Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Lab Invest ; 104(5): 100341, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38280634

RESUMEN

Ki-67 is a nuclear protein associated with proliferation, and a strong potential biomarker in breast cancer, but is not routinely measured in current clinical management owing to a lack of standardization. Digital image analysis (DIA) is a promising technology that could allow high-throughput analysis and standardization. There is a dearth of data on the clinical reliability as well as intra- and interalgorithmic variability of different DIA methods. In this study, we scored and compared a set of breast cancer cases in which manually counted Ki-67 has already been demonstrated to have prognostic value (n = 278) to 5 DIA methods, namely Aperio ePathology (Lieca Biosystems), Definiens Tissue Studio (Definiens AG), Qupath, an unsupervised immunohistochemical color histogram algorithm, and a deep-learning pipeline piNET. The piNET system achieved high agreement (interclass correlation coefficient: 0.850) and correlation (R = 0.85) with the reference score. The Qupath algorithm exhibited a high degree of reproducibility among all rater instances (interclass correlation coefficient: 0.889). Although piNET performed well against absolute manual counts, none of the tested DIA methods classified common Ki-67 cutoffs with high agreement or reached the clinically relevant Cohen's κ of at least 0.8. The highest agreement achieved was a Cohen's κ statistic of 0.73 for cutoffs 20% and 25% by the piNET system. The main contributors to interalgorithmic variation and poor cutoff characterization included heterogeneous tumor biology, varying algorithm implementation, and setting assignments. It appears that image segmentation is the primary explanation for semiautomated intra-algorithmic variation, which involves significant manual intervention to correct. Automated pipelines, such as piNET, may be crucial in developing robust and reproducible unbiased DIA approaches to accurately quantify Ki-67 for clinical diagnosis in the future.


Asunto(s)
Neoplasias de la Mama , Procesamiento de Imagen Asistido por Computador , Antígeno Ki-67 , Humanos , Antígeno Ki-67/análisis , Antígeno Ki-67/metabolismo , Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/patología , Femenino , Reproducibilidad de los Resultados , Procesamiento de Imagen Asistido por Computador/métodos , Biomarcadores de Tumor/metabolismo , Biomarcadores de Tumor/análisis , Algoritmos , Inmunohistoquímica/métodos
2.
Nature ; 538(7625): 378-382, 2016 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-27732578

RESUMEN

Pancreatic cancer, a highly aggressive tumour type with uniformly poor prognosis, exemplifies the classically held view of stepwise cancer development. The current model of tumorigenesis, based on analyses of precursor lesions, termed pancreatic intraepithelial neoplasm (PanINs) lesions, makes two predictions: first, that pancreatic cancer develops through a particular sequence of genetic alterations (KRAS, followed by CDKN2A, then TP53 and SMAD4); and second, that the evolutionary trajectory of pancreatic cancer progression is gradual because each alteration is acquired independently. A shortcoming of this model is that clonally expanded precursor lesions do not always belong to the tumour lineage, indicating that the evolutionary trajectory of the tumour lineage and precursor lesions can be divergent. This prevailing model of tumorigenesis has contributed to the clinical notion that pancreatic cancer evolves slowly and presents at a late stage. However, the propensity for this disease to rapidly metastasize and the inability to improve patient outcomes, despite efforts aimed at early detection, suggest that pancreatic cancer progression is not gradual. Here, using newly developed informatics tools, we tracked changes in DNA copy number and their associated rearrangements in tumour-enriched genomes and found that pancreatic cancer tumorigenesis is neither gradual nor follows the accepted mutation order. Two-thirds of tumours harbour complex rearrangement patterns associated with mitotic errors, consistent with punctuated equilibrium as the principal evolutionary trajectory. In a subset of cases, the consequence of such errors is the simultaneous, rather than sequential, knockout of canonical preneoplastic genetic drivers that are likely to set-off invasive cancer growth. These findings challenge the current progression model of pancreatic cancer and provide insights into the mutational processes that give rise to these aggressive tumours.


Asunto(s)
Carcinogénesis/genética , Carcinogénesis/patología , Reordenamiento Génico/genética , Genoma Humano/genética , Modelos Biológicos , Mutagénesis/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Carcinoma in Situ/genética , Cromotripsis , Variaciones en el Número de Copia de ADN/genética , Progresión de la Enfermedad , Evolución Molecular , Femenino , Genes Relacionados con las Neoplasias/genética , Humanos , Masculino , Mitosis/genética , Mutación/genética , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/patología , Poliploidía , Lesiones Precancerosas/genética
4.
Blood ; 112(3): 760-9, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18502826

RESUMEN

D-cyclins are regulators of cell division that act in a complex with cyclin-dependent kinases to commit cells to a program of DNA replication. D-cyclins are overexpressed in many tumors, including multiple myeloma and leukemia, and contribute to disease progression and chemoresistance. To better understand the role and impact of D-cyclins in hematologic malignancies, we conducted a high throughput screen for inhibitors of the cyclin D2 promoter and identified the drug cyproheptadine. In myeloma and leukemia cells, cyproheptadine decreased expression of cyclins D1, D2, and D3 and arrested these cells in the G(0)/G(1) phase. After D-cyclin suppression, cyproheptadine induced apoptosis in myeloma and leukemia cell lines and primary patient samples preferentially over normal hematopoietic cells. In mouse models of myeloma and leukemia, cyproheptadine inhibited tumor growth without significant toxicity. Cyproheptadine-induced apoptosis was preceded by activation of the mitochondrial pathway of caspase activation and was independent of the drug's known activity as an H1 histamine and serotonin receptor antagonist. Thus, cyproheptadine represents a lead for a novel therapeutic agent for the treatment of malignancy. Because the drug is well tolerated and already approved in multiple countries for clinical use as an antihistamine and appetite stimulant, it could be moved directly into clinical trials for cancer.


Asunto(s)
Ciclinas/genética , Ciproheptadina/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Leucemia Mieloide Aguda/tratamiento farmacológico , Mieloma Múltiple/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Línea Celular Tumoral , Ciclina D1/genética , Ciclina D2 , Ciclina D3 , Ciproheptadina/uso terapéutico , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Leucemia Mieloide Aguda/patología , Ratones , Mieloma Múltiple/patología
5.
Clin Cancer Res ; 26(8): 1997-2010, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-31964786

RESUMEN

PURPOSE: The molecular drivers of antitumor immunity in pancreatic ductal adenocarcinoma (PDAC) are poorly understood, posing a major obstacle for the identification of patients potentially amenable for immune-checkpoint blockade or other novel strategies. Here, we explore the association of chemokine expression with effector T-cell infiltration in PDAC. EXPERIMENTAL DESIGN: Discovery cohorts comprised 113 primary resected PDAC and 107 PDAC liver metastases. Validation cohorts comprised 182 PDAC from The Cancer Genome Atlas and 92 PDACs from the Australian International Cancer Genome Consortium. We explored associations between immune cell counts by immunohistochemistry, chemokine expression, and transcriptional hallmarks of antitumor immunity by RNA sequencing (RNA-seq), and mutational burden by whole-genome sequencing. RESULTS: Among all known human chemokines, a coregulated set of four (CCL4, CCL5, CXCL9, and CXCL10) was strongly associated with CD8+ T-cell infiltration (P < 0.001). Expression of this "4-chemokine signature" positively correlated with transcriptional metrics of T-cell activation (ZAP70, ITK, and IL2RB), cytolytic activity (GZMA and PRF1), and immunosuppression (PDL1, PD1, CTLA4, TIM3, TIGIT, LAG3, FASLG, and IDO1). Furthermore, the 4-chemokine signature marked tumors with increased T-cell activation scores (MHC I presentation, T-cell/APC costimulation) and elevated expression of innate immune sensing pathways involved in T-cell priming (STING and NLRP3 inflammasome pathways, BATF3-driven dendritic cells). Importantly, expression of this 4-chemokine signature was consistently indicative of a T-cell-inflamed phenotype across primary PDAC and PDAC liver metastases. CONCLUSIONS: A conserved 4-chemokine signature marks resectable and metastatic PDAC tumors with an active antitumor phenotype. This could have implications for the appropriate selection of PDAC patients in immunotherapy trials.


Asunto(s)
Biomarcadores de Tumor/genética , Linfocitos T CD8-positivos/inmunología , Quimiocina CCL4/genética , Quimiocina CCL5/genética , Quimiocina CXCL10/genética , Quimiocina CXCL9/genética , Neoplasias Hepáticas/secundario , Neoplasias Pancreáticas/patología , Biomarcadores de Tumor/inmunología , Quimiocina CCL4/inmunología , Quimiocina CCL5/inmunología , Quimiocina CXCL10/inmunología , Quimiocina CXCL9/inmunología , Estudios de Cohortes , Biología Computacional/métodos , Bases de Datos Genéticas/estadística & datos numéricos , Humanos , Proteínas de Punto de Control Inmunitario/genética , Inmunoterapia/métodos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/inmunología , Mutación , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/inmunología , RNA-Seq/métodos
7.
Nat Genet ; 52(2): 231-240, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31932696

RESUMEN

Pancreatic adenocarcinoma presents as a spectrum of a highly aggressive disease in patients. The basis of this disease heterogeneity has proved difficult to resolve due to poor tumor cellularity and extensive genomic instability. To address this, a dataset of whole genomes and transcriptomes was generated from purified epithelium of primary and metastatic tumors. Transcriptome analysis demonstrated that molecular subtypes are a product of a gene expression continuum driven by a mixture of intratumoral subpopulations, which was confirmed by single-cell analysis. Integrated whole-genome analysis uncovered that molecular subtypes are linked to specific copy number aberrations in genes such as mutant KRAS and GATA6. By mapping tumor genetic histories, tetraploidization emerged as a key mutational process behind these events. Taken together, these data support the premise that the constellation of genomic aberrations in the tumor gives rise to the molecular subtype, and that disease heterogeneity is due to ongoing genomic instability during progression.


Asunto(s)
Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/mortalidad , Estudios de Cohortes , Femenino , Factor de Transcripción GATA6/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Inestabilidad Genómica , Humanos , Masculino , Persona de Mediana Edad , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/mortalidad , Fenotipo , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteína Smad4/genética
8.
Cancer Cell ; 35(2): 267-282.e7, 2019 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-30686769

RESUMEN

We integrated clinical, genomic, and transcriptomic data from 224 primaries and 95 metastases from 289 patients to characterize progression of pancreatic ductal adenocarcinoma (PDAC). Driver gene alterations and mutational and expression-based signatures were preserved, with truncations, inversions, and translocations most conserved. Cell cycle progression (CCP) increased with sequential inactivation of tumor suppressors, yet remained higher in metastases, perhaps driven by cell cycle regulatory gene variants. Half of the cases were hypoxic by expression markers, overlapping with molecular subtypes. Paired tumor heterogeneity showed cancer cell migration by Halstedian progression. Multiple PDACs arising synchronously and metachronously in the same pancreas were actually intra-parenchymal metastases, not independent primary tumors. Established clinical co-variates dominated survival analyses, although CCP and hypoxia may inform clinical practice.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinoma Ductal Pancreático/genética , Ciclo Celular/genética , Movimiento Celular/genética , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Hepáticas/genética , Mutación , Neoplasias Pancreáticas/genética , Transcripción Genética , Animales , Biomarcadores de Tumor/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/secundario , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Predisposición Genética a la Enfermedad , Humanos , Israel , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/secundario , Ratones , Invasividad Neoplásica , América del Norte , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Fenotipo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcriptoma , Hipoxia Tumoral
9.
Clin Cancer Res ; 13(2 Pt 1): 621-9, 2007 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-17255285

RESUMEN

PURPOSE: The aim of this study is to investigate the antimyeloma activity of a novel Bcl-2 family inhibitor, ABT-737, in preclinical treatment of multiple myeloma. EXPERIMENTAL DESIGN: The antimyeloma activity of ABT-737 was evaluated in cultured myeloma cell lines and patient myeloma samples, and in a xenograft mouse myeloma model. Drug combination therapy using ABT-737 with other commonly used myeloma drugs was also investigated. RESULTS: MY5 and JJN3 cell lines exhibited the most sensitivity to ABT-737 with an EC(50) of 0.2 and 0.5 micromol/L, respectively, with increased cell apoptosis and elevated activated caspase-3. We identified two distinct groups of myeloma patient samples that were either sensitive or resistant to the drug. Four of 15 patient bone marrow samples (27%) were highly sensitive to ABT-737 at doses of 0.25 and 0.5 micromol/L, which eliminated 80% to 90% of myeloma cells as a result of cellular apoptosis 3 days after drug treatment. ABT-737 showed a synergistic effect when combined with dexamethasone or melphalan in inducing myeloma cell death. Furthermore, the dexamethasone-resistant MM1(Dex)R myeloma cell line was highly sensitive to 0.2 micromol/L ABT-737. As determined by colony assay, little or no detectable toxicity to patient hematologic progenitor cells was observed at 1 micromol/L ABT-737. ABT-737 dose dependently suppressed tumor growth in a xenograft MY5 mouse model. CONCLUSIONS: These studies show substantial antimyeloma activity of ABT-737 as a single agent or in combination with dexamethasone or melphalan and suggest a rationale for future clinical trials.


Asunto(s)
Compuestos de Bifenilo/farmacología , Dexametasona/farmacología , Sinergismo Farmacológico , Melfalán/farmacología , Mieloma Múltiple/tratamiento farmacológico , Nitrofenoles/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Sulfonamidas/farmacología , Animales , Protocolos de Quimioterapia Combinada Antineoplásica , Apoptosis , Línea Celular Tumoral , Supervivencia Celular , Humanos , Interleucina-6/metabolismo , Ratones , Trasplante de Neoplasias , Piperazinas/farmacología , Células del Estroma/metabolismo , Células Tumorales Cultivadas
10.
Mol Ther ; 15(3): 618-27, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17228315

RESUMEN

Hematopoietic cell transplantation can impact lysosomal storage disorders (LSDs) and will be enhanced by gene therapy. Transduced cells in LSDs often secrete the therapeutic hydrolase, which can be used by bystander cells. However, toxicity associated with myeloablative transplant preparative regimens limits many applications of this approach in gene therapy. We hypothesized that reduced-intensity (RI) conditioning regimens would allow stable engraftment of therapeutically transduced cells and allow correction of Fabry disease. We transplanted transduced cells into Fabry mice receiving eight different clinically relevant chemotherapy- and/or radiotherapy-based RI conditioning regimens generating modest and transient lymphoid/myeloid cell depletion. Two comprehensive transplantation Protocols were performed. Firstly, transplantation of 0.38 x 10(6) gene-modified stem/progenitor cells was nominally effective; none of the RI regimens led to stable alpha-galactosidase A (alpha-gal A) correction. Secondly, transduced cells were preselected for functional transgene expression and transplanted at a higher dose (0.72 x 10(6) cells). Each RI regimen yielded engraftment of functional transgene-positive cells through 180 days along with increased plasma alpha-gal A activity. Importantly, the RI regimens mediated broad organ enzyme correction and were not associated with immune responses against alpha-gal A. RI conditioning thus has an important role in gene therapy for LSDs; a variety of regimens can be effective in this context.


Asunto(s)
Trasplante de Médula Ósea/métodos , Enfermedad de Fabry/terapia , Acondicionamiento Pretrasplante/métodos , Animales , Biomarcadores , Enfermedad de Fabry/genética , Enfermedad de Fabry/inmunología , Enfermedad de Fabry/cirugía , Expresión Génica , Terapia Genética , Humanos , Subunidad alfa del Receptor de Interleucina-2/genética , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Masculino , Ratones , Bazo/metabolismo
11.
Clin Cancer Res ; 24(6): 1344-1354, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29288237

RESUMEN

Purpose: To perform real-time whole genome sequencing (WGS) and RNA sequencing (RNASeq) of advanced pancreatic ductal adenocarcinoma (PDAC) to identify predictive mutational and transcriptional features for better treatment selection.Experimental Design: Patients with advanced PDAC were prospectively recruited prior to first-line combination chemotherapy. Fresh tumor tissue was acquired by image-guided percutaneous core biopsy for WGS and RNASeq. Laser capture microdissection was performed for all cases. Primary endpoint was feasibility to report WGS results prior to first disease assessment CT scan at 8 weeks. The main secondary endpoint was discovery of patient subsets with predictive mutational and transcriptional signatures.Results: Sixty-three patients underwent a tumor biopsy between December 2015 and June 2017. WGS and RNASeq were successful in 62 (98%) and 60 (95%), respectively. Genomic results were reported at a median of 35 days (range, 19-52 days) from biopsy, meeting the primary feasibility endpoint. Objective responses to first-line chemotherapy were significantly better in patients with the classical PDAC RNA subtype compared with those with the basal-like subtype (P = 0.004). The best progression-free survival was observed in those with classical subtype treated with m-FOLFIRINOX. GATA6 expression in tumor measured by RNA in situ hybridization was found to be a robust surrogate biomarker for differentiating classical and basal-like PDAC subtypes. Potentially actionable genetic alterations were found in 30% of patients.Conclusions: Prospective genomic profiling of advanced PDAC is feasible, and our early data indicate that chemotherapy response differs among patients with different genomic/transcriptomic subtypes. Clin Cancer Res; 24(6); 1344-54. ©2017 AACR.


Asunto(s)
Genómica , Neoplasias Pancreáticas/genética , Medicina de Precisión , Adulto , Anciano , Biomarcadores de Tumor , Ensayos Clínicos como Asunto , Daño del ADN , Manejo de la Enfermedad , Progresión de la Enfermedad , Femenino , Factor de Transcripción GATA6/genética , Genómica/métodos , Humanos , Masculino , Persona de Mediana Edad , Mutación , Metástasis de la Neoplasia , Estadificación de Neoplasias , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/terapia , Medicina de Precisión/métodos , Transcriptoma , Secuenciación del Exoma
12.
Cancer Gene Ther ; 12(6): 540-51, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15678150

RESUMEN

Gene therapy for prostate cancer may be realized through transduction of whole genes, such as PSA or PSMA, into immunotherapeutic dendritic cells (DCs). An oncoretroviral vector encoding human PSMA and a bicistronic oncoretroviral vector encoding human PSA and cell surface CD25 cDNAs were constructed. Remarkably, transfer of PSA/CD25 or PSMA cDNA during murine hematopoietic cell differentiation into DCs occurred with approximately 80% efficiency. In vitro, transduced DCs retained allostimulatory function and primed syngeneic T cells for tumor antigen-specific IFN-gamma secretion. In test experiments designed to elucidate mechanisms in vivo, syngeneic recipients of transduced DCs had increased anti-human PSA antibody titers and tumor-specific CD8(+) T cell IFN-gamma secretion with no detectable immune response to CD25. Gene-modified DC recipients had increased protection from specific tumor challenge for at least 18 weeks post-vaccination. DC vaccination also protected both male and female recipients. Gene-modified DC vaccination mediated regression of established, specific gene-expressing, TRAMP-C1 prostate cancer cell tumors. These findings indicate that antibody and cellular responses generated through PSA and PSMA gene transfer into DC yielded protective immunity, thereby providing further preclinical support for the implementation of immuno-gene therapy approaches for prostate cancer.


Asunto(s)
Antígenos de Superficie/genética , Antígenos de Superficie/inmunología , Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Terapia Genética/métodos , Glutamato Carboxipeptidasa II/genética , Glutamato Carboxipeptidasa II/inmunología , Antígeno Prostático Específico/genética , Antígeno Prostático Específico/inmunología , Neoplasias de la Próstata/prevención & control , Animales , Anticuerpos Antineoplásicos/metabolismo , Linfocitos T CD8-positivos/inmunología , ADN Complementario/genética , Células Dendríticas/metabolismo , Femenino , Vectores Genéticos/genética , Humanos , Masculino , Ratones , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/terapia , Receptores de Interleucina-2/genética , Receptores de Interleucina-2/inmunología
13.
J Invest Dermatol ; 118(6): 1080-4, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12060406

RESUMEN

T-cadherin is a unique member of the cadherin superfamily that shares the ectodomain organization with classical cadherins, but lacks both transmembrane and cytoplasmic regions and is instead anchored to the plasma membrane through a glycosyl-phosphatidylinositol (GPI) moiety. The function of T-cadherin has not been revealed yet. The special structure of T-cadherin might endow this molecule with specific intracellular targeting properties and functions that are distinct from classical cadherins. T-cadherin was originally cloned from chicken embryo brain and then was also found in mouse and human nervous and cardiovascular systems; however, T-cadherin in the keratinocytes and skin tissue is still an unknown area that remains to be explored. To test whether the unusual truncated T-cadherin is expressed in keratinocytes, we performed the reverse transcriptase-polymerase chain reaction of T-cadherin, as well as several classical cadherins (E-, P-, and N-cadherin), on the mouse keratinocyte cell line Pam212, fibroblast NIH3T3, and melanoma cell B16. The result indicated that mouse keratinocytes expressed the mRNA of truncated T-cadherin apart from classical cadherins, E-, and P-cadherin. To confirm the expression of T-cadherin in mouse keratinocytes, immunocytochemistry staining was carried out on Pam212 cells by using rabbit anti-T-cadherin antibody and rat antimouse E- and P-cadherin antibody. The result of immunofluorescence staining proved that T-cadherin was expressed in mouse keratinocytes. In order to analyze the distribution patterns of T-cadherin and classical cadherins on the keratinocytes, 3D scanning was performed by using a confocal microscope. From the Z-sections and XZ-sections, it was clearly demonstrated that T-cadherin was distributed diffusely on the whole cell surface, while E- and P-cadherin were concentrated on the cell-cell contacts. To examine the expression and the localization of T-cadherin on skin tissue, the frozen sections of the mouse back skin were immunohistochemically labeled by using anti-T-cadherin antibody. It was found that T-cadherin was intensively expressed only on the basal cell layer of the mouse skin. Apart from mouse keratinocytes and mouse skin, we further examined the expression of T-cadherin in human keratinocytes and human skin by western blot, immunocytochemistry, and immunohistochemistry staining. The same results were achieved with human samples. In this study, we found and verified that T-cadherin was expressed on the mouse and human keratinocytes and specifically localized on the basal cell layer of skin. The nature of T-cadherin function and its mechanism of localization at the basal cell layer of skin are important issues to be addressed concerning this unique member of the cadherin family and its physiologic and pathologic roles in the skin.


Asunto(s)
Cadherinas/análisis , Cadherinas/genética , Queratinocitos/química , Queratinocitos/fisiología , Células 3T3 , Animales , Expresión Génica/fisiología , Humanos , Queratinocitos/citología , Melanoma , Ratones , ARN Mensajero/análisis , Células Tumorales Cultivadas
14.
Clin Cancer Res ; 17(14): 4693-704, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21632854

RESUMEN

PURPOSE: Therapeutic strategies that target insulin-like growth factor 1 receptor (IGF-1R) hold promise in a wide variety of cancers including multiple myeloma (MM). In this study, we describe GTx-134, a novel small-molecule inhibitor of IGF-1R and insulin receptor (IR) and characterized its antitumor activity in preclinical models of MM. EXPERIMENTAL DESIGN: The activity of GTx-134 as a single agent and in combination was tested in MM cell lines and primary patient samples. Downstream effector proteins and correlation with apoptosis was evaluated. Cytotoxcity in bone marrow stroma coculture experiments was assessed. Finally, the in vivo efficacy was evaluated in a human myeloma xenograft model. RESULTS: GTx-134 inhibited the growth of 10 of 14 myeloma cell lines (<5 µmol/L) and induced apoptosis. Sensitivity to GTx-134 correlated with IGF-1R signal inhibition. Expression of MDR-1 and CD45 were associated with resistance to GTx-134. Coculture with insulin-growth factor-1 (IGF-1) or adherence to bone marrow stroma conferred modest resistance, but did not overcome GTx-134-induced cytotoxicity. GTx-134 showed in vitro synergies when combined with dexamethasone or lenalidomide. Further, GTx-134 enhanced the activity of PD173074, a fibroblast growth factor receptor 3 (FGFR3) inhibitor, against t(4;14) myeloma cells. Therapeutic efficacy of GTx-134 was shown against primary cells and xenograft tumors. Although dysregulation of glucose homeostasis was observed in GTx-134-treated mice, impairment of glucose tolerance was modest. CONCLUSIONS: These studies support the potential therapeutic efficacy of GTx-134 in MM. Further, they provide a rationale for clinical application in combination with established antimyeloma treatments and novel targeted therapies.


Asunto(s)
Antineoplásicos/farmacología , Compuestos Heterocíclicos con 3 Anillos/farmacología , Mieloma Múltiple/metabolismo , Receptor IGF Tipo 1/antagonistas & inhibidores , Sulfonas/farmacología , Animales , Antineoplásicos/química , Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Femenino , Compuestos Heterocíclicos con 3 Anillos/química , Compuestos Heterocíclicos con 3 Anillos/toxicidad , Humanos , Ratones , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Sulfonas/química , Sulfonas/toxicidad , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Blood ; 107(10): 4039-46, 2006 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-16467200

RESUMEN

The association of fibroblast growth factor receptor 3 (FGFR3) expression with t(4;14) multiple myeloma (MM) and the demonstration of the transforming potential of this receptor tyrosine kinase (RTK) make it a particularly attractive target for drug development. We report here a novel and highly specific anti-FGFR3-neutralizing antibody (PRO-001). PRO-001 binds to FGFR3 expressed on transformed cells and inhibits FGFR3 autophosphorylation and downstream signaling. The antibody inhibited the growth of FGFR3-expressing FDCP cells (IC(50) of 0.5 microg/mL) but not that of cells expressing FGFR1 or FGFR2, and potently inhibited FGFR3-dependent solid tumor growth in a mouse xenograft model. Furthermore, PRO-001 inhibited the growth of the FGFR3-expressing, human myeloma cell line, UTMC2. Inhibition of viability was still observed when cells were cocultured with stroma or in the presence of IL-6 or IGF-1. PRO-001 did not inhibit constitutive activation of K650E, G384D, and Y373C FGFR3 in myeloma cell lines and failed to inhibit the growth of these cells. Most importantly, however, PRO-001 induced cytotoxic responses in primary t(4;14)(+) MM samples with an increase in apoptotic index of 20% to 80% as determined by annexin V staining. The data demonstrate that PRO-001 is a potent and specific inhibitor of FGFR3 and deserves further study for the treatment of FGFR3-expressing myeloma.


Asunto(s)
Anticuerpos/farmacología , Cromosomas Humanos Par 14 , Cromosomas Humanos Par 4 , Mieloma Múltiple/genética , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Translocación Genética , Animales , División Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular , Citotoxicidad Inmunológica/efectos de los fármacos , Humanos , Ratones , Ratones Transgénicos , Trasplante Heterólogo
16.
Mol Carcinog ; 35(4): 173-9, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12489108

RESUMEN

T-cadherin appears to act as a tumor-suppressor factor in various cancers. Downregulation of T-cadherin is caused by a combination of allelic loss and hypermethylation of the T-cadherin promoter region and is related to cancer invasion. To elucidate the molecular mechanism of invasiveness of basal cell carcinoma of the skin, T-cadherin expression was investigated in archival pathological tissue sections made up of normal counterparts of skin and various types of basal cell carcinoma. Immunohistochemical staining showed that T-cadherin was not expressed in 38 of 51 (75%) basal cell carcinoma specimens, whereas normal counterparts of the skin appeared to express abundant T-cadherin. Loss of heterozygosity in intron 1 of the T-cadherin gene was found in four of 20 informative cases that did not express T-cadherin. Aberrant methylation of the T-cadherin promoter region also was found in six of 25 basal cell carcinomas by methylation-specific polymerase chain reaction. In contrast, no structural alternations were found in two loss of heterozygosity-positive basal cell carcinomas on sequence analysis. These findings indicated that T-cadherin expression was downregulated by a combination of allelic loss and aberrant methylation in basal cell carcinoma of the skin. Loss of T-cadherin expression might be related to the biological behavior of basal cell carcinoma. In addition, results of the present study suggested that downregulation of T-cadherin in various cancers might be related to tumor invasiveness rather than metastasis, because basal cell carcinoma of the skin principally lacks metastatic activity.


Asunto(s)
Cadherinas/genética , Carcinoma Basocelular/genética , Regulación hacia Abajo , Neoplasias Cutáneas/genética , Secuencia de Aminoácidos , Secuencia de Bases , Carcinoma Basocelular/patología , Cartilla de ADN , Humanos , Inmunohistoquímica , Pérdida de Heterocigocidad , Datos de Secuencia Molecular , Invasividad Neoplásica , Neoplasias Cutáneas/patología
17.
Am J Pathol ; 163(4): 1395-404, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14507647

RESUMEN

In the present study, we cloned and characterized a novel actin-binding molecule, designated skeletrophin, from aggregated neuroblastoma cells. The putative amino acid sequence of human skeletrophin cDNA contained a cysteine-rich zinc-finger motif which was also found in dystrophin and five ankyrin repeats. Northern blot analysis revealed that the 3.2-kb skeletrophin mRNA was expressed in normal skeletal muscle, and to a lesser extent in heart, brain, and kidney. Specific antibody was prepared to human skeletrophin peptide, and a single protein band with an approximate molecular weight of 70 kd was detected in tissue extracts by immunoblotting using the antibody. To better understand the biological properties of skeletrophin, we used a yeast two-hybrid system to screen for molecules interacting with skeletrophin and found that skeletrophin bound to actin monomer. Co-immunoprecipitation experiments also demonstrated that skeletrophin was able to bind to actin monomer. Fluorescence in situ hybridization mapped the skeletrophin gene on human chromosome 1p36.2-36.3, in which putative tumor suppressor genes for malignant melanoma have been postulated to exist. We therefore immunohistochemically stained benign nevi and malignant melanoma tissues. Notably, 23 of 25 benign nevi expressed skeletrophin in cytoplasm, but 18 of 38 cases of primary skin melanoma appeared to lack skeletrophin expression. Treatment with a demethylating agent, 5'-aza-2-deoxycytidine, restored skeletrophin expression in cultured Mewo melanoma cells. The present findings suggest that skeletrophin may be a novel actin-binding cytoskeleton-related molecule, expression of which is silenced in a considerable number of melanoma specimens.


Asunto(s)
Actinas/metabolismo , Azacitidina/análogos & derivados , Melanoma/metabolismo , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Secuencia de Aminoácidos/genética , Azacitidina/farmacología , Northern Blotting , Western Blotting , Clonación Molecular , ADN Complementario/genética , Decitabina , Regulación hacia Abajo , Humanos , Inmunohistoquímica/métodos , Hibridación Fluorescente in Situ , Datos de Secuencia Molecular , Pruebas de Precipitina , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Coloración y Etiquetado , Transfección , Células Tumorales Cultivadas , Técnicas del Sistema de Dos Híbridos , Ubiquitina-Proteína Ligasas , Levaduras
18.
Lab Invest ; 82(8): 1023-9, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12177241

RESUMEN

We previously reported that T-cadherin (CDH13, H-cadherin), a unique cadherin molecule, was expressed on the basal cell layer in normal murine and human epidermis. In the present study, T-cadherin expression in archival human skin specimens comprising a spectrum of human squamous cell neoplasia was investigated. T-cadherin expression was observed in both normal epidermal basal cells and adnexal epithelial cells of formalin-fixed and paraffin-embedded tissue sections. Western immunoblotting also revealed that mature T-cadherin protein was expressed in cultured human skin tissue equivalent. Atypical keratinocytes in 27 of 53 specimens of actinic keratosis and 23 of 30 specimens of Bowen's disease expressed T-cadherin. In contrast, T-cadherin was focally expressed in 6 of 56 invasive cutaneous squamous cell carcinomas. To explore the molecular mechanism of down-regulation of T-cadherin expression in invasive squamous cell carcinoma, loss of heterozygosity, genetic alternations, and methylation status in the 5' region of the T-cadherin gene were investigated. Loss of heterozygosity at intron 1 of the T-cadherin gene was observed in 8 of 28 informative cases of invasive squamous cell carcinoma. Although no structural genomic alternations were found by sequence analysis, aberrant promoter methylation of the T-cadherin gene was found in 12 of 28 invasive squamous cell carcinomas. T-cadherin expression was restored in cultured A431 cells, in which aberrant methylation was found by treatment with the demethylating agent 5'-aza-2-deoxycytidine. These findings suggest that a combination of deletion and aberrant methylation of the T-cadherin gene may play a role in loss of gene expression in a considerable number of invasive cutaneous squamous cell carcinomas.


Asunto(s)
Cadherinas/biosíntesis , Carcinoma de Células Escamosas/metabolismo , Neoplasias Cutáneas/metabolismo , Cadherinas/genética , Carcinoma de Células Escamosas/genética , Metilación de ADN , Regulación hacia Abajo , Eliminación de Gen , Humanos , Neoplasias Cutáneas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA