Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Oncoimmunology ; 11(1): 2008110, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35141051

RESUMEN

Carcinoembryonic antigen-related cell adhesion molecule 6 (CEACAM6), a cell surface receptor, is expressed on normal epithelial tissue and highly expressed in cancers of high unmet medical need, such as non-small cell lung, pancreatic, and colorectal cancer. CEACAM receptors undergo homo- and heterophilic interactions thereby regulating normal tissue homeostasis and angiogenesis, and in cancer, tumor invasion and metastasis. CEACAM6 expression on malignant plasma cells inhibits antitumor activity of T cells, and we hypothesize a similar function on epithelial cancer cells. The interactions between CEACAM6 and its suggested partner CEACAM1 on T cells were studied. A humanized CEACAM6-blocking antibody, BAY 1834942, was developed and characterized for its immunomodulating effects in co-culture experiments with T cells and solid cancer cells and in comparison to antibodies targeting the immune checkpoints programmed cell death protein 1 (PD-1), programmed death-ligand 1 (PD-L1), and T cell immunoglobulin mucin-3 (TIM-3). The immunosuppressive activity of CEACAM6 was mediated by binding to CEACAM1 expressed by activated tumor-specific T cells. BAY 1834942 increased cytokine secretion by T cells and T cell-mediated killing of cancer cells. The in vitro efficacy of BAY 1834942 correlated with the degree of CEACAM6 expression on cancer cells, suggesting potential in guiding patient selection. BAY 1834942 was equally or more efficacious compared to blockade of PD-L1, and at least an additive efficacy was observed in combination with anti-PD-1 or anti-TIM-3 antibodies, suggesting an efficacy independent of the PD-1/PD-L1 axis. In summary, CEACAM6 blockade by BAY 1834942 reactivates the antitumor response of T cells. This warrants clinical evaluation.


Asunto(s)
Antígenos CD , Neoplasias , Receptor de Muerte Celular Programada 1 , Antígenos CD/inmunología , Antígeno B7-H1/inmunología , Moléculas de Adhesión Celular/inmunología , Proteínas Ligadas a GPI/inmunología , Humanos , Neoplasias/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Linfocitos T
3.
Gastric Cancer ; 25(3): 598-608, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35050442

RESUMEN

BACKGROUND: In the phase 3 GRID trial, regorafenib improved progression-free survival (PFS) independent of KIT mutations in exons 9 and 11. In this retrospective, exploratory analysis of the GRID trial, we investigated whether a more comprehensive KIT mutation analysis could identify mutations that impact treatment outcome with regorafenib and a regorafenib-induced mutation pattern. METHODS: Archived tumor samples, collected at any time prior to enrollment in GRID, were analyzed by Sanger sequencing (n = 102) and next-generation sequencing (FoundationONE; n = 47). Plasma samples collected at baseline were analyzed by BEAMing (n = 163) and SafeSEQ (n = 96). RESULTS: In archived tumor samples, 67% (68/102) had a KIT mutation; 61% (62/102) had primary KIT mutations (exons 9 and 11) and 12% (12/102) had secondary mutations (exons 13, 14, 17, and 18). At baseline, 81% of samples (78/96) had KIT mutations by SafeSEQ, including the M541L polymorphism (sole event in 6 patients). Coexisting mutations in other oncogenes were rare, as were mutations in PDGFR, KRAS, and BRAF. Regorafenib showed PFS benefit across all primary and secondary KIT mutational subgroups examined. Available patient-matched samples taken at baseline and end of treatment (n = 41; SafeSEQ), revealed heterogeneous KIT mutational changes with no specific mutation pattern emerging upon regorafenib treatment. CONCLUSION: These data support the results of the GRID trial, and suggest that patients may benefit from regorafenib in the presence of KIT mutations and without the selection of particular mutation patterns that confer resistance. The study was not powered to address biomarker-related questions, and the results are exploratory and hypothesis-generating.


Asunto(s)
Antineoplásicos , Tumores del Estroma Gastrointestinal , Neoplasias Gástricas , Antineoplásicos/uso terapéutico , Tumores del Estroma Gastrointestinal/tratamiento farmacológico , Tumores del Estroma Gastrointestinal/genética , Tumores del Estroma Gastrointestinal/patología , Humanos , Mutación , Compuestos de Fenilurea/uso terapéutico , Proteínas Proto-Oncogénicas c-kit/genética , Piridinas , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Estudios Retrospectivos
4.
Leukemia ; 34(8): 2184-2197, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32060403

RESUMEN

Patients with relapsed/refractory diffuse large B-cell lymphoma (DLBCL) have adverse outcomes. We evaluated the efficacy and safety of the phosphatidylinositol 3-kinase inhibitor copanlisib in patients with relapsed/refractory DLBCL and assessed the relationship between efficacy and DLBCL cell of origin (COO; activated B-cell like [ABC] and germinal center B-cell like [GCB]) and other biomarkers. The primary endpoint was objective response rate (ORR) in DLBCL COO subgroups (ABC, GCB, and unclassifiable) and by CD79B mutational status (NCT02391116). Sixty-seven patients received copanlisib (ABC DLBCL, n = 19; GCB DLBCL, n = 30; unclassifiable, n = 3; missing, n = 15). The ORR was 19.4%; 31.6% and 13.3% in ABC and GCB DLBCL patients, respectively. ORR was 22.2%/20.0% for patients with/without CD79B mutations (wild type, n = 45; mutant, n = 9; missing, n = 13). Overall median progression-free survival and duration of response were 1.8 and 4.3 months, respectively. Adverse events included hypertension (40.3%), diarrhea (37.3%), and hyperglycemia (32.8%). Aberrations were detected in 338 genes, including BCL2 (53.7%) and MLL2 (53.7%). A 16-gene signature separating responders from nonresponders was identified. Copanlisib treatment demonstrated a manageable safety profile in patients with relapsed/refractory DLBCL and a numerically higher response rate in ABC vs. GCB DLBCL patients.


Asunto(s)
Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Inhibidores de las Quinasa Fosfoinosítidos-3/uso terapéutico , Pirimidinas/uso terapéutico , Quinazolinas/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Antígenos CD79/genética , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/mortalidad , Masculino , Persona de Mediana Edad , Mutación , Pirimidinas/efectos adversos , Quinazolinas/efectos adversos , Recurrencia
5.
BMC Cancer ; 16: 622, 2016 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-27515171

RESUMEN

BACKGROUND: The single hotspot mutation AKT1 [G49A:E17K] has been described in several cancers, with the highest incidence observed in breast cancer. However, its precise role in disease etiology remains unknown. METHODS: We analyzed more than 600 breast cancer tumor samples and circulating tumor DNA for AKT1 (E17K) and alterations in other cancer-associated genes using Beads, Emulsions, Amplification, and Magnetics digital polymerase chain reaction technology and targeted exome sequencing. RESULTS: Overall AKT1 (E17K) mutation prevalence was 6.3 % and not correlated with age or menopausal stage. AKT1 (E17K) mutation frequency tended to be lower in patients with grade 3 disease (1.9 %) compared with those with grade 1 (11.1 %) or grade 2 (6 %) disease. In two cohorts of patients with advanced metastatic disease, 98.0 % (n = 50) and 97.1 % (n = 35) concordance was obtained between tissue and blood samples for the AKT1 (E17K) mutation, and mutation capture rates of 66.7 % (2/3) and 85.7 % (6/7) in blood versus tissue samples were observed. Although AKT1-mutant tumor specimens were often found to harbor concurrent alterations in other driver genes, a subset of specimens harboring AKT1 (E17K) as the only known driver alteration was also identified. Initial follow-up survival data suggest that AKT1 (E17K) could be associated with increased mortality. These findings warrant additional long-term follow-up. CONCLUSIONS: The data suggest that AKT1 (E17K) is the most likely disease driver in certain breast cancer patients. Blood-based mutation detection is achievable in advanced-stage disease. These findings underpin the need for a further enhanced-precision medicine paradigm in the treatment of breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Carcinoma Ductal de Mama/genética , Mutación Missense , Proteínas Proto-Oncogénicas c-akt/genética , Neoplasias de la Mama/sangre , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/terapia , Carcinogénesis/genética , Carcinoma Ductal de Mama/sangre , Carcinoma Ductal de Mama/mortalidad , Carcinoma Ductal de Mama/terapia , Fosfatidilinositol 3-Quinasa Clase I/genética , Análisis Mutacional de ADN , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Estimación de Kaplan-Meier , Prevalencia , Modelos de Riesgos Proporcionales , Proteínas Proto-Oncogénicas c-akt/sangre
6.
J Biol Chem ; 291(21): 11252-67, 2016 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-27030009

RESUMEN

The spindle assembly checkpoint (SAC) is an essential safeguarding mechanism devised to ensure equal chromosome distribution in daughter cells upon mitosis. The proteins Bub3 and BubR1 are key components of the mitotic checkpoint complex, an essential part of the molecular machinery on which the SAC relies. In the present work we have performed a detailed functional and biochemical characterization of the interaction between human Bub3 and BubR1 in cells and in vitro Our results demonstrate that genetic knockdown of Bub3 abrogates the SAC, promotes apoptosis, and inhibits the proliferation of human cancer cells. We also show that the integrity of the human mitotic checkpoint complex depends on the specific recognition between BubR1 and Bub3, for which the BubR1 Gle2 binding sequence motif is essential. This 1:1 binding event is high affinity, enthalpy-driven and with slow dissociation kinetics. The affinity, kinetics, and thermodynamic parameters of the interaction are differentially modulated by small regions in the N and C termini of the Gle2 binding domain sequence, suggesting the existence of "hotspots" for this protein-protein interaction. Furthermore, we show that specific disruption of endogenous BubR1·Bub3 complexes in human cancer cells phenocopies the effects observed in gene targeting experiments. Our work enhances the current understanding of key members of the SAC and paves the road for the pursuit of novel targeted cancer therapies based on SAC inhibition.


Asunto(s)
Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Puntos de Control de la Fase M del Ciclo Celular/fisiología , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Huso Acromático/metabolismo , Apoptosis , Proteínas de Ciclo Celular/genética , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Cinética , Puntos de Control de la Fase M del Ciclo Celular/genética , Células MCF-7 , Modelos Moleculares , Proteínas de Unión a Poli-ADP-Ribosa , Dominios y Motivos de Interacción de Proteínas , Proteínas Serina-Treonina Quinasas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Huso Acromático/genética , Termodinámica
7.
Mol Cancer Ther ; 15(2): 287-98, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26590165

RESUMEN

Multiple myeloma is a plasma cell malignancy characterized by marked heterogeneous genomic instability including frequent genetic alterations in epigenetic enzymes. In particular, the histone methyltransferase Enhancer of Zeste Homolog 2 (EZH2) is overexpressed in multiple myeloma. EZH2 is the catalytic component of the polycomb repressive complex 2 (PRC2), a master transcriptional regulator of differentiation. EZH2 catalyzes methylation of lysine 27 on histone H3 and its deregulation in cancer has been reported to contribute to silencing of tumor suppressor genes, resulting in a more undifferentiated state, and thereby contributing to the multiple myeloma phenotype. In this study, we propose the use of EZH2 inhibitors as a new therapeutic approach for the treatment of multiple myeloma. We demonstrate that EZH2 inhibition causes a global reduction of H3K27me3 in multiple myeloma cells, promoting reexpression of EZH2-repressed tumor suppressor genes in a subset of cell lines. As a result of this transcriptional activation, multiple myeloma cells treated with EZH2 inhibitors become more adherent and less proliferative compared with untreated cells. The antitumor efficacy of EZH2 inhibitors is also confirmed in vivo in a multiple myeloma xenograft model in mice. Together, our data suggest that EZH2 inhibition may provide a new therapy for multiple myeloma treatment and a promising addition to current treatment options. Mol Cancer Ther; 15(2); 287-98. ©2015 AACR.


Asunto(s)
Inhibidores Enzimáticos/administración & dosificación , Histonas/metabolismo , Mieloma Múltiple/tratamiento farmacológico , Complejo Represivo Polycomb 2/metabolismo , Proteínas Supresoras de Tumor/genética , Animales , Línea Celular Tumoral , Proteína Potenciadora del Homólogo Zeste 2 , Inhibidores Enzimáticos/farmacología , Epigénesis Genética/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Mieloma Múltiple/enzimología , Mieloma Múltiple/genética , Complejo Represivo Polycomb 2/antagonistas & inhibidores , Microambiente Tumoral/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
8.
J Alzheimers Dis ; 44(2): 613-24, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25318543

RESUMEN

The pathogenesis of Alzheimer's disease (AD) is characterized by the aggregation of amyloid-ß (Aß) peptides leading to deposition of senile plaques and a progressive decline of cognitive functions, which currently remains the main criterion for its diagnosis. Robust biomarkers for AD do not yet exist, although changes in the cerebrospinal fluid levels of tau and Aß represent promising candidates in addition to brain imaging and genetic risk profiling. Although concentrations of soluble Aß42 correlate with symptoms of AD, less is known about the biological activities of Aß peptides which are generated from the amyloid-ß protein precursor. An unbiased DNA microarray study showed that Aß42, at sub-lethal concentrations, specifically increases expression of several genes in neuroblastoma cells, notably the insulin-like growth factor binding proteins 3 and 5 (IGFBP3/5), the transcription regulator inhibitor of DNA binding, and the transcription factor Lim only domain protein 4. Using qRT-PCR, we confirmed that mRNA levels of the identified candidate genes were exclusively increased by the potentially neurotoxic Aß42 wild-type peptide, as both the less toxic Aß40 and a non-toxic substitution peptide Aß42 G33A did not affect mRNA levels. In vivo immunohistochemistry revealed a corresponding increase in both hippocampal and cortical IGFBP5 expression in an AD mouse model. Proteomic analyses of human AD cerebrospinal fluid displayed increased in vivo concentrations of IGFBPs. IGFBPs and transcription factors, as identified here, are modulated by soluble Aß42 and may represent useful early biomarkers.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Corteza Cerebral/metabolismo , Hipocampo/metabolismo , Fragmentos de Péptidos/metabolismo , Enfermedad de Alzheimer/líquido cefalorraquídeo , Péptidos beta-Amiloides/genética , Animales , Línea Celular Tumoral , Regulación de la Expresión Génica/fisiología , Humanos , Inmunohistoquímica , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Proteína 5 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Ratones Transgénicos , Análisis por Micromatrices , Fragmentos de Péptidos/genética , Escalas de Valoración Psiquiátrica , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
9.
Cancer Med ; 4(2): 253-67, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25490861

RESUMEN

Molecular mechanisms underlying the development of resistance to platinum-based treatment in patients with ovarian cancer remain poorly understood. This is mainly due to the lack of appropriate in vivo models allowing the identification of resistance-related factors. In this study, we used human whole-genome microarrays and linear model analysis to identify potential resistance-related genes by comparing the expression profiles of the parental human ovarian cancer model A2780 and its platinum-resistant variant A2780cis before and after carboplatin treatment in vivo. Growth differentiation factor 15 (GDF15) was identified as one of five potential resistance-related genes in the A2780cis tumor model. Although A2780-bearing mice showed a strong carboplatin-induced increase of GDF15 plasma levels, the basal higher GDF15 plasma levels of A2780cis-bearing mice showed no further increase after short-term or long-term carboplatin treatment. This correlated with a decreased DNA damage response, enhanced AKT survival signaling and abrogated cell cycle arrest in the carboplatin-treated A2780cis tumors. Furthermore, knockdown of GDF15 in A2780cis cells did not alter cell proliferation but enhanced cell migration and colony size in vitro. Interestingly, in vivo knockdown of GDF15 in the A2780cis model led to a basal-enhanced tumor growth, but increased sensitivity to carboplatin treatment as compared to the control-transduced A2780cis tumors. This was associated with larger necrotic areas, a lobular tumor structure and increased p53 and p16 expression of the carboplatin-treated shGDF15-A2780cis tumors. Furthermore, shRNA-mediated GDF15 knockdown abrogated p27 expression as compared to control-transduced A2780cis tumors. In conclusion, these data show that GDF15 may contribute to carboplatin resistance by suppressing tumor growth through p27. These data show that GDF15 might serve as a novel treatment target in women with platinum-resistant ovarian cancer.


Asunto(s)
Antineoplásicos/administración & dosificación , Carboplatino/administración & dosificación , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Resistencia a Antineoplásicos , Factor 15 de Diferenciación de Crecimiento/genética , Neoplasias Ováricas/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Carboplatino/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Ratones SCID , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias Ováricas/genética , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Breast Cancer Res Treat ; 138(2): 437-56, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23483306

RESUMEN

Tumorigenesis is a multi-step process involving several consecutive genetic alterations resulting in loss of genomic stability and deregulated signal transduction pathways. To study these deregulated processes in vitro, typically established cancer cell lines derived from primary tumors, ascites, or from metastases are used. However, these cancer cell lines reflect only late stages of the tumorigenic process. To better understand the consequences of the sequential genetic alterations in an in vitro model system, we applied consecutive immortalization and transformation of primary human mammary epithelial cells (HMECs) combining shRNA-mediated knockdown of tumor suppressor genes and overexpression of oncogenes. Thereby, we developed a panel of isogenic HMEC-derived cell lines reflecting the multi-step process of tumorigenesis. The immortalized cell lines have a normal epithelial morphology and proliferate indefinitely and anchorage-dependently. In contrast, the transformed cells exhibit mesenchymal-like morphological changes and strong colony-forming activity in soft agar. SNP array analysis showed that none of the cell lines displayed gross chromosomal aberrations in 80 % of the chromosomes. However, massive changes were observed in some chromosomes of the transformed cells indicating that the transformed phenotype is characterized by chromosomal alterations. The isogenic immortalized and transformed cells described here provide a powerful tool for the in vitro validation of target genes for cancer therapy.


Asunto(s)
Células Epiteliales/fisiología , Transición Epitelial-Mesenquimal , Glándulas Mamarias Humanas/citología , Antineoplásicos/farmacología , Apoptosis , Línea Celular , Proliferación Celular , Forma de la Célula , Supervivencia Celular/efectos de los fármacos , Variaciones en el Número de Copia de ADN , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Redes Reguladoras de Genes , Humanos , Hidroxiurea/farmacología , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Polimorfismo de Nucleótido Simple , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , ARN Interferente Pequeño/genética , Transducción de Señal/efectos de los fármacos , Telomerasa/metabolismo , Transcriptoma , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
11.
Int J Colorectal Dis ; 26(7): 847-58, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21465190

RESUMEN

PURPOSE: Published multigene classifiers suggesting outcome prediction for patients with stage UICC II colon cancer have not been translated into a clinical application so far. Therefore, we aimed at validating own and published gene expression signatures employing methods which enable their reconstruction in routine diagnostic specimens. METHODS: Immunohistochemistry was applied to 68 stage UICC II colon cancers to determine the protein expression of previously published prognostic classifier genes (CDH17, LAT, CA2, EMR3, and TNFRSF11A). RNA from macrodissected tumor samples from 53 of these 68 patients was profiled on Affymetrix GeneChips (HG-U133 Plus 2.0). Prognostic signatures were generated by "nearest shrunken centroids" with cross-validation. Previously published gene signatures were applied to our data set using "global tests" and leave-one-out cross-validation RESULTS: Correlation of protein expression with clinical outcome failed to separate patients with disease-free follow-up (group DF) and relapse (group R). Although gene expression profiling allowed the identification of differentially expressed genes ("DF" vs. "R"), a stable classification/prognosis signature was not discernable. Furthermore, the application of previously published gene signatures to our data was unable to predict clinical outcome (prediction rate 75.5% and 64.2%; n.s.). T-stage was the only independent prognostic factor for relapse with established clinical and pathological parameters including microsatellite status (multivariate analysis). CONCLUSIONS: Our protein and gene expression analyses do not support application of molecular classifiers for prediction of clinical outcome in current routine diagnostic as a basis for patient-orientated therapy in stage UICC II colon cancer. Further studies are needed to develop prognosis signatures applicable in patient care.


Asunto(s)
Neoplasias del Colon/genética , Neoplasias del Colon/terapia , Perfilación de la Expresión Génica , Anciano , Neoplasias del Colon/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/patología , Estadificación de Neoplasias , Resultado del Tratamiento
12.
BMC Genomics ; 11: 676, 2010 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-21118496

RESUMEN

BACKGROUND: Treatment of non-small cell lung cancer with novel targeted therapies is a major unmet clinical need. Alternative splicing is a mechanism which generates diverse protein products and is of functional relevance in cancer. RESULTS: In this study, a genome-wide analysis of the alteration of splicing patterns between lung cancer and normal lung tissue was performed. We generated an exon array data set derived from matched pairs of lung cancer and normal lung tissue including both the adenocarcinoma and the squamous cell carcinoma subtypes. An enhanced workflow was developed to reliably detect differential splicing in an exon array data set. In total, 330 genes were found to be differentially spliced in non-small cell lung cancer compared to normal lung tissue. Microarray findings were validated with independent laboratory methods for CLSTN1, FN1, KIAA1217, MYO18A, NCOR2, NUMB, SLK, SYNE2, TPM1, (in total, 10 events) and ADD3, which was analysed in depth. We achieved a high validation rate of 69%. Evidence was found that the activity of FOX2, the splicing factor shown to cause cancer-specific splicing patterns in breast and ovarian cancer, is not altered at the transcript level in several cancer types including lung cancer. CONCLUSIONS: This study demonstrates how alternatively spliced genes can reliably be identified in a cancer data set. Our findings underline that key processes of cancer progression in NSCLC are affected by alternative splicing, which can be exploited in the search for novel targeted therapies.


Asunto(s)
Empalme Alternativo/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Sondas de ADN/metabolismo , Exones/genética , Genes Relacionados con las Neoplasias/genética , Neoplasias Pulmonares/genética , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Bases de Datos Genéticas , Reacciones Falso Positivas , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reproducibilidad de los Resultados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...