed that the SB treatment (32 mg) delayed prostate tumor progression in the TRAMP mice. Caspase 3 activation was observed in SB-treated prostate tissue. Positive TUNEL assay results were detected in TRAMP-C1 and LNCaP cells treated with SB (1 mg/ml), which indicated significant apoptosis induction. Western blotting of SB-treated LNCaP cells also showed elevated expression of Bax, p53, Akt, and JNK. In-vivo data showed that the SB delayed tumor development in TRAMP mice. Complementary in-vitro data indicated that SB might exert this function by upregulating the apoptotic pathway and downregulating the survival pathway in prostate cancer cells, thus suggesting that SB possesses chemopreventive properties and has potential for cancer treatment.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Medicamentos Herbarios Chinos/uso terapéutico , Neoplasias de la Próstata/tratamiento farmacológico , Scutellaria , Adenocarcinoma/patología , Animales , Apoptosis/fisiología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Medicamentos Herbarios Chinos/farmacología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias de la Próstata/patología
15.
Cancer Res ; 68(9): 3495-504, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18451178

RESUMEN

Insulin-like growth factor-I (IGF-I) is a polypeptide hormone that can influence growth, differentiation, and survival of cells expressing the cognate type 1 receptor (IGF-IR). To better understand cell autonomous IGF-IR signaling in the epithelial compartment of the prostate gland, we generated a conditional (Cre/loxP) prostate-specific IGF-IR knockout mouse model. In contrast to epidemiologic studies that established a correlation between elevated serum IGF-I and the risk of developing prostate cancer, we show that abrogation of IGF-IR expression in the dorsal and lateral prostate could activate extracellular signal-regulated kinase 1/2 signaling and cause cell autonomous proliferation and hyperplasia. Moreover, persistent loss of IGF-IR expression in dorsal and ventral lobes induced p53-regulated apoptosis and cellular senescence rescue programs, predicting that titration of IGF-IR signaling might facilitate growth of tumors with compromised p53 activity. Therefore, we crossed the mice carrying the prostate-specific IGF-IR knockout alleles into the transgenic adenocarcinoma of the mouse prostate model that is driven, in part, by T antigen-mediated functional inactivation of p53. Consistent with our prediction, prostate epithelial-specific deletion of IGF-IR accelerated the emergence of aggressive prostate cancer when p53 activity was compromised. Collectively, these data support a critical role for IGF-IR signaling in prostate tumorigenesis and identify an important IGF-IR-dependent growth control mechanism.


Asunto(s)
Adenocarcinoma/genética , Epitelio/metabolismo , Eliminación de Gen , Próstata/metabolismo , Neoplasias de la Próstata/genética , Receptor IGF Tipo 1/genética , Adenocarcinoma/metabolismo , Factores de Edad , Animales , Apoptosis/genética , Proliferación Celular , Senescencia Celular/genética , Regulación Neoplásica de la Expresión Génica , Marcación de Gen , Integrasas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Especificidad de Órganos/genética , Hiperplasia Prostática/genética , Hiperplasia Prostática/patología , Neoplasias de la Próstata/metabolismo , Receptor IGF Tipo 1/metabolismo
16.
Immunity ; 28(4): 571-80, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18394936

RESUMEN

Ligands for the NKG2D stimulatory receptor are frequently upregulated on tumor lines, rendering them sensitive to natural killer (NK) cells, but the role of NKG2D in tumor surveillance has not been addressed in spontaneous cancer models. Here, we provided the first characterization of NKG2D-deficient mice, including evidence that NKG2D was not necessary for NK cell development but was critical for immunosurveillance of epithelial and lymphoid malignancies in two transgenic models of de novo tumorigenesis. In both models, we detected NKG2D ligands on the tumor cell surface ex vivo, providing needed evidence for ligand expression by primary tumors. In a prostate cancer model, aggressive tumors arising in NKG2D-deficient mice expressed higher amounts of NKG2D ligands than did similar tumors in wild-type mice, suggesting an NKG2D-dependent immunoediting of tumors in this model. These findings provide important genetic evidence for surveillance of primary tumors by an NK receptor.


Asunto(s)
Adenocarcinoma/inmunología , Fibrosarcoma/inmunología , Síndromes de Inmunodeficiencia/inmunología , Vigilancia Inmunológica , Linfoma de Células B/inmunología , Neoplasias de la Próstata/inmunología , Receptores Inmunológicos/deficiencia , Receptores Inmunológicos/genética , Adenocarcinoma/genética , Animales , Benzo(a)Antracenos/toxicidad , Modelos Animales de Enfermedad , Femenino , Fibrosarcoma/inducido químicamente , Fibrosarcoma/genética , Síndromes de Inmunodeficiencia/genética , Vigilancia Inmunológica/genética , Linfoma de Células B/genética , Masculino , Metilcolantreno , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Subfamilia K de Receptores Similares a Lectina de Células NK , Neoplasias de la Próstata/genética , Receptores Inmunológicos/fisiología , Receptores de Células Asesinas Naturales
17.
Prostate ; 68(10): 1105-15, 2008 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-18437689

RESUMEN

BACKGROUND: Poor penetration and uneven distribution of doxorubicin in tumors limits the efficacy of this drug in patients with prostate cancer (PC). Aim of the study was to investigate whether pre-treatment with NGR-TNF, a tumor necrosis factor-alpha derivative able to target tumor vessels and alter vessel permeability, increases the penetration and the efficacy of doxorubicin in pre-clinical models of PC. METHODS: Wild type C57BL/6 mice bearing androgen-independent TRAMP-C1 PC and transgenic adenocarcinoma of the mouse prostate (TRAMP) mice, which spontaneously develop PC and metastasis, were treated with repeated cycles of doxorubicin, administered either alone or following NGR-TNF. Tumor growth and drug uptake by cancer cells was evaluated. RESULTS: Doxorubicin as a single agent blocked the growth of TRAMP-C1 cells in vitro but not in vivo. Pre-treatment of mice bearing subcutaneous TRAMP-C1 tumors with NGR-TNF favored doxorubicin penetration into the tumor mass, and in both TRAMP-C1 and TRAMP models significantly delayed tumor growth without increasing drug-related toxicity. CONCLUSIONS: Pre-treatment with NGR-TNF significantly expanded the therapeutic index of doxorubicin in mouse models of hormone-dependent and -independent PC.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Doxorrubicina/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Factor de Necrosis Tumoral alfa/farmacología , Andrógenos/metabolismo , Animales , División Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Quimioterapia Combinada , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/patología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Receptores Androgénicos/metabolismo
18.
Proc Natl Acad Sci U S A ; 105(9): 3509-14, 2008 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-18303116

RESUMEN

Discovery of immunologically relevant antigens in prostate cancer forms the basis for developing more potent active immunotherapy. We report here a strategy using the transgenic adenocarcinoma of mouse prostate (TRAMP) model, which allows for the functional identification of immunogenic prostate tumor antigens with relevance for human immunotherapy. Using a combination of active tumor vaccination in the presence of CTL-associated antigen 4 (CTLA-4) in vivo blockade, we elicited tumor-specific T cells used to expression clone the first T cell-defined TRAMP tumor antigen, called Spas-1 (stimulator of prostatic adenocarcinoma specific T cells-1). Spas-1 expression was increased in advanced primary TRAMP tumors. We show that the immunodominant SPAS-1 epitope SNC9-H(8) arose from a point mutation in one allele of the gene in TRAMP tumor cells, and that immunization with dendritic cells pulsed with SNC9-H(8) peptide resulted in protection against TRAMP-C2 tumor challenge. In humans, the Spas-1 ortholog SH3GLB2 has been reported to be overexpressed in prostate cancer metastases. Additionally, we identified a nonmutated HLA-A2-binding epitope in the human ortholog SH3GLB2, which primed T cells from healthy HLA-A2(+) individuals in vitro. Importantly, in vitro-primed T cells also recognized naturally processed and presented SH3GLB2. Our findings demonstrate that our in vivo CTLA-4 blockade-based T cell expression cloning can identify immunogenic cancer antigens with potential relevance for human immunotherapy.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/aislamiento & purificación , Antígenos de Neoplasias/aislamiento & purificación , Clonación Molecular/métodos , Neoplasias de la Próstata/química , Proteínas Adaptadoras Transductoras de Señales/inmunología , Animales , Antígenos CD , Antígenos de Diferenciación , Antígenos de Neoplasias/inmunología , Secuencia de Bases , Antígeno CTLA-4 , Vacunas contra el Cáncer/inmunología , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Humanos , Epítopos Inmunodominantes/genética , Masculino , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Mutación Puntual , Neoplasias de la Próstata/inmunología , Especificidad del Receptor de Antígeno de Linfocitos T , Linfocitos T/inmunología
19.
Prostate ; 68(6): 629-39, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18213636

RESUMEN

Prostate cancer continues to be a major cause of morbidity and mortality in men around the world. The field of prostate cancer research continues to be hindered by the lack of relevant preclinical models to study tumorigenesis and to further development of effective prevention and therapeutic strategies. The Prostate Cancer Foundation held a Prostate Cancer Models Working Group (PCMWG) Summit on August 6th and 7th, 2007 to address these issues. The PCMWG reviewed the state of prostate cancer preclinical models and identified the current limitations of cell line, xenograft and genetically engineered mouse models that have hampered the transition of scientific findings from these models to human clinical trials. In addition the PCMWG identified administrative issues that inhibit the exchange of models and impede greater interactions between academic centers and these centers with industry. The PCMWG identified potential solutions for discovery bottlenecks that include: (1) insufficient number of models with insufficient molecular and biologic diversity to reflect human cancer, (2) a lack of understanding of the molecular events that define tumorigenesis, (3) a lack of tools for studying tumor-host interactions, (4) difficulty in accessing model systems across institutions, and (5) addressing why preclinical studies appear not to be predictive of human clinical trials. It should be possible to apply the knowledge gained molecular and epigenetic studies to develop new cell lines and models that mimic progressive and fatal prostate cancer and ultimately improve interventions.


Asunto(s)
Modelos Animales de Enfermedad , Neoplasias Experimentales/patología , Neoplasias de la Próstata/patología , Animales , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Masculino , Ratones , Ratones Transgénicos , Neoplasias Experimentales/etiología , Neoplasias Experimentales/prevención & control , Neoplasias de la Próstata/etiología , Neoplasias de la Próstata/prevención & control
20.
Development ; 135(4): 775-84, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18184727

RESUMEN

The fibroblast growth factor (FGF) regulates a broad spectrum of biological activities by activation of transmembrane FGF receptor (FGFR) tyrosine kinases and their coupled intracellular signaling pathways. FGF receptor substrate 2alpha (FRS2alpha) is an FGFR interactive adaptor protein that links multiple signaling pathways to the activated FGFR kinase. We previously showed that FGFR2 in the prostate epithelium is important for branching morphogenesis and for the acquisition of the androgen responsiveness. Here we show in mice that FRS2alpha is uniformly expressed in the epithelial cells of developing prostates, whereas it is expressed only in basal cells of the mature prostate epithelium. However, expression of FRS2alpha was apparent in luminal epithelial cells of regenerating prostates and prostate tumors. To investigate FRS2alpha function in the prostate, the Frs2alpha alleles were ablated specifically in the prostatic epithelial precursor cells during prostate development. Similar to the ablation of Fgfr2, ablation of Frs2alpha disrupted MAP kinase activation, impaired prostatic ductal branching morphogenesis and compromised cell proliferation. Unlike the Fgfr2 ablation, disrupting Frs2alpha had no effect on the response of the prostate to androgens. More importantly, ablation of Frs2alpha inhibited prostatic tumorigenesis induced by oncogenic viral proteins. The results suggest that FRS2alpha-mediated signals in prostate epithelial cells promote branching morphogenesis and proliferation, and that aberrant activation of FRS2-linked pathways might promote tumorigenesis. Thus, the prostate-specific Frs2alpha(cn) mice provide a useful animal model for scrutinizing the molecular mechanisms underlying prostatic development and tumorigenesis.


Asunto(s)
Células Epiteliales/metabolismo , Proteínas de la Membrana/metabolismo , Organogénesis , Próstata/crecimiento & desarrollo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Regeneración , Alelos , Andrógenos/farmacología , Animales , Proliferación Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/enzimología , Células Epiteliales/patología , Epitelio/metabolismo , Epitelio/patología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Masculino , Proteínas de la Membrana/genética , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Especificidad de Órganos/efectos de los fármacos , Próstata/efectos de los fármacos , Próstata/enzimología , Próstata/patología , Regeneración/efectos de los fármacos , Maduración Sexual/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...