Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Pathol ; 263(3): 360-371, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38779852

RESUMEN

Mutations are abundantly present in tissues of healthy individuals, including the breast epithelium. Yet it remains unknown whether mutant cells directly induce lesion formation or first spread, leading to a field of mutant cells that is predisposed towards lesion formation. To study the clonal and spatial relationships between morphologically normal breast epithelium adjacent to pre-cancerous lesions, we developed a three-dimensional (3D) imaging pipeline combined with spatially resolved genomics on archival, formalin-fixed breast tissue with the non-obligate breast cancer precursor ductal carcinoma in situ (DCIS). Using this 3D image-guided characterization method, we built high-resolution spatial maps of DNA copy number aberration (CNA) profiles within the DCIS lesion and the surrounding normal mammary ducts. We show that the local heterogeneity within a DCIS lesion is limited. However, by mapping the CNA profiles back onto the 3D reconstructed ductal subtree, we find that in eight out of 16 cases the healthy epithelium adjacent to the DCIS lesions has overlapping structural variations with the CNA profile of the DCIS. Together, our study indicates that pre-malignant breast transformations frequently develop within mutant clonal fields of morphologically normal-looking ducts. © 2024 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Asunto(s)
Neoplasias de la Mama , Carcinoma Intraductal no Infiltrante , Variaciones en el Número de Copia de ADN , Mutación , Humanos , Carcinoma Intraductal no Infiltrante/genética , Carcinoma Intraductal no Infiltrante/patología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Femenino , Imagenología Tridimensional , Lesiones Precancerosas/genética , Lesiones Precancerosas/patología , Células Clonales
2.
Cell ; 186(18): 3968-3982.e15, 2023 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-37586362

RESUMEN

Ductal carcinoma in situ (DCIS) is a common precursor of invasive breast cancer. Our understanding of its genomic progression to recurrent disease remains poor, partly due to challenges associated with the genomic profiling of formalin-fixed paraffin-embedded (FFPE) materials. Here, we developed Arc-well, a high-throughput single-cell DNA-sequencing method that is compatible with FFPE materials. We validated our method by profiling 40,330 single cells from cell lines, a frozen tissue, and 27 FFPE samples from breast, lung, and prostate tumors stored for 3-31 years. Analysis of 10 patients with matched DCIS and cancers that recurred 2-16 years later show that many primary DCIS had already undergone whole-genome doubling and clonal diversification and that they shared genomic lineages with persistent subclones in the recurrences. Evolutionary analysis suggests that most DCIS cases in our cohort underwent an evolutionary bottleneck, and further identified chromosome aberrations in the persistent subclones that were associated with recurrence.


Asunto(s)
Neoplasias de la Mama , Carcinoma Ductal de Mama , Carcinoma Intraductal no Infiltrante , Femenino , Humanos , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/genética , Carcinoma Intraductal no Infiltrante/genética , Carcinoma Intraductal no Infiltrante/patología , Progresión de la Enfermedad , Genómica/métodos , Análisis de Expresión Génica de una Sola Célula , Línea Celular Tumoral
3.
Cancer Cell ; 41(5): 986-1002.e9, 2023 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-37116492

RESUMEN

Ductal carcinoma in situ (DCIS) is a non-obligate precursor of invasive breast cancer (IBC). Due to a lack of biomarkers able to distinguish high- from low-risk cases, DCIS is treated similar to early IBC even though the minority of untreated cases eventually become invasive. Here, we characterized 115 patient-derived mouse-intraductal (MIND) DCIS models reflecting the full spectrum of DCIS observed in patients. Utilizing the possibility to follow the natural progression of DCIS combined with omics and imaging data, we reveal multiple prognostic factors for high-risk DCIS including high grade, HER2 amplification, expansive 3D growth, and high burden of copy number aberrations. In addition, sequential transplantation of xenografts showed minimal phenotypic and genotypic changes over time, indicating that invasive behavior is an intrinsic phenotype of DCIS and supporting a multiclonal evolution model. Moreover, this study provides a collection of 19 distributable DCIS-MIND models spanning all molecular subtypes.


Asunto(s)
Neoplasias de la Mama , Carcinoma Intraductal no Infiltrante , Humanos , Animales , Ratones , Femenino , Carcinoma Intraductal no Infiltrante/genética , Carcinoma Intraductal no Infiltrante/patología , Bancos de Muestras Biológicas , Xenoinjertos , Biomarcadores de Tumor/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Factores de Riesgo , Progresión de la Enfermedad
4.
Nat Commun ; 14(1): 1958, 2023 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-37029129

RESUMEN

The high frequency of homologous recombination deficiency (HRD) is the main rationale of testing platinum-based chemotherapy in triple-negative breast cancer (TNBC), however, the existing methods to identify HRD are controversial and there is a medical need for predictive biomarkers. We assess the in vivo response to platinum agents in 55 patient-derived xenografts (PDX) of TNBC to identify determinants of response. The HRD status, determined from whole genome sequencing, is highly predictive of platinum response. BRCA1 promoter methylation is not associated with response, in part due to residual BRCA1 gene expression and homologous recombination proficiency in different tumours showing mono-allelic methylation. Finally, in 2 cisplatin sensitive tumours we identify mutations in XRCC3 and ORC1 genes that are functionally validated in vitro. In conclusion, our results demonstrate that the genomic HRD is predictive of platinum response in a large cohort of TNBC PDX and identify alterations in XRCC3 and ORC1 genes driving cisplatin response.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Humanos , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología , Cisplatino/farmacología , Cisplatino/uso terapéutico , Platino (Metal)/uso terapéutico , Proteína BRCA1/genética , Recombinación Homóloga , Mutación , Secuenciación Completa del Genoma , Proteína BRCA2/genética
5.
Res Sq ; 2023 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-38168198

RESUMEN

Ductal carcinoma in situ (DCIS) may progress to ipsilateral invasive breast cancer (iIBC), but often never will. Because DCIS is treated as early breast cancer, many women with harmless DCIS face overtreatment. To identify these women that may forego treatment, we hypothesized that DCIS morphometric features relate to the risk of subsequent iIBC. We developed an artificial intelligence-based DCIS morphometric analysis pipeline (AIDmap) to detect DCIS as a pathologist and measure morphological structures in hematoxylin-eosin-stained (H&E) tissue sections. These were from a case-control study of patients diagnosed with primary DCIS, treated by breast-conserving surgery without radiotherapy. We analyzed 689 WSIs of DCIS of which 226 were diagnosed with subsequent iIBC (cases) and 463 were not (controls). The distribution of 15 duct morphological measurements in each H&E was summarized in 55 morphometric variables. A ridge regression classifier with cross validation predicted 5-years-free of iIBC with an area-under the curve of 0.65 (95% CI 0.55-0.76). A morphometric signature based on the 30 variables most associated with outcome, identified lesions containing small-sized ducts, low number of cells and low DCIS/stroma area ratio. This signature was associated with lower iIBC risk in a multivariate regression model including grade, ER, HER2 and COX-2 expression (HR = 0.56; 95% CI 0.28-0.78). AIDmap has potential to identify harmless DCIS that may not need treatment.

6.
Nat Genet ; 54(6): 850-860, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35681052

RESUMEN

Ductal carcinoma in situ (DCIS) is the most common form of preinvasive breast cancer and, despite treatment, a small fraction (5-10%) of DCIS patients develop subsequent invasive disease. A fundamental biologic question is whether the invasive disease arises from tumor cells in the initial DCIS or represents new unrelated disease. To address this question, we performed genomic analyses on the initial DCIS lesion and paired invasive recurrent tumors in 95 patients together with single-cell DNA sequencing in a subset of cases. Our data show that in 75% of cases the invasive recurrence was clonally related to the initial DCIS, suggesting that tumor cells were not eliminated during the initial treatment. Surprisingly, however, 18% were clonally unrelated to the DCIS, representing new independent lineages and 7% of cases were ambiguous. This knowledge is essential for accurate risk evaluation of DCIS, treatment de-escalation strategies and the identification of predictive biomarkers.


Asunto(s)
Neoplasias de la Mama , Carcinoma Ductal de Mama , Carcinoma Intraductal no Infiltrante , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/genética , Carcinoma Intraductal no Infiltrante/genética , Carcinoma Intraductal no Infiltrante/patología , Femenino , Genómica , Humanos , Recurrencia Local de Neoplasia/genética
7.
NPJ Breast Cancer ; 8(1): 60, 2022 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-35523804

RESUMEN

When locally advanced breast cancer is treated with neoadjuvant chemotherapy, the recurrence risk is significantly higher if no complete pathologic response is achieved. Identification of the underlying resistance mechanisms is essential to select treatments with maximal efficacy and minimal toxicity. Here we employed gene expression profiles derived from 317 HER2-negative treatment-naïve breast cancer biopsies of patients who underwent neoadjuvant chemotherapy, deep whole exome, and RNA-sequencing profiles of 22 matched pre- and post-treatment tumors, and treatment outcome data to identify biomarkers of response and resistance mechanisms. Molecular profiling of treatment-naïve breast cancer samples revealed that expression levels of proliferation, immune response, and extracellular matrix (ECM) organization combined predict response to chemotherapy. Triple negative patients with high proliferation, high immune response and low ECM expression had a significantly better treatment response and survival benefit (HR 0.29, 95% CI 0.10-0.85; p = 0.02), while in ER+ patients the opposite was seen (HR 4.73, 95% CI 1.51-14.8; p = 0.008). The characterization of paired pre-and post-treatment samples revealed that aberrations of known cancer genes were either only present in the pre-treatment sample (CDKN1B) or in the post-treatment sample (TP53, APC, CTNNB1). Proliferation-associated genes were frequently down-regulated in post-treatment ER+ tumors, but not in triple negative tumors. Genes involved in ECM were upregulated in the majority of post-chemotherapy samples. Genomic and transcriptomic differences between pre- and post-chemotherapy samples are common and may reveal potential mechanisms of therapy resistance. Our results show a wide range of distinct, but related mechanisms, with a prominent role for proliferation- and ECM-related genes.

8.
J Natl Cancer Inst ; 108(11)2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27381626

RESUMEN

BACKGROUND: Although BRCA1-deficient tumors are extremely sensitive to DNA-damaging drugs and poly(ADP-ribose) polymerase (PARP) inhibitors, recurrences do occur and, consequently, resistance to therapy remains a serious clinical problem. To study the underlying mechanisms, we induced therapy resistance in patient-derived xenograft (PDX) models of BRCA1-mutated and BRCA1-methylated triple-negative breast cancer. METHODS: A cohort of 75 mice carrying BRCA1-deficient breast PDX tumors was treated with cisplatin, melphalan, nimustine, or olaparib, and treatment sensitivity was determined. In tumors that acquired therapy resistance, BRCA1 expression was investigated using quantitative real-time polymerase chain reaction and immunoblotting. Next-generation sequencing, methylation-specific multiplex ligation-dependent probe amplification (MLPA) and Target Locus Amplification (TLA)-based sequencing were used to determine mechanisms of BRCA1 re-expression in therapy-resistant tumors. RESULTS: BRCA1 protein was not detected in therapy-sensitive tumors but was found in 31 out of 42 resistant cases. Apart from previously described mechanisms involving BRCA1-intragenic deletions and loss of BRCA1 promoter hypermethylation, a novel resistance mechanism was identified in four out of seven BRCA1-methylated PDX tumors that re-expressed BRCA1 but retained BRCA1 promoter hypermethylation. In these tumors, we found de novo gene fusions that placed BRCA1 under the transcriptional control of a heterologous promoter, resulting in re-expression of BRCA1 and acquisition of therapy resistance. CONCLUSIONS: In addition to previously described clinically relevant resistance mechanisms in BRCA1-deficient tumors, we describe a novel resistance mechanism in BRCA1-methylated PDX tumors involving de novo rearrangements at the BRCA1 locus, demonstrating that BRCA1-methylated breast cancers may acquire therapy resistance via both epigenetic and genetic mechanisms.


Asunto(s)
Antineoplásicos/uso terapéutico , Resistencia a Antineoplásicos/genética , Fusión Génica , Genes BRCA1 , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética , Animales , Proteína BRCA1/deficiencia , Cisplatino/uso terapéutico , Metilación de ADN , Femenino , Expresión Génica , Humanos , Melfalán/uso terapéutico , Ratones , Mutación , Trasplante de Neoplasias , Nimustina/uso terapéutico , Ftalazinas/uso terapéutico , Piperazinas/uso terapéutico , Regiones Promotoras Genéticas
9.
Hum Mutat ; 33(1): 100-3, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22009941

RESUMEN

Mutations in the chromatin remodeling gene ARID1A have recently been identified in the majority of ovarian clear cell carcinomas (OCCCs). To determine the prevalence of mutations in other tumor types, we evaluated 759 malignant neoplasms including those of the pancreas, breast, colon, stomach, lung, prostate, brain, and blood (leukemias). We identified truncating mutations in 6% of the neoplasms studied; nontruncating somatic mutations were identified in an additional 0.4% of neoplasms. Mutations were most commonly found in gastrointestinal samples with 12 of 119 (10%) colorectal and 10 of 100 (10%) gastric neoplasms, respectively, harboring changes. More than half of the mutated colorectal and gastric cancers displayed microsatellite instability (MSI) and the mutations in these tumors were out-of-frame insertions or deletions at mononucleotide repeats. Mutations were also identified in 2-8% of tumors of the pancreas, breast, brain (medulloblastomas), prostate, and lung, and none of these tumors displayed MSI. These findings suggest that the aberrant chromatin remodeling consequent to ARID1A inactivation contributes to a variety of different types of neoplasms.


Asunto(s)
Adenocarcinoma de Células Claras/genética , Cromatina/genética , Neoplasias Colorrectales/genética , Mutación del Sistema de Lectura , Proteínas Nucleares/genética , Neoplasias Gástricas/genética , Factores de Transcripción/genética , Adenocarcinoma de Células Claras/patología , Secuencia de Bases , Mama/metabolismo , Mama/patología , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Colon/metabolismo , Colon/patología , Neoplasias Colorrectales/patología , Proteínas de Unión al ADN , Femenino , Mucosa Gástrica/metabolismo , Humanos , Masculino , Inestabilidad de Microsatélites , Repeticiones de Microsatélite , Datos de Secuencia Molecular , Páncreas/metabolismo , Páncreas/patología , Estómago/patología , Neoplasias Gástricas/patología
10.
Clin Cancer Res ; 16(2): 651-63, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-20068109

RESUMEN

PURPOSE: Several prognostic gene expression profiles have been identified in breast cancer. In spite of this progress in prognostic classification, the underlying mechanisms that drive these gene expression patterns remain unknown. Specific genomic alterations, such as copy number alterations, are an important factor in tumor development and progression and are also associated with changes in gene expression. EXPERIMENTAL DESIGN: We carried out array comparative genomic hybridization in 68 human breast carcinomas for which gene expression and clinical data were available. We used a two-class supervised algorithm, Supervised Identification of Regions of Aberration in aCGH data sets, for the identification of regions of chromosomal alterations that are associated with specific sample labeling. Using gene expression data from the same tumors, we identified genes in the altered regions for which the expression level is significantly correlated with the copy number and validated our results in public available data sets. RESULTS: Specific chromosomal aberrations are related to clinicopathologic characteristics and prognostic gene expression signatures. The previously identified poor prognosis, 70-gene expression signature is associated with the gain of 3q26.33-27.1, 8q22.1-24.21, and 17q24.3-25.1; the 70-gene good prognosis profile is associated with the loss at 16q12.1-13 and 16q22.1-24.1; basal-like tumors are associated with the gain of 6p12.3-23, 8q24.21-22, and 10p12.33-14 and losses at 4p15.31, 5q12.3-13.1, 5q33.1, 10q23.33, 12q13.13-3, 15q15.1, and 15q21.1; HER2+ breast show amplification at 17q11.1-12 and 17q21.31-23.2 (including HER2 gene). CONCLUSIONS: There is a strong correlation between the different gene expression signatures and underlying genomic changes. These findings help to establish a link between genomic changes and gene expression signatures, enabling a better understanding of the tumor biology that causes poor prognosis.


Asunto(s)
Neoplasias de la Mama/diagnóstico , Carcinoma/diagnóstico , Dosificación de Gen , Perfilación de la Expresión Génica , Técnicas de Diagnóstico Molecular/métodos , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de la Mama/genética , Carcinoma/genética , Hibridación Genómica Comparativa , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Polimorfismo Genético/fisiología , Pronóstico
11.
Proc Natl Acad Sci U S A ; 106(37): 15855-60, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19717424

RESUMEN

We previously identified a SNF1/AMPK-related protein kinase, Hunk, from a mammary tumor arising in an MMTV-neu transgenic mouse. The function of this kinase is unknown. Using targeted deletion in mice, we now demonstrate that Hunk is required for the metastasis of c-myc-induced mammary tumors, but is dispensable for normal development. Reconstitution experiments revealed that Hunk is sufficient to restore the metastatic potential of Hunk-deficient tumor cells, as well as defects in migration and invasion, and does so in a manner that requires its kinase activity. Consistent with a role for this kinase in the progression of human cancers, the human homologue of Hunk is overexpressed in aggressive subsets of carcinomas of the ovary, colon, and breast. In addition, a murine gene expression signature that distinguishes Hunk-wild type from Hunk-deficient mammary tumors predicts clinical outcome in women with breast cancer in a manner consistent with the pro-metastatic function of Hunk in mice. These findings identify a direct role for Hunk kinase activity in metastasis and establish an in vivo function for this kinase.


Asunto(s)
Neoplasias de la Mama/enzimología , Neoplasias de la Mama/secundario , Neoplasias Mamarias Experimentales/enzimología , Neoplasias Mamarias Experimentales/secundario , Proteínas Quinasas/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Animales , Neoplasias de la Mama/genética , Movimiento Celular/genética , Movimiento Celular/fisiología , Supervivencia sin Enfermedad , Femenino , Expresión Génica , Humanos , Neoplasias Mamarias Experimentales/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Invasividad Neoplásica/genética , Invasividad Neoplásica/fisiopatología , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/fisiopatología , Pronóstico , Proteínas Quinasas/deficiencia , Proteínas Quinasas/genética , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética
12.
Breast Cancer Res ; 9(5): R68, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17922920

RESUMEN

INTRODUCTION: Before any new methodology can be introduced into the routine diagnostic setting it must be technically validated against the established standards. To this end, a ring study involving five international pathology laboratories was initiated to validate chromogenic in situ hybridisation (CISH) against fluorescence in situ hybridisation (FISH) and immunohistochemistry (IHC) as a test for assessing human epidermal growth factor receptor 2 (HER2) status in breast cancer. METHODS: Each laboratory performed CISH, FISH and IHC on its own samples. Unstained sections from each case were also sent to another participating laboratory for blinded retesting by CISH ('outside CISH'). RESULTS: A total of 211 invasive breast carcinoma cases were tested. In 76 cases with high amplification (HER2/CEP17 ratio >4.0) by FISH, 73 cases (96%) scored positive (scores >or= 6) by 'outside CISH'. For FISH-negative cases (HER2/CEP17 ratio <2.0), 94 of 100 cases (94%) had CISH scores indicating no amplification (score or= 6. CISH intra-laboratory concordance with IHC was 92% for IHC-negative cases (IHC 0/1+) and 91% for IHC 3+ cases. Among IHC 2+ cases, CISH was 100% concordant with samples showing high amplification by FISH, and 94% concordant with FISH-negative samples. CONCLUSION: These results show that CISH inter- and intra-laboratory concordance to FISH and IHC is very high, even in equivocal IHC 2+ cases. Therefore, we conclude that CISH is a methodology that is a viable alternative to FISH in the HER2 testing algorithm.


Asunto(s)
Neoplasias de la Mama/genética , Compuestos Cromogénicos , Hibridación Fluorescente in Situ , Receptor ErbB-2/genética , Neoplasias de la Mama/patología , Femenino , Amplificación de Genes , Humanos , Receptor ErbB-2/metabolismo
13.
Clin Cancer Res ; 12(19): 5705-12, 2006 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17020974

RESUMEN

PURPOSE: Several risk factors for local recurrence of breast cancer after breast-conserving therapy (BCT) have been identified. The identification of additional risk factors would be very useful in guiding optimal therapy and also in improving understanding of the mechanisms underlying local recurrence. We used cDNA microarray analysis to identify gene expression profiles associated with local recurrence. EXPERIMENTAL DESIGN: Using 18K cDNA microarrays, gene expression profiles were obtained from 50 patients who underwent BCT. Of these 50 patients, 19 developed a local recurrence; the remaining 31 patients were selected as controls as they were free of local recurrence at least 11 years after treatment. For 9 of 19 patients, the local recurrence was also available for gene expression profiling. Unsupervised and supervised methods of classification were used to separate patients in groups corresponding to disease outcome and to study the overall gene expression pattern of primary tumors and their recurrences. RESULTS: Hierarchical clustering of patients did not show any grouping reflecting local recurrence status. Supervised analysis revealed no significant set of genes that was able to distinguish recurring tumors from nonrecurring tumors. Paired-data analysis of primary tumors and local recurrences showed a remarkable similarity in gene expression profile between primary tumors and their recurrences. CONCLUSIONS: No significant differences in gene expression between primary breast cancer tumors in patients with or without local recurrence after BCT were identified. Furthermore, analyses of primary tumors and local recurrences show a preservation of the overall gene expression pattern in the local recurrence, even after radiotherapy.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias de la Mama/genética , Perfilación de la Expresión Génica , Mastectomía Segmentaria , Recurrencia Local de Neoplasia/genética , Adulto , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/cirugía , Carcinoma Ductal de Mama/genética , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/cirugía , Carcinoma Lobular/genética , Carcinoma Lobular/metabolismo , Carcinoma Lobular/cirugía , Femenino , Humanos , Persona de Mediana Edad , Recurrencia Local de Neoplasia/diagnóstico , Recurrencia Local de Neoplasia/metabolismo , Estadificación de Neoplasias , Análisis de Secuencia por Matrices de Oligonucleótidos , Pronóstico , Estudios Prospectivos , Factores de Riesgo
14.
Anticancer Res ; 24(5A): 2931-9, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15517899

RESUMEN

BACKGROUND: Multidrug resistance associated proteins MRP1, MRP2 and MRP3 confer in vitro multidrug resistance. We investigated their role in breast cancer resistance to anthracycline-based chemotherapy. MATERIALS AND METHODS: Using real-time reverse transcriptase polymerase chain reaction (RT-PCR) and immunohistochemistry (IHC), the expression of MRP1 - 3 was quantified in nine breast cancer cell lines and 30 breast carcinoma samples. RESULTS: MRP1 - 3 mRNA was detectable in all breast cancer cell lines and tumor samples. No increase of expression was detected between untreated carcinoma and post-neoadjuvant anthracycline treatment tumor samples. IHC failed to detect the proteins. MRP1 - 3 expression was not associated with tumor response to treatment or with outcome. CONCLUSION: MRP1 - 3 are expressed in breast cancer cells, but are not detected with IHC. We have found no evidence linking these proteins to clinical drug resistance in a small but well-documented series of breast cancer samples.


Asunto(s)
Antraciclinas/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Genes MDR/efectos de los fármacos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/biosíntesis , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Antraciclinas/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Neoplasias de la Mama/genética , Neoplasias de la Mama/cirugía , Línea Celular Tumoral , Ensayos Clínicos Fase II como Asunto , Ciclofosfamida/administración & dosificación , Supervivencia sin Enfermedad , Resistencia a Antineoplásicos , Epirrubicina/administración & dosificación , Fluorouracilo/administración & dosificación , Expresión Génica/efectos de los fármacos , Humanos , Inmunohistoquímica , Proteínas de Transporte de Membrana/biosíntesis , Proteínas de Transporte de Membrana/genética , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Terapia Neoadyuvante , ARN Mensajero/biosíntesis , ARN Mensajero/genética
15.
J Pathol ; 201(1): 75-82, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12950019

RESUMEN

Human carcinoma in situ of the breast already demonstrates genomic changes found in invasive lesions. However, no specific genetic alterations have previously been identified that are associated with progression from the in situ to the invasive stage. By comparative genomic hybridization (CGH) and fluorescence in situ hybridization (FISH) analysis of an invasive breast carcinoma with a large associated in situ component, high-level amplification of C-MYC was found in the invasive component only. To determine the frequency of this correlation in a panel of 188 invasive breast carcinomas, 18 additional cases with C-MYC amplification were identified. Nine of these cases had a detectable adjacent in situ component. FISH analysis demonstrated increased (>5) C-MYC signals per nucleus in seven invasive components and increased (>4) C-MYC/centromere 8 signal ratios in five of these. None of the associated in situ components demonstrated these increases. The minimal amplified region was defined at 8q24.13-8qter. C-MYC amplification was correlated with overexpression of C-MYC and two of its target genes, TERT and FBL. Thus, C-MYC amplification is the first identified genetic alteration that is associated with progression from the in situ to the invasive stage of breast carcinoma.


Asunto(s)
Neoplasias de la Mama/genética , Carcinoma in Situ/genética , Carcinoma Intraductal no Infiltrante/genética , Genes myc , Adulto , Neoplasias de la Mama/patología , Carcinoma in Situ/patología , Carcinoma Intraductal no Infiltrante/patología , Cromosomas Humanos Par 8/genética , ADN de Neoplasias/genética , Progresión de la Enfermedad , Femenino , Amplificación de Genes , Humanos , Hibridación Fluorescente in Situ , Persona de Mediana Edad , Invasividad Neoplásica , Hibridación de Ácido Nucleico , Reacción en Cadena de la Polimerasa/métodos
16.
Clin Cancer Res ; 8(4): 1068-74, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11948115

RESUMEN

PURPOSE: The breast cancer resistance protein (BCRP) is involved in in vitro multidrug resistance and was first identified in the breast cancer cell line MCF7/AdrVp. The aim of this study was to investigate the role of BCRP in resistance of breast cancer to anthracycline treatment. EXPERIMENTAL DESIGN: BCRP mRNA was determined with real-time reverse transcription-PCR and immunostaining in nine breast cancer cell lines and in samples of 25 primary breast carcinomas and 27 patients who received preoperative anthracycline-based therapy. Tumor response to treatment and patient survival were recorded. RESULTS: In cell lines, only MCF7 and BT20 had BCRP mRNA levels coinciding with membrane-bound immunostaining. In clinical samples, BCRP expression varied widely (range, 0.01-0.86). With immunohistochemistry, BCRP was detected in vessels and normal breast epithelium but not in tumor cells. There was no difference in BCRP expression between anthracycline-naïve and treated tumor samples. BCRP expression was not associated with decreased response or survival. CONCLUSIONS: There is no indication that elevated BCRP expression in breast carcinomas confers resistance to anthracyclines. Expression was not detectable with immunohistochemistry.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Neoplasias de la Mama/genética , Proteínas de Neoplasias , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/metabolismo , Antibióticos Antineoplásicos/uso terapéutico , Northern Blotting , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Epirrubicina/uso terapéutico , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de Supervivencia , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...