Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Ann Oncol ; 33(11): 1186-1199, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35988656

RESUMEN

BACKGROUND: Germline variant evaluation in precision oncology opens new paths toward the identification of patients with genetic tumor risk syndromes and the exploration of therapeutic relevance. Here, we present the results of germline variant analysis and their clinical implications in a precision oncology study for patients with predominantly rare cancers. PATIENTS AND METHODS: Matched tumor and control genome/exome and RNA sequencing was carried out for 1485 patients with rare cancers (79%) and/or young adults (77% younger than 51 years) in the National Center for Tumor Diseases/German Cancer Consortium (NCT/DKTK) Molecularly Aided Stratification for Tumor Eradication Research (MASTER) trial, a German multicenter, prospective, observational precision oncology study. Clinical and therapeutic relevance of prospective pathogenic germline variant (PGV) evaluation was analyzed and compared to other precision oncology studies. RESULTS: Ten percent of patients (n = 157) harbored PGVs in 35 genes associated with autosomal dominant cancer predisposition, whereof up to 75% were unknown before study participation. Another 5% of patients (n = 75) were heterozygous carriers for recessive genetic tumor risk syndromes. Particularly, high PGV yields were found in patients with gastrointestinal stromal tumors (GISTs) (28%, n = 11/40), and more specifically in wild-type GISTs (50%, n = 10/20), leiomyosarcomas (21%, n = 19/89), and hepatopancreaticobiliary cancers (16%, n = 16/97). Forty-five percent of PGVs (n = 100/221) supported treatment recommendations, and its implementation led to a clinical benefit in 40% of patients (n = 10/25). A comparison of different precision oncology studies revealed variable PGV yields and considerable differences in germline variant analysis workflows. We therefore propose a detailed workflow for germline variant evaluation. CONCLUSIONS: Genetic germline testing in patients with rare cancers can identify the very first patient in a hereditary cancer family and can lead to clinical benefit in a broad range of entities. Its routine implementation in precision oncology accompanied by the harmonization of germline variant evaluation workflows will increase clinical benefit and boost research.


Asunto(s)
Neoplasias , Adulto Joven , Humanos , Neoplasias/diagnóstico , Neoplasias/genética , Neoplasias/terapia , Mutación de Línea Germinal , Predisposición Genética a la Enfermedad , Estudios Prospectivos , Síndrome , Medicina de Precisión/métodos
2.
ESMO Open ; 6(5): 100253, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34487971

RESUMEN

BACKGROUND: Targeted therapies have improved survival and quality of life for patients with non-small-cell lung cancer with actionable driver mutations. However, epidermal growth factor receptor (EGFR) and human epidermal growth factor receptor 2 gene (HER2, also known as ERBB2) exon 20 insertions (Ex20mut) are characterized by a poor response to currently approved tyrosine kinase inhibitors and immunotherapies. The underlying immune biology is not well understood. MATERIALS AND METHODS: We carried out messenger RNA expression profiling of lung adenocarcinomas (ADCs) with ERBB2 (n = 19) and EGFR exon 20-insertion mutations (n = 13) and compared these to tumors with classical EGFR mutations (n = 40, affecting EGFR exons 18, 19 or 21) and EGFR/ERBB2 mutation-negative lung ADC (EGFR/ERBB2wt, n = 26) focusing on immunologically relevant transcripts. Tumor-infiltrating immune cells were estimated from gene expression profiles. RESULTS: Cytotoxic cells were significantly lower in EGFR-mutated tumors regardless of the affected exon, while Th1 cells were significantly lower in EGFR-Ex20mut compared to EGFR/ERBB2wt tumors. We assessed the differentially expressed genes of ERBB2-Ex20mut and EGFR-Ex20mut tumors compared to EGFR-Ex18/19/21mut and EGFR/ERBB2wt tumors. Of these, the genes GUSB, HDAC11, IFNGR2, PUM1, RASGRF1 and RBL2 were up-regulated, while a lower expression of CBLC, GBP1, GBP2, GBP4 and MYC was observed in all three comparison groups. The omnibus test revealed 185 significantly (FDR = 5%) differentially expressed genes and we found these four most significant gene expression changes in the study cohort: VHL and JAK1 were overexpressed in ERBB2-Ex20mut and EGFR-Ex20mut tumors compared to both EGFR-Ex18/19/21mut and EGFR/ERBB2wt tumors. RIPK1 and STK11IP showed the highest expression in ERBB2-Ex20mut tumors. CONCLUSIONS: Targeted gene expression profiling is a promising tool to read out the characteristics of the tumor microenvironment from routine diagnostic lung cancer biopsies. Significant immune reactivity and specific immunosuppressive characteristics in ERBB2-Ex20mut and EGFR-Ex20mut lung ADC with at least some degree of immune infiltration support further clinical evaluation of immune-modulators as partners of immune checkpoint inhibitors in such tumors.


Asunto(s)
Adenocarcinoma del Pulmón , Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Adenocarcinoma del Pulmón/genética , Receptores ErbB/genética , Histona Desacetilasas , Humanos , Neoplasias Pulmonares/genética , Calidad de Vida , Proteínas de Unión al ARN , Receptor ErbB-2/genética , Microambiente Tumoral/genética
3.
Lung Cancer ; 148: 105-112, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32871455

RESUMEN

OBJECTIVE: Panel-based next-generation sequencing (NGS) is increasingly used for the diagnosis of EGFR-mutated non-small-cell lung cancer (NSCLC) and could improve risk assessment in combination with clinical parameters. MATERIALS AND METHODS: To this end, we retrospectively analyzed the outcome of 400 tyrosine kinase inhibitor (TKI)-treated EGFR+ NSCLC patients with validation of results in an independent cohort (n = 130). RESULTS: EGFR alterations other than exon 19 deletions (non-del19), TP53 co-mutations, and brain metastases at baseline showed independent associations of similar strengths with progression-free (PFS hazard ratios [HR] 2.1-2.3) and overall survival (OS HR 1.7-2.2), in combination defining patient subgroups with distinct outcome (EGFR+NSCLC risk Score, "ENS", p < 0.001). Co-mutations beyond TP53 were rarely detected by our multigene panel (<5%) and not associated with clinical endpoints. Smoking did not affect outcome independently, but was associated with non-del19 EGFR mutations (p < 0.05) and comorbidities (p < 0.001). Laboratory parameters, like the blood lymphocyte-to-neutrophil ratio and serum LDH, correlated with the metastatic pattern (p < 0.01), but had no independent prognostic value. Reduced ECOG performance status (PS) was associated with comorbidities (p < 0.05) and shorter OS (p < 0.05), but preserved TKI efficacy. Non-adenocarcinoma histology was also associated with shorter OS (p < 0.05), but rare (2-3 %). The ECOG PS and non-adenocarcinoma histology could not be validated in our independent cohort, and did not increase the range of prognostication alongside the ENS. CONCLUSIONS: EGFR variant, TP53 status and brain metastases predict TKI efficacy and survival in EGFR+ NSCLC irrespective of other currently available parameters ("ENS"). Together, they constitute a practical and reproducible approach for risk stratification of newly diagnosed metastatic EGFR+ NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Carcinoma de Pulmón de Células no Pequeñas/diagnóstico , Carcinoma de Pulmón de Células no Pequeñas/genética , Receptores ErbB/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/genética , Mutación , Inhibidores de Proteínas Quinasas/uso terapéutico , Estudios Retrospectivos , Medición de Riesgo
4.
Pathologe ; 40(3): 235-242, 2019 May.
Artículo en Alemán | MEDLINE | ID: mdl-31089797

RESUMEN

Precision oncology is obtaining a central role in the therapy of malignant diseases. The indication for targeted therapy is based on the identification of molecular targets for which next-generation sequencing (NGS) is commonly used nowadays. All approved predictive biomarkers and molecular targets, including gene fusions and copy number alterations, can be identified depending on panel design and method applied. Some clinical scenarios, however, may require more holistic genomic approaches, such as whole-genome/whole-exome and transcriptome analysis, which must be embedded in a clinical trial. Here, key aspects and applications of each method are summarized and discussed.


Asunto(s)
Genómica , Neoplasias , Biomarcadores de Tumor , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Mutación , Neoplasias/genética , Neoplasias/patología , Medicina de Precisión
5.
Int J Cancer ; 144(1): 190-199, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30255938

RESUMEN

Tyrosine kinase inhibitors (TKI) have improved prognosis in metastatic anaplastic lymphoma kinase (ALK)-driven lung adenocarcinoma, but patient outcomes vary widely. We retrospectively analyzed the clinical course of all cases with assessable baseline TP53 status and/or ALK fusion variant treated at our institutions (n = 102). TP53 mutations were present in 17/87 (20%) and the echinoderm microtubule-associated protein-like 4 (EML4)-ALK variant 3 (V3) in 41/92 (45%) patients. The number of metastatic sites at diagnosis was affected more by the presence of V3 than by TP53 mutations, and highest with both factors (mean 5.3, p < 0.001). Under treatment with ALK TKI, progression-free survival (PFS) was shorter with either TP53 mutations or V3, while double positive cases appeared to have an even higher risk (hazard ratio [HR] = 2.9, p = 0.015). The negative effect of V3 on PFS of TKI-treated patients was strong already in the first line (HR = 2.5, p = 0.037) and decreased subsequently, whereas a trend for PFS impairment under first-line TKI by TP53 mutations became stronger and statistically significant only when considering all treatment lines together. Overall survival was impaired more by TP53 mutations (HR = 4.9, p = 0.003) than by V3 (HR = 2.4, p = 0.018), while patients with TP53 mutated V3-driven tumors carried the highest risk of death (HR = 9.1, p = 0.02). Thus, TP53 mutations and V3 are independently associated with enhanced metastatic spread, shorter TKI responses and inferior overall survival in ALK+ lung adenocarcinoma. Both markers could assist selection of cases for more aggressive management and guide development of novel therapeutic strategies. In combination, they define a patient subset with very poor outcome.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Neoplasias Pulmonares/genética , Mutación , Proteínas de Fusión Oncogénica/genética , Proteína p53 Supresora de Tumor/genética , Anciano , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Supervivencia sin Enfermedad , Femenino , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Masculino , Persona de Mediana Edad , Proteínas de Fusión Oncogénica/metabolismo , Evaluación de Resultado en la Atención de Salud , Inhibidores de Proteínas Quinasas/uso terapéutico , Estudios Retrospectivos , Proteína p53 Supresora de Tumor/metabolismo
6.
Oncogenesis ; 6(5): e337, 2017 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-28504689

RESUMEN

Multiple myeloma (MM) is a plasma cell malignancy that is still considered to be incurable in most cases. A dominant mutation cluster has been identified in RAS/RAF genes, emphasizing the potential significance of RAS/RAF/MEK/ERK signaling as a therapeutic target. As yet, however, the clinical relevance of this finding is unclear as clinical responses to MEK inhibition in RAS-mutant MM have been mixed. We therefore assessed RAS/RAF mutation status and MEK/ERK pathway activation by both targeted sequencing and phospho-ERK immunohistochemistry in 180 tissue biopsies from 103 patients with newly diagnosed MM (NDMM) and 77 patients with relapsed/refractory MM (rrMM). We found a significant enrichment of RAS/BRAF mutations in rrMM compared to NDMM (P=0.011), which was mainly due to an increase of NRAS mutations (P=0.010). As expected, BRAF mutations were significantly associated with activated downstream signaling. However, only KRAS and not NRAS mutations were associated with pathway activation compared to RAS/BRAFwt (P=0.030). More specifically, only KRASG12D and BRAFV600E were consistently associated with ERK activation (P<0.001 and P=0.006, respectively). Taken together, these results suggest the need for a more specific stratification strategy consisting of both confirmation of protein-level pathway activation as well as detailed RAS/RAF mutation status to allow for a more precise and more effective application of targeted therapies, for example, with BRAF/MEK inhibitors in MM.

8.
Ann Oncol ; 28(1): 142-148, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27687311

RESUMEN

Background: Sinonasal carcinomas (SNCs) comprise various rare tumor types that are characterized by marked histologic diversity and largely unknown molecular profiles, yet share an overall poor prognosis owing to an aggressive clinical course and frequent late-stage diagnosis. The lack of effective systemic therapies for locally advanced or metastatic SNC poses a major challenge to therapeutic decision making for individual patients. We here aimed to identify actionable genetic alterations in a patient with metastatic SNC whose tumor, despite all diagnostic efforts, could not be assigned to any known SNC category and was refractory to multimodal therapy. Patients and methods: We used whole-exome and transcriptome sequencing to identify a KIT exon 11 mutation (c.1733_1735del, p.D579del) as potentially druggable target in this patient and carried out cancer hotspot panel sequencing to detect secondary resistance-conferring mutations in KIT. Furthermore, as a step towards clinical exploitation of the recently described signatures of mutational processes in cancer genomes, we established and applied a novel bioinformatics algorithm that enables supervised analysis of the mutational catalogs of individual tumors. Results: Molecularly guided treatment with imatinib in analogy to the management of gastrointestinal stromal tumor (GIST) resulted in a dramatic and durable response with remission of nearly all tumor manifestations, indicating a dominant driver function of mutant KIT in this tumor. KIT dependency was further validated by a secondary KIT exon 17 mutation (c.2459_2462delATTCinsG, p.D820_S821delinsG) that was detected upon tumor progression after 10 months of imatinib treatment and provided a rationale for salvage therapy with regorafenib, which has activity against KIT exon 11/17 mutant GIST. Conclusions: These observations highlight the potential of unbiased genomic profiling for uncovering the vulnerabilities of individual malignancies, particularly in rare and unclassifiable tumors, and underscore that KIT exon 11 mutations represent tractable therapeutic targets across different histologies.


Asunto(s)
Carcinoma/diagnóstico , Carcinoma/genética , Neoplasias de los Senos Paranasales/diagnóstico , Neoplasias de los Senos Paranasales/genética , Proteínas Proto-Oncogénicas c-kit/genética , Adulto , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/análisis , Carcinoma/tratamiento farmacológico , Análisis Mutacional de ADN , Perfilación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Mesilato de Imatinib/uso terapéutico , Inmunohistoquímica , Masculino , Mutación , Neoplasias de los Senos Paranasales/tratamiento farmacológico
9.
Pathologe ; 36(2): 154-63, 2015 Mar.
Artículo en Alemán | MEDLINE | ID: mdl-25820445

RESUMEN

Lung cancer is the prototypical tumor entity for the development of new diagnostic and individualized therapeutic strategies based on molecular patient stratification. Developments in this field specifically concentrate on predictive biomarkers for the response to conventional therapeutic agents, novel drugs targeting specific mutations and also new immunomodulatory drugs. The multitude of upcoming new predictive biomarkers requires the development and implementation of efficient test strategies and comprehensive technical methods, specifically when tissue restrictions inherent to lung cancer diagnostics are also taken into account. Novel procedures and technical aspects of these issues are discussed in this review.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Marcadores Genéticos/genética , Técnicas de Diagnóstico Molecular , Carcinoma de Pulmón de Células no Pequeñas/diagnóstico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Sistemas de Liberación de Medicamentos , Humanos , Inmunoterapia , Pulmón/patología , Medicina de Precisión , Pronóstico
11.
Pathologe ; 35(6): 565-73, 2014 Nov.
Artículo en Alemán | MEDLINE | ID: mdl-25106123

RESUMEN

Lung cancer is the most frequent cause of cancer-related death in Germany in men and women alike. While in the last decades a classification of epithelial lung tumors into non-small cell and small cell lung cancer was clearly sufficient from the therapeutic viewpoint, the dawn of the era of personalized medicine together with tremendous developments in the field of high throughput technologies have led to a molecular individualization of these tumors and, even more important, to a molecularly defined individualization of tumor therapy. This development resulted in the definition of a wide array of molecularly divergent tumor families. In this article we will give an overview on relevant molecular alterations in non-small cell lung cancers, comprising adenocarcinomas, squamous cell carcinomas and large cell carcinomas and also small cell carcinomas and carcinoids. Besides some similarities data gathered in the last few years specifically highlighted the immense diversity of molecular alterations that might underlie tumorigenesis of lung neoplasms. The knowledge on how to detect these alterations is of utmost importance in pathology, as treatment decisions are increasingly based on their presence or absence, putting molecular pathology in the central focus of the novel era of personalized medicine in oncology.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Células Pequeñas/genética , Carcinoma de Células Pequeñas/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Carcinoma de Pulmón de Células no Pequeñas/clasificación , Carcinoma de Pulmón de Células no Pequeñas/terapia , Carcinoma de Células Pequeñas/clasificación , Carcinoma de Células Pequeñas/terapia , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Progresión de la Enfermedad , Humanos , Inmunohistoquímica , Pulmón/patología , Neoplasias Pulmonares/clasificación , Neoplasias Pulmonares/terapia , Patología Molecular , Medicina de Precisión , Organización Mundial de la Salud
12.
Oncogene ; 33(42): 5006-16, 2014 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-24166495

RESUMEN

Synovial sarcoma is a high-grade soft tissue malignancy characterized by a specific reciprocal translocation t(X;18), which leads to the fusion of the SS18 (SYT) gene to one of three SSX genes (SSX1, SSX2 or SSX4). The resulting chimeric SS18-SSX protein is suggested to act as an oncogenic transcriptional regulator. Despite multimodal therapeutic approaches, metastatic disease is often lethal and the development of novel targeted therapeutic strategies is required. Several expression-profiling studies identified distinct gene expression signatures, implying a consistent role of Wnt/ß-catenin signaling in synovial sarcoma tumorigenesis. Here we investigate the functional and therapeutic relevance of Wnt/ß-catenin pathway activation in vitro and in vivo. Immunohistochemical analyses of nuclear ß-catenin and Wnt downstream targets revealed activation of canonical Wnt signaling in a significant subset of 30 primary synovial sarcoma specimens. Functional aspects of Wnt signaling including dependence of Tcf/ß-catenin complex activity on the SS18-SSX fusion proteins were analyzed. Efficient SS18-SSX-dependent activation of the Tcf/ß-catenin transcriptional complex was confirmed by TOPflash reporter luciferase assays and immunoblotting. In five human synovial sarcoma cell lines, inhibition of the Tcf/ß-catenin protein-protein interaction significantly blocked the canonical Wnt/ß-catenin signaling cascade, accompanied by the effective downregulation of Wnt targets (AXIN2, CDC25A, c-MYC, DKK1, CyclinD1 and Survivin) and the specific suppression of cell viability associated with the induction of apoptosis. In SYO-1 synovial sarcoma xenografts, administration of small molecule Tcf/ß-catenin complex inhibitors significantly reduced tumor growth, associated with diminished AXIN2 protein levels. In summary, SS18-SSX-induced Wnt/ß-catenin signaling appears to be of crucial biological importance in synovial sarcoma tumorigenesis and progression, representing a potential molecular target for the development of novel therapeutic strategies.


Asunto(s)
Proteínas de Fusión Oncogénica/fisiología , Sarcoma Sinovial/metabolismo , Vía de Señalización Wnt , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Núcleo Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Expresión Génica , Células HEK293 , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Perileno/análogos & derivados , Perileno/farmacología , Pirimidinonas/farmacología , Sarcoma Sinovial/tratamiento farmacológico , Triazinas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , beta Catenina/genética , beta Catenina/metabolismo
14.
Dtsch Med Wochenschr ; 138(20): 1061-8, 2013 May.
Artículo en Alemán | MEDLINE | ID: mdl-23670263

RESUMEN

Tissue-based molecular diagnostics is a fast growing diagnostic field, which already complements morphologic classifications in many cases. Pathology based molecular diagnosis is performed almost exclusively on paraffin embedded material and always in conjunction with histopathology. Besides the classic field of tissue based detection of pathogenic organisms such as bacteria, viruses and fungi, molecular diagnostics of tumor tissue is one of the current hot topics in oncology. In this context the detection of predictive molecular biomarkers, such as specific mutations, allows patient stratification for individually tailored treatment strategies and thereby is one of the key components of individualized patient care in oncology. The rapidly growing number of clinically relevant predictive biomarkers together with impressive technical advances, specifically the development of massive parallel sequencing, will modify the care of patients with malignant diseases. Pathology, therefore, has returned in the very center of interdisciplinary patient care.


Asunto(s)
Técnicas de Diagnóstico Molecular/métodos , Técnicas de Diagnóstico Molecular/tendencias , Neoplasias/patología , Patología Molecular/métodos , Patología Molecular/tendencias , Biomarcadores de Tumor/genética , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Aberraciones Cromosómicas , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Neoplasias/genética , Neoplasias/terapia , Adhesión en Parafina , Pronóstico , Análisis de Secuencia de ADN/métodos
16.
Pathologe ; 32(1): 47-56, 2011 Feb.
Artículo en Alemán | MEDLINE | ID: mdl-21234572

RESUMEN

The conceptual evolution in the classification of pleomorphic high-grade sarcomas is a paradigm of how the integrative morphological, immunohistochemical and molecular genetic analysis has contributed to a clinical, prognostic and therapy-oriented characterization of this complex group of tumors. The clinical and prognostic relevance of a refined subtyping of pleomorphic high-grade sarcomas, which until recently was considered a mere academic exercise, is now undisputed. It is imperative to unequivocally differentiate sarcomas from non-sarcomatous, clearly defined malignancies to start adequate therapy. Furthermore, pleomorphic sarcomas which are particularly aggressive and prone to poor prognosis, have to be separated from sarcomas which, in contrast to the pleomorphic phenotype, are characterized by a less aggressive behavior. Also, morphologically pleomorphic but benign mesenchymal tumors must be recognized. Finally, it is important to promote the promising, array-based identification of diagnostic, prognostic and clinically relevant gene signatures on larger collections of pathomorphologically and clinically precisely defined subtypes of pleomorphic high-grade sarcomas.


Asunto(s)
Transformación Celular Neoplásica/clasificación , Transformación Celular Neoplásica/patología , Sarcoma/clasificación , Sarcoma/patología , Neoplasias de los Tejidos Blandos/clasificación , Neoplasias de los Tejidos Blandos/patología , Transformación Celular Neoplásica/genética , Tejido Conectivo/patología , Diagnóstico Diferencial , Marcadores Genéticos/genética , Histiocitoma Fibroso Maligno/clasificación , Histiocitoma Fibroso Maligno/genética , Histiocitoma Fibroso Maligno/patología , Humanos , Liposarcoma/clasificación , Liposarcoma/genética , Liposarcoma/patología , Técnicas de Diagnóstico Molecular , Pronóstico , Sarcoma/genética , Neoplasias de los Tejidos Blandos/genética
17.
Pathologe ; 31 Suppl 2: 211-4, 2010 Oct.
Artículo en Alemán | MEDLINE | ID: mdl-20711583

RESUMEN

To establish precise diagnostic algorithms and standardised treatment of sarcomas in specialized centers, the interdisciplinary research group KoSar (sarcoma competence network) has been funded by German Cancer Aid. A sarcoma tissue repository and a diagnostic reference center have been set up, presently containing about 1000 accurately diagnosed sarcomas of different entities. Significant gene expression profiles for synovial sarcomas, leiomyosarcomas, myxoid liposarcomas and a small profile for myxofibrosarcomas as well as a new classification of angiosarcomas were defined. We systematically searched for activated signal transduction pathways in sarcoma cell lines and xenograft transplant models and candidate targets for molecular therapies were identified. Based on these results first clinical studies have been initiated by the German Interdisciplinary Sarcoma Study Group (GISG).


Asunto(s)
Sarcoma/genética , Sarcoma/patología , Animales , Investigación Biomédica , Línea Celular Tumoral , Conducta Cooperativa , Evaluación Preclínica de Medicamentos , Fibrosarcoma/diagnóstico , Fibrosarcoma/tratamiento farmacológico , Fibrosarcoma/genética , Fibrosarcoma/patología , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Comunicación Interdisciplinaria , Leiomiosarcoma/diagnóstico , Leiomiosarcoma/tratamiento farmacológico , Leiomiosarcoma/genética , Leiomiosarcoma/patología , Liposarcoma Mixoide/diagnóstico , Liposarcoma Mixoide/tratamiento farmacológico , Liposarcoma Mixoide/genética , Liposarcoma Mixoide/patología , Técnicas de Diagnóstico Molecular , Terapia Molecular Dirigida , Trasplante de Neoplasias , Sarcoma/diagnóstico , Sarcoma/tratamiento farmacológico , Sarcoma Sinovial/diagnóstico , Sarcoma Sinovial/tratamiento farmacológico , Sarcoma Sinovial/genética , Sarcoma Sinovial/patología , Transducción de Señal/genética
18.
Pathologe ; 29 Suppl 2: 353-6, 2008 Nov.
Artículo en Alemán | MEDLINE | ID: mdl-18712391

RESUMEN

Flat epithelial atypia (FEA) of the breast has recently gained attention as a possible precursor lesion of highly differentiated breast cancer. Especially tubular carcinomas, with which FEA shares cytological features, often occur in close proximity to each other. To examine a possible clonal relationship, we analysed mutations of the highly variable region of the mitochondrial genome in a series of tubular carcinomas, associated FEA and normal glands. Multiple sequence alignment showed identical mtDNA mutations in approximately 50% of paired FEA and tumour samples, indicative of a clonal relationship. Our data indicate a possible precursor role of FEA in the development of tubular breast cancer.


Asunto(s)
Adenocarcinoma/genética , Neoplasias de la Mama/genética , Transformación Celular Neoplásica/genética , Análisis Mutacional de ADN , ADN Mitocondrial/genética , Células Epiteliales/patología , Lesiones Precancerosas/genética , Adenocarcinoma/patología , Anciano , Mama/patología , Neoplasias de la Mama/patología , Calcinosis/genética , Calcinosis/patología , Núcleo Celular/genética , Núcleo Celular/patología , Transformación Celular Neoplásica/patología , Femenino , Humanos , Persona de Mediana Edad , Estadificación de Neoplasias , Lesiones Precancerosas/patología , Pronóstico , Análisis de Secuencia de ADN
19.
J Clin Pathol ; 61(9): 1034-7, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18559375

RESUMEN

AIMS: Two different forms of vulvar intraepithelial neoplasia (VIN) are recognised: (1) usual-type (bowenoid) VIN, which is related to high-risk papillomavirus infection, and (2) differentiated (simplex) VIN, which is associated with chronic inflammation. The aim of this study was to investigate the presence of chromosome 3q26 gains in the spectrum of precancerous lesions and invasive squamous cell carcinomas (SCCs) of the vulva. METHODS: 3q26 gains were analysed using fluorescence in situ hybridisation in a series of usual-type VINs, VINs of the differentiated type and invasive squamous cell carcinomas. In addition, all cases were examined for human papillomavirus (HPV) DNA, p53 mutations, and p16 and p53 protein expression. RESULTS: Gains of chromosome 3q26 were present in all VINs of the differentiated type and in 50% of the usual-type VIN lesions. 81% of SCCs were positive for 3q26 gains irrespective of the HPV status and of the associated precursor lesion. HPV-associated lesions exhibited the typical, strong cytoplasmic p16 accumulation while mutated p53 was only detected in HPV-negative VINs or SCCs, and was associated with an overexpression of p53 protein. CONCLUSIONS: Immunohistochemical evaluation of p16 and p53 expression aids in the differential diagnosis of squamous cell alterations of the vulva. However, detection of 3q26 imbalance is of additional diagnostic value in difficult cases of HPV-unrelated usual-type VINs and VINs of the differentiated type.


Asunto(s)
Carcinoma in Situ/genética , Carcinoma de Células Escamosas/genética , Aberraciones Cromosómicas , Cromosomas Humanos Par 3/genética , Neoplasias de la Vulva/genética , Anciano , Carcinoma in Situ/virología , Carcinoma de Células Escamosas/virología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/análisis , Femenino , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Genes p16 , Genes p53 , Humanos , Hibridación Fluorescente in Situ , Persona de Mediana Edad , Invasividad Neoplásica/genética , Infecciones por Papillomavirus/complicaciones , Reacción en Cadena de la Polimerasa , Proteína p53 Supresora de Tumor/análisis , Infecciones Tumorales por Virus/complicaciones , Neoplasias de la Vulva/virología
20.
J Pathol ; 215(2): 195-203, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18421760

RESUMEN

In chronic pancreatitis (CP), both the progressive loss of acinar parenchyma and aggressive fibro-inflammatory reactions ultimately lead to irreversible organ destruction. Dying cells are normally removed by macrophages and elimination is associated with anti-inflammatory cytokine switch. We investigated whether defective clearance of damaged acini by macrophages such as compromised phagocytosis or altered cytokine reaction occurs in CP and thus represents a causative link between acinar loss and fibro-inflammation. In a checkerboard-like co-culture system, we assessed normal and CP macrophages for their phagocytic and cytokine responses to dying pancreatic acinar cells of normal or CP origin by FACS, confocal microscopy, QRT-PCR, and ELISA. In CP, phagocytosis of apoptotic acini by macrophages was not impaired; however, the associated cytokine responses were gradually perturbed. Most interestingly, only normal acini suppressed TGFbeta1 expression and accumulation specifically in normal macrophage cultures, while CP acini lost this ability. Both types of apoptotic acini induced pro-inflammatory cytokine bursts of varying strength in both types of macrophages; however, the most significant difference (more than 50-fold higher expression of IL-1beta, IL-6, and IL-8) was evident between CP/CP and normal/normal combinations, indicating that acinar and macrophage alterations synergistically lead to the ultimate CP-specific bias. In combination with in situ data comparing circulating inflammatory cells to pancreatic resident ones, our results indicate that cytokine expression in inflammatory cells undergoes spatiotemporal modulation, most likely through a successive interplay of acinar, stromal, and circulating factors. Thus, clearance of injured pancreatic acini by macrophages is associated with a unique cytokine reaction which may constitute a basis for progression of SAPE (sentinel acute pancreatitis event) to the irreversible fibro-inflammation in CP.


Asunto(s)
Macrófagos/fisiología , Páncreas/inmunología , Pancreatitis Crónica/inmunología , Apoptosis , Técnicas de Cocultivo , Ensayo de Inmunoadsorción Enzimática/métodos , Citometría de Flujo , Humanos , Interleucina-1/inmunología , Interleucina-6/inmunología , Interleucina-8/inmunología , Macrófagos/inmunología , Microscopía Confocal , Páncreas/patología , Pancreatitis Crónica/patología , Fagocitosis , Reacción en Cadena de la Polimerasa/métodos , Factor de Necrosis Tumoral alfa/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...