19 research outputs found

    Application of FISH in the diagnosis of lung cancer

    No full text
    Rapid advancements in the area of lung cancer therapy were determined by the discovery of molecular markers and the possibility of their therapeutic exploitation. Today's lung cancer diagnosis places high demands on pathologists. In the majority of cases, small tissue samples must suffice for diagnosis and testing of all relevant biomarkers. The minimum panel required for advanced non-small-cell lung carcinoma (NSCLC) with nonsquamous histology includes testing ofEGFR,BRAF,ALK,ROS1, and PD-L1-expression. So far, only PD-L1-IHC (immunohistochemistry, IHC) is required for squamous cell carcinoma. If possible, newer biomarkers such asRET,MET,HER2,NTRK,andKRASshould be integrated in test panels. Fluorescence in situ hybridization (FISH) is a well-established molecular method for the detection of chromosomal aberrations, such asALK-,ROS1-,andRET- translocations and amplifications, such asHer2/neuorMET. The relevance ofMET-FISH for the detection of amplifications in the first-line setting is controversial, but of high importance in the recurrent setting. All equivocal or discrepant results should be validated using orthogonal methods. IHC is a suitable, thoroughly validated method forALKandROS1aberration detection with the advantage of quick and cost-efficient test results and tissue conservation. All other translocations, or discrepancy between IHC and FISH, require a sequencing-based confirmation procedure. The low frequency ofNTRKfusions, and high sensitivity of NTRK-IHC, suggest using IHC as a prescreening tool with subsequent sequencing-based analysis for IHC positive cases

    Anwendungen der FISH in der Diagnostik von Lungenkarzinomen

    No full text
    <jats:title>Zusammenfassung</jats:title><jats:p>Die rasante Entwicklung im Bereich der Lungenkrebstherapie wurde maßgeblich auch durch die Entdeckung molekularer Marker und der damit verbundenen Möglichkeit einer personalisierten Therapie bestimmt. Die heutige Lungenkrebsdiagnostik stellt hohe Anforderungen an den Pathologen. An kleinen Gewebeproben muss nicht nur die Diagnose gestellt, sondern müssen auch alle therapierelevanten Biomarker getestet werden. Das verlangte Mindestmaß bei fortgeschrittenem nichtkleinzelligen Lungenkarzinom („non small cell lung cancer“, NSCLC) mit Nicht-Plattenepithel-Histologie umfasst die Testung von <jats:italic>EGFR, BRAF, ALK, ROS1</jats:italic> und PD-L1. Für Plattenepithelkarzinome ist bislang nur die PD-L1-IHC (Immunhistochemie, IHC) gefordert. Nach Möglichkeit sollten neuere Biomarker wie <jats:italic>RET, MET, HER2, NTRK</jats:italic> und <jats:italic>KRAS</jats:italic> integriert werden. Die Fluoreszenz-in-situ-Hybridisierung („fluorescence in situ hybridization“, FISH) ist eine gut-etablierte Methode zum Nachweis einer <jats:italic>ALK</jats:italic>-, <jats:italic>ROS1</jats:italic>- und <jats:italic>RET</jats:italic>-Translokation, wobei die ALK-IHC als gleichwertig anerkannt wurde. Die Relevanz der <jats:italic>MET</jats:italic>-FISH zum Amplifikationsnachweis im <jats:italic>First-line</jats:italic>-<jats:italic>Setting</jats:italic> ist umstritten. Nicht eindeutige Fälle sollten immer mit einem orthogonalen Verfahren validiert werden. Hierzu eignet sich bei <jats:italic>ALK</jats:italic> und <jats:italic>ROS1</jats:italic> die IHC mit dem Vorteil schneller und kostengünstiger Testergebnisse sowie geringen Gewebeverbrauchs. Bei allen anderen Translokationen oder bei Diskrepanz zwischen IHC und FISH sollte ein sequenzierungsbasiertes Verfahren ergänzt werden. Zur Detektion der seltenen <jats:italic>NTRK</jats:italic>-Fusionen eignet sich bei hoher Sensitivität ein IHC-Vorscreening; die sequenzierungsbasierte Analyse ist hier bei Positivität zur Bestätigung indiziert.</jats:p&gt

    Interdisciplinary assessment and management of a patient with a fibrous gingival enlargement of unknown origin: A case report

    No full text
    A gingival enlargement of unclear cause could only be diagnosed after interdisciplinary cooperation as plasma cell gingivitis of unknown origin. Interdisciplinary approaches remain crucial when diagnosing rare gum diseases

    First reported case of a collision tumor composed of pancreatic adenocarcinoma and retroperitoneal liposarcoma: a case report

    No full text
    BackgroundCollision tumors are rare cases with two different tumor entities growing synchronously. While adenocarcinoma of the pancreas is the most common pancreatic tumor with an incidence of 10 per 100.000, retroperitoneal liposarcoma remains very rare. This is the first report of a collision tumor between these two tumor entities.Case presentationDemographic details:The tumor was diagnosed in a 64 male Caucasian patient. Besides atrial fibrillation, arterial hypertension and a hypothyroidism there is no relevant medical history especially no history of cancer.Clinical details:During a routine check-up an unclassified tumor of the pancreatic tail was diagnosed. The lab showed no pathologies. Tumor markers were negative for carbohydrate antigen 19-9 and 72-4 (CA 19-9, CA 72-4) and carcinoembryonic antigen (CEA). Alpha-fetoprotein (AFP) and neuron specific enolase (NSE) were both elevated (AFP 97kU/l, (<5,8kU/l) and NSE 30,0g/l (16,4g/l)). A computed tomography-guided core needle biopsy was performed which revealed a low-grade liposarcoma (G1). A CT scan showed no metastases. A surgical resection was recommended by the interdisciplinary tumor board.Interventions: A systematic left sided retroperitoneal compartment resection including en-bloc-left sided pancreatectomy, splenectomy, nephrectomy, hemicolectomy, adrenalectomy, partial gastrectomy and partial resection of the diaphragm was performed. Pathology revealed a collision tumor consisting of pancreatic adenocarcinoma that was classified pT3, pN2 (11/33 ece+) L1V0 Pn0, R0; G2 [UICC Stage III] and a liposarcoma pT2, pN0 (0/33) L0V0 Pn0, G1 [UICC Stage Ib].The postoperative tumor board recommended an adjuvant chemotherapy with gemcitabine and capecitabine for the locally advanced pancreatic adenocarcinoma.Outcome: At the latest follow-up (1year after surgery) the patient was in good clinical condition and without evidence of tumor recurrence.ConclusionCollision tumors are rare and difficult to diagnose. This is the first description of a collision tumor composed of pancreatic adenocarcinoma and retroperitoneal liposarcoma.The reported case demonstrates that inconsistent diagnostic results (e.g. imaging and pathology) should raise suspicion concerning the diagnosis. Awareness of these rare cases might protect us from underdiagnosing patients and therefore leading to better patient care.There is evolving evidence that will lead to more personalized treatment options for somatic BRCA mutated pancreatic cancer

    Molecular In-Depth Characterization of Chondrosarcoma for Current and Future Targeted Therapies

    No full text
    Chondrosarcoma (CHS) are heterogenous, but as a whole, represent the second most common primary malignant bone tumor entity. Although knowledge on tumor biology has grown exponentially during the past few decades, surgical resection remains the gold standard for the treatment of these tumors, while radiation and differentiated chemotherapy do not result in sufficient cancer control. An in-depth molecular characterization of CHS reveals significant differences compared to tumors of epithelial origin. Genetically, CHS are heterogenous, but there is no characteristic mutation defining CHS, and yet, IDH1 and IDH2 mutations are frequent. Hypovascularization, extracellular matrix composition of collagen, proteoglycans, and hyaluronan create a mechanical barrier for tumor suppressive immune cells. Comparatively low proliferation rates, MDR-1 expression and an acidic tumor microenvironment further limit therapeutic options in CHS. Future advances in CHS therapy depend on the further characterization of CHS, especially the tumor immune microenvironment, for improved and better targeted therapies

    Tumor budding assessed according to the criteria of the International Tumor Budding Consensus Conference determines prognosis in resected esophageal adenocarcinoma

    No full text
    Only few studies examined the prognostic effect of tumor budding in esophageal adenocarcinomas so far. However, different quantification approaches were used, so results cannot be directly compared. Recently, the International Tumor Budding Consensus Conference (ITBCC) published consensus criteria for the evaluation of tumor budding in colorectal cancer, which we applied in our study. Hematoxylin and eosin (H&E) and cytokeratin (AE1/AE3) stained whole tissue slides of 104 resected esophageal adenocarcinomas were evaluated. The mean count of tumor buds was analyzed in one high power field according to the ITBCC criteria and assigned to budding groups Bd1-3. Tumor budding was significantly associated with a worse overall survival. Regardless of the quantification approach, an increased number of tumor buds was significantly associated with reduced overall survival (OS) (H&E: HR = 1.05 (95% CI 1.029-1.073),p < 0.001; cytokeratin: HR = 1.073 (95% CI 1.045-1.101),p < 0.001). In multivariable analysis tumor budding according to ITBCC criteria on H&E stained slides was an independent prognostic factor. Tumor budding, according to ITBCC criteria, is an independent prognostic factor in resected esophageal adenocarcinoma. Prospective studies using ITBCC criteria are useful in the near future to validate our results

    International Tumor Budding Consensus Conference criteria determine the prognosis of oesophageal adenocarcinoma with poor response to neoadjuvant treatment

    No full text
    Background: Neoadjuvant therapy regimens followed by surgery represent the current standard treatment of locally advanced oesophageal adenocarcinomas. Tumour regression determines prognosis, but more than half of patients do have more than 10% residual tumour after neoadjuvant therapy. In these cases, classical histopathological parameters for the determination of prognosis are of limited value. Therefore, we investigated whether tumour budding could be an additional prognostic factor for tumours with poor response to neoadjuvant therapy. Methods: Tumour budding was assessed according to a standardized consensus quantification method as proposed by the International Tumor Budding Consensus Conference (ITBCC) in H&E-stained whole tissue slides of 278 formalin-fixed paraffin-embedded (FFPE) resected oesophageal adenocarcinomas with a poor response (> 10% vital residual tumour) to neoadjuvant therapy. Results: We could demonstrate a strong positive correlation (p < 0.05) between the budding group, ypN stage and UICC tumour stage. Further, high numbers of tumour buds were a significant and independent negative prognostic marker for OS in all studied patients (HR = 1.039 (95% CI 1.012-1.066), p = 0.004). ITBCC budding groups were an independent prognostic parameter. Conclusions: Tumour budding assessed in accordance with the ITBCC criteria may aid in the prognostic stratification of locally advanced oesophageal adenocarcinoma with poor response to neoadjuvant treatment

    Cardiac metastasis causes paradoxical malignant embolism

    No full text
    Background Embolic events play an important role in clinical everyday practice. Malignant arterial embolism is a rare nevertheless often fatal entity for cardiac, cerebral or systemic ischemia, requiring immediate diagnosis and treatment. Case This is a case report of a 65 years-old female, suffering from pulmonal adenocarcinoma, who was hospitalized due to neurological deficits caused by an acute ischemic stroke, followed by anterior myocardial infarction within 3 days. Diagnostic work-up revealed metastasis of the pulmonal adenocarcinoma in the right atrium and a patent foramen ovale. Histopathological examination of the coronary embolus verified paradoxical arterial embolism of the pulmonal adenocarcinoma into a coronary vessel and consequently cerebral arteries. Conclusion The present case underlines the need for (i), consideration of malignant embolism, (ii) histopathological examination of the embolus to determine its etiology, and (iii) interdisciplinary discussion of individual therapeutic and prevention strategies in cancer patients with cerebral, cardiac or systemic embolic events

    Mismatch Repair Deficiency and Somatic Mutations in Human Sinonasal Tumors

    No full text
    Simple Summary Sinonasal carcinomas are rare tumors with an overall poor prognosis. Due to limitations in local therapeutic approaches, systemic neo-adjuvant or adjuvant therapies are becoming increasingly important in order to improve patient outcome. This study aimed to examine potentially therapeutic targetable molecular alterations in different sinonasal tumors, including deficiency in mismatch repair proteins and microsatellite instability as well as driver mutations. According to our results, immunohistochemical (IHC) analysis of mismatch repair (MMR) proteins and sequencing-based panel analysis should be integrated into the diagnostics of clinically aggressive inverted sinonasal papilloma (ISP) and sinonasal squamous cell carcinoma (SNSCC) in order to enable the therapeutic possibility of a targeted therapy. Due to limitations in local therapy approaches for sinonasal tumors, improvement in systemic therapies plays a pivotal role for prolongation of the patient's survival. The aim of this study was to examine potential biomarkers, including deficiency in mismatch repair proteins (dMMR)/microsatellite instability (MSI-H) in sinonasal cancers and their precancerous lesions. A comprehensive analysis of 10 sinonasal cancer cell lines by whole exome sequencing, screening 174 sinonasal tumors by immunohistochemistry (IHC) for mismatch repair deficiency and next generation sequencing (NGS) of 136 tumor samples revealed a dMMR/MSI-H sinonasal squamous cell carcinoma (SNSCC) cell line based on a somatic missense mutation in MLH1 and an overall frequency of dMMR/MSI-H SNSCC of 3.2% (4/125). Targetable EGFR mutations were found in 89.3% (25/28) of inverted sinonasal papilloma (ISP) and in 60% (6/10) of ISP-associated carcinomas. While PIK3CA and EGFR mutations were not mutually exclusive, KRAS mutated tumors were an EGFR-wildtype. The effect of potential driver mutations in FGFR2, FGFR3, BRAF, HRAS, MAP2K1, PTEN, NOTCH1 and CARD11 need further investigation. Our results suggest that biomarker testing, including MMR-IHC and NGS panel analysis, should be integrated into the diagnostics of clinically aggressive ISPs and SNSCC to assess prognosis and facilitate therapeutic decisions

    FGFR2-amplified tumor clones are markedly heterogeneously distributed in carcinomas of the upper gastrointestinal tract

    No full text
    Background FGFR2 is a therapy-relevant target in tumors of the upper gastrointestinal tract (GIT), and clinical trials are currently underway to test the efficacy of FGFR2 inhibitors. Tumor heterogeneity is one of the relevant causes of treatment failure. Almost nothing is known about the heterogeneous distribution of FGFR2-amplified clones in adenocarcinomas of the upper GIT. Patients and methods To assess FGFR2 gene copy number alteration and intratumoral heterogeneity of upper GIT adenocarcinomas, we analyzed 893 patient-derived formalin-fixed paraffin-embedded tumor specimens, including primary operated and neoadjuvant-treated tumors (462 gastric carcinomas and 429 esophageal adenocarcinomas) as well as complementary lymph node and distant metastasis by fluorescence in situ hybridization. Results Twenty-six gastric tumors (5.6%) and 21 esophageal adenocarcinomas (4.9%) showed FGFR2 amplification. Overall, 93% of gastric carcinomas and 83% of esophageal carcinomas showed heterogeneous amplification. FGFR2 amplification was found in different histological growth patterns, including intestinal and diffuse type according to the Lauren classification. In the primary gastric carcinoma group, FGFR2 amplification was associated with poor prognosis (p = 0.005). Conclusion Homogeneous FGFR2 amplification in tumors of the upper GIT is the exception. This has highly relevant implications in the nature of FGFR2 diagnostics (sufficient tumor cell number, determination of amplification at metastasis versus primary tumor, etc.) and on the response probability of appropriate inhibitors. It is relevant that the often poorly treatable and aggressive subtype of diffuse carcinomas (poorly cohesive carcinomas) also shows FGFR2 amplification and that an individualized therapy option with FGFR2 inhibitors could be an option in this group
    corecore