Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Pathologica ; 116(3): 170-175, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38979591

ABSTRACT

Embryonal tumors with multilayered rosettes (ETMR) are highly aggressive and therapy-resistant pediatric central nervous system (CNS) tumors that have three histological patters: embryonal tumor with abundant neuropil and true rosettes, ependymoblastoma, and medulloepithelioma. We present a case of ETMR in an 18-year-old woman with DICER1 syndrome. This report confirms the important role of DNA-methylation analysis in the classification of CNS embryonal tumors and the importance of investigating somatic and germline DICER1 mutations in all CNS embryonal tumors.


Subject(s)
DEAD-box RNA Helicases , Neoplasms, Germ Cell and Embryonal , Ribonuclease III , Humans , Female , Ribonuclease III/genetics , DEAD-box RNA Helicases/genetics , Adolescent , Neoplasms, Germ Cell and Embryonal/genetics , Neoplasms, Germ Cell and Embryonal/pathology , DNA Methylation
2.
Histopathology ; 84(4): 683-696, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38084641

ABSTRACT

AIMS: Ovarian Wilms tumour (WT)/nephroblastoma is an extremely rare neoplasm that has been reported to occur in pure form or as a component of a teratomatous neoplasm. We hypothesized that teratoma-associated and pure ovarian WT may represent different tumour types with diverging molecular backgrounds. To test this hypothesis, we comprehensively characterized a series of five tumours originally diagnosed as ovarian WT. METHODS AND RESULTS: The five cases comprised three teratoma-associated (two mature and one immature) and two pure WTs. Two of the teratoma-associated WTs consisted of small nodular arrangements of "glandular"/epithelial structures, while the third consisted of both an epithelial and a diffuse spindle cell/blastemal component. The pure WTs consisted of "glandular" structures, which were positive for sex cord markers (including inhibin and SF1) together with a rhabdomyosarcomatous component. The two pure WTs harboured DICER1 pathogenic variants (PVs), while the three associated with teratomas were DICER1 wildtype. Panel-based DNA sequencing of four of the cases did not identify PVs in the other genes investigated. Analysis of the HA19/IGF2 imprinting region showed retention of imprinting in the pure WTs but loss of heterozygosity with hypomethylation of the ICR1 region in two of three teratoma-associated WTs. Furthermore, copy number variation and clustering-based whole-genome DNA methylation analyses identified divergent molecular profiles for pure and teratoma-associated WTs. CONCLUSION: Based on the morphological features, immunophenotype, and molecular findings (DICER1 PVs, copy number, and DNA methylation profiles), we suggest that the two cases diagnosed as pure primary ovarian WT represent moderately to poorly differentiated Sertoli Leydig cell tumours (SLCTs), while the tumours arising in teratomas represent true WTs. It is possible that at least some prior cases reported as pure primary ovarian WT represent SLCTs.


Subject(s)
Kidney Neoplasms , Ovarian Neoplasms , Sex Cord-Gonadal Stromal Tumors , Teratoma , Wilms Tumor , Male , Female , Humans , DNA Copy Number Variations , Wilms Tumor/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Teratoma/genetics , Teratoma/pathology , Kidney Neoplasms/genetics , Ribonuclease III/genetics , DEAD-box RNA Helicases/genetics
3.
Mod Pathol ; 37(1): 100374, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37925057

ABSTRACT

Dedifferentiated and undifferentiated ovarian carcinomas (DDOC/UDOC) are rare neoplasms defined by the presence of an undifferentiated carcinoma. In this study, we detailed the clinical, pathological, immunohistochemical, and molecular features of a series of DDOC/UDOC. We collected a multi-institutional cohort of 23 DDOC/UDOC and performed immunohistochemistry for core switch/sucrose nonfermentable (SWI/SNF) complex proteins (ARID1A, ARID1B, SMARCA4, and SMARCB1), mismatch repair (MMR) proteins, and p53. Array-based genome-wide DNA methylation and copy number variation analyses were performed on a subset of cases with comparison made to a previously reported cohort of undifferentiated endometrial carcinoma (UDEC), small cell carcinoma of the ovary, hypercalcemic type (SCCOHT), and tubo-ovarian high-grade serous carcinoma (HGSC). The age of all 23 patients with DDOC/UDOC ranged between 22 and 71 years (with an average age of 50 years), and a majority of them presented with extraovarian disease (16/23). Clinical follow-up was available for 19 patients. Except for 2 patients, the remaining 17 patients died from disease, with rapid disease progression resulting in mortality within a year in stage II-IV settings (median disease-specific survival of 3 months). Eighteen of 22 cases with interpretable immunohistochemistry results showed loss of expression of core SWI/SNF protein(s) that are expected to result in SWI/SNF complex inactivation as 10 exhibited coloss of ARID1A and ARID1B, 7 loss of SMARCA4, and 1 loss of SMARCB1. Six of 23 cases were MMR-deficient. Two of 20 cases exhibited mutation-type p53 immunoreactivity. Methylation profiles showed coclustering of DDOC/UDOC with UDEC, which collectively were distinct from SCCOHT and HGSC. However, DDOC/UDOC showed an intermediate degree of copy number variation, which was slightly greater, compared with SCCOHT but much less compared with HGSC. Overall, DDOC/UDOC, like its endometrial counterpart, is highly aggressive and is characterized by frequent inactivation of core SWI/SNF complex proteins and MMR deficiency. Its molecular profile overlaps with UDEC while being distinct from SCCOHT and HGSC.


Subject(s)
Brain Neoplasms , Carcinoma, Small Cell , Carcinoma , Colorectal Neoplasms , Endometrial Neoplasms , Neoplastic Syndromes, Hereditary , Ovarian Neoplasms , Female , Humans , Middle Aged , Young Adult , Adult , Aged , Tumor Suppressor Protein p53/genetics , DNA Copy Number Variations , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Carcinoma/pathology , Carcinoma, Ovarian Epithelial , Endometrial Neoplasms/pathology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , DNA Helicases/genetics , DNA Helicases/metabolism , Nuclear Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Histopathology ; 84(4): 712-714, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38114269

ABSTRACT

Currently available molecular data support a dichotomous classification of Sertoli-Leydig cell tumours (SLCTs) based on DICER1 mutational status. This correspondence suggests a possible roadmap towards a molecular-based classification of SLCT.


Subject(s)
Ovarian Neoplasms , Sertoli-Leydig Cell Tumor , Sex Cord-Gonadal Stromal Tumors , Female , Humans , Sertoli-Leydig Cell Tumor/genetics , Sertoli-Leydig Cell Tumor/pathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Sex Cord-Gonadal Stromal Tumors/pathology , Ribonuclease III/genetics , Genomics , DEAD-box RNA Helicases/genetics
6.
Nat Struct Mol Biol ; 30(11): 1640-1652, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37735617

ABSTRACT

The SS18-SSX fusion drives oncogenic transformation in synovial sarcoma by bridging SS18, a member of the mSWI/SNF (BAF) complex, to Polycomb repressive complex 1 (PRC1) target genes. Here we show that the ability of SS18-SSX to occupy H2AK119ub1-rich regions is an intrinsic property of its SSX C terminus, which can be exploited by fusion to transcriptional regulators beyond SS18. Accordingly, SS18-SSX recruitment occurs in a manner that is independent of the core components and catalytic activity of BAF. Alternative SSX fusions are also recruited to H2AK119ub1-rich chromatin and reproduce the expression signatures of SS18-SSX by engaging with transcriptional activators. Variant Polycomb repressive complex 1.1 (PRC1.1) acts as the main depositor of H2AK119ub1 and is therefore required for SS18-SSX occupancy. Importantly, the SSX C terminus not only depends on H2AK119ub1 for localization, but also further increases it by promoting PRC1.1 complex stability. Consequently, high H2AK119ub1 levels are a feature of murine and human synovial sarcomas. These results uncover a critical role for SSX-C in mediating gene deregulation in synovial sarcoma by providing specificity to chromatin and further enabling oncofusion binding by enhancing PRC1.1 stability and H2AK119ub1 deposition.


Subject(s)
Sarcoma, Synovial , Humans , Animals , Mice , Sarcoma, Synovial/genetics , Sarcoma, Synovial/metabolism , Polycomb Repressive Complex 1/genetics , Transcriptional Activation , Cell Nucleus/metabolism , Chromatin/metabolism , Oncogene Proteins, Fusion/metabolism , Cell Cycle Proteins/metabolism
7.
Gynecol Oncol ; 176: 162-172, 2023 09.
Article in English | MEDLINE | ID: mdl-37556934

ABSTRACT

OBJECTIVE: Dedifferentiated endometrial cancer (DDEC) is an uncommon and clinically highly aggressive subtype of endometrial cancer characterized by genomic inactivation of SWItch/Sucrose Non-Fermentable (SWI/SNF) complex protein. It responds poorly to conventional systemic treatment and its rapidly progressive clinical course limits the therapeutic windows to trial additional lines of therapies. This underscores a pressing need for biologically accurate preclinical tumor models to accelerate therapeutic development. METHODS: DDEC tumor from surgical samples were implanted into immunocompromised mice for patient-derived xenograft (PDX) and cell line development. The histologic, immunophenotypic, genetic and epigenetic features of the patient tumors and the established PDX models were characterized. The SMARCA4-deficienct DDEC model was evaluated for its sensitivity toward a KDM6A/B inhibitor (GSK-J4) that was previously reported to be effective therapy for other SMARCA4-deficient cancer types. RESULTS: All three DDEC models exhibited rapid growth in vitro and in vivo, with two PDX models showing spontaneous development of metastases in vivo. The PDX tumors maintained the same undifferentiated histology and immunophenotype, and exhibited identical genomic and methylation profiles as seen in the respective parental tumors, including a mismatch repair (MMR)-deficient DDEC with genomic inactivation of SMARCA4, and two MMR-deficient DDECs with genomic inactivation of both ARID1A and ARID1B. Although the SMARCA4-deficient cell line showed low micromolecular sensitivity to GSK-J4, no significant tumor growth inhibition was observed in the corresponding PDX model. CONCLUSIONS: These established patient tumor-derived models accurately depict DDEC and represent valuable preclinical tools to gain therapeutic insights into this aggressive tumor type.


Subject(s)
Brain Neoplasms , Colorectal Neoplasms , Endometrial Neoplasms , Female , Humans , Animals , Mice , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/genetics , Endometrial Neoplasms/metabolism , Cell Differentiation , Biomarkers, Tumor/genetics , DNA Helicases , Nuclear Proteins/genetics , Transcription Factors/genetics , DNA-Binding Proteins/genetics
8.
Cancer Res ; 83(21): 3517-3528, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37494476

ABSTRACT

DICER1 is an RNase III enzyme essential for miRNA biogenesis through cleaving precursor-miRNA hairpins. Germline loss-of-function DICER1 mutations underline the development of DICER1 syndrome, a rare genetic disorder that predisposes children to cancer development in organs such as lung, gynecologic tract, kidney, and brain. Unlike classical tumor suppressors, the somatic "second hit" in DICER1 syndrome-associated cancers does not fully inactivate DICER1 but impairs its RNase IIIb activity only, suggesting a noncanonical two-hit hypothesis. Here, we developed a genetically engineered conditional compound heterozygous Dicer1 mutant mouse strain that fully recapitulates the biallelic DICER1 mutations in DICER1 syndrome-associated human cancers. Crossing this tool strain with tissue-specific Cre strains that activate Dicer1 mutations in gynecologic tract cells at two distinct developmental stages revealed that embryonic biallelic Dicer1 mutations caused infertility in females by disrupting oviduct and endometrium development and ultimately drove cancer development. These multicystic tubal and intrauterine tumors histologically resembled a subset of DICER1 syndrome-associated human cancers. Molecular analysis uncovered accumulation of additional oncogenic events (e.g., aberrant p53 expression, Kras mutation, and Myc activation) in murine Dicer1 mutant tumors and validated miRNA biogenesis defects in 5P miRNA strand production, of which, loss of let-7 family miRNAs was identified as a putative key player in transcriptomic rewiring and tumor development. Thus, this DICER1 syndrome-associated cancer model recapitulates the biology of human cancer and provides a unique tool for future investigation and therapeutic development. SIGNIFICANCE: Generation of a Dicer1 mutant mouse model establishes the oncogenicity of missense mutations in the DICER1 RNase IIIb domain and provides a faithful model of DICER1 syndrome-associated cancer for further investigation.


Subject(s)
MicroRNAs , Neoplastic Syndromes, Hereditary , Child , Humans , Female , Animals , Mice , Ribonuclease III/genetics , Ribonuclease III/metabolism , MicroRNAs/genetics , Mutation , Mutation, Missense , DEAD-box RNA Helicases/genetics
10.
Nat Commun ; 14(1): 1677, 2023 03 25.
Article in English | MEDLINE | ID: mdl-36966138

ABSTRACT

DICER1 syndrome is a tumor predisposition syndrome that is associated with up to 30 different neoplastic lesions, usually affecting children and adolescents. Here we identify a group of mesenchymal tumors which is highly associated with DICER1 syndrome, and molecularly distinct from other DICER1-associated tumors. This group of DICER1-associated mesenchymal tumors encompasses multiple well-established clinicopathological tumor entities and can be further divided into three clinically meaningful classes designated "low-grade mesenchymal tumor with DICER1 alteration" (LGMT DICER1), "sarcoma with DICER1 alteration" (SARC DICER1), and primary intracranial sarcoma with DICER1 alteration (PIS DICER1). Our study not only provides a combined approach to classify DICER1-associated neoplasms for improved clinical management but also suggests a role for global hypomethylation and other recurrent molecular events in sarcomatous differentiation in mesenchymal tumors with DICER1 alteration. Our results will facilitate future investigations into prognostication and therapeutic approaches for affected patients.


Subject(s)
Neoplastic Syndromes, Hereditary , Sarcoma , Child , Adolescent , Humans , Germ-Line Mutation , Sarcoma/genetics , Neoplastic Syndromes, Hereditary/genetics , Genomics , Ribonuclease III/genetics , Genetic Predisposition to Disease , Rare Diseases , Mutation , DEAD-box RNA Helicases/genetics
11.
Mod Pathol ; 36(3): 100044, 2023 03.
Article in English | MEDLINE | ID: mdl-36788095

ABSTRACT

High-grade endometrial stromal sarcomas (HGESSs) are aggressive uterine tumors harboring oncogenic fusion proteins. We performed a molecular study of 36 HGESSs with YWHAE::NUTM2 gene fusion, assessing co-occurring genetic events, and showed that these tumors frequently harbor recurrent events involving the CDKN2A locus on chromosome 9p. Using array-based copy number profiling and CDKN2A fluorescence in situ hybridization, we identified homozygous and hemizygous deletions of CDKN2A in 18% and 14% of tumors (n = 22 analyzed), respectively. While all YWHAE-rearranged HGESSs with retained disomy for CDKN2A were immunohistochemically positive for p16INK4 (p16), all tumors with homozygous deletion of CDKN2A showed complete absence of p16 staining. Of the 2 tumors with a hemizygous deletion of CDKN2A, 1 showed diffuse and strong p16 positivity, whereas the other showed complete absence of staining. In the p16-negative case, we did not find intragenic mutations or DNA promoter methylation to explain the p16 protein loss, implicating other mechanisms in the regulation of protein expression. In our cohort, subclonal or complete absence of p16 staining was associated with worse overall survival compared with positive p16 staining (1-year overall survival: 28.6% vs 90.7%, respectively; n = 32; P < .001), with all 7 patients in the p16-negative group having succumbed to their disease within 2 years of diagnosis. Our results suggested CDKN2A alterations as a cooperative driver of tumorigenesis in a subset of HGESSs with the YWHAE::NUTM2 gene fusion and showed p16 to be a potential prognostic marker.


Subject(s)
Endometrial Neoplasms , Sarcoma, Endometrial Stromal , Sarcoma , Female , Humans , Endometrial Neoplasms/pathology , Prognosis , In Situ Hybridization, Fluorescence , Sarcoma, Endometrial Stromal/genetics , Sarcoma, Endometrial Stromal/pathology , Homozygote , Sequence Deletion , Sarcoma/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Gene Fusion , 14-3-3 Proteins/genetics , 14-3-3 Proteins/metabolism
12.
Tumori ; 109(1): 112-120, 2023 Feb.
Article in English | MEDLINE | ID: mdl-34724840

ABSTRACT

BACKGROUND: Vaginal cancer is a rare disease for which prospective randomized trials do not exist. We aimed to assess survival outcomes, patterns of recurrence, prognostic factors, and toxicity in the curative treatment using image-guided radiotherapy (RT). METHODS: In this retrospective review, we identified 53 patients who were treated at a single center with external beam radiotherapy and brachytherapy with or without concomitant chemotherapy from 2000 to 2021. RESULTS: With a median follow-up of 64.5 months, the Kaplan-Meier 2-, 5-, and 7-year overall survival (OS) was found to be 74.8%, 62.8%, and 58.9%, respectively. Local and distant control were 67.8%, 65.0%, and 65.0% and 74.4%, 62.6%, and 62.6% at 2, 5, and 7 years, respectively. In univariate Cox proportional hazards ratio analysis, OS was significantly correlated to FIGO stage (hazard ratio [HR] 1.78, p = 0.042), postoperative RT (HR 0.41, p = 0.044), and concomitant chemotherapy (HR 0.31, p = 0.009). Local control rates were superior when an equivalent dose in 2-Gy fractions (EQD2) of ⩾65 Gy was delivered (HR 0.216, p = 0.028) and with the use of concurrent chemotherapy (HR 0.248, p = 0.011). Not surprisingly, local control was inferior for patients with a higher TNM stage (HR 3.303, p = 0.027). Minimal toxicity was observed with no patients having documentation of high-grade toxicity (CTCAE grade 3+). CONCLUSION: In treatment of vaginal cancer, high-dose RT in combination with brachytherapy is well tolerated and results in effective local control rates, which significantly improve with an EQD2(α/ß=10) ⩾65 Gy. Multivariate analyses revealed concomitant chemotherapy was a positive prognostic factor for overall and progression-free survival.


Subject(s)
Brachytherapy , Uterine Cervical Neoplasms , Vaginal Neoplasms , Female , Humans , Prognosis , Vaginal Neoplasms/drug therapy , Vaginal Neoplasms/radiotherapy , Prospective Studies , Radiotherapy Dosage , Brachytherapy/adverse effects , Retrospective Studies , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/radiotherapy , Neoplasm Recurrence, Local/etiology , Uterine Cervical Neoplasms/radiotherapy
13.
Br J Cancer ; 128(1): 137-147, 2023 01.
Article in English | MEDLINE | ID: mdl-36323878

ABSTRACT

BACKGROUND: Recently, we showed a >60% difference in 5-year survival for patients with tubo-ovarian high-grade serous carcinoma (HGSC) when stratified by a 101-gene mRNA expression prognostic signature. Given the varied patient outcomes, this study aimed to translate prognostic mRNA markers into protein expression assays by immunohistochemistry and validate their survival association in HGSC. METHODS: Two prognostic genes, FOXJ1 and GMNN, were selected based on high-quality antibodies, correlation with protein expression and variation in immunohistochemical scores in a preliminary cohort (n = 134 and n = 80, respectively). Six thousand four hundred and thirty-four (FOXJ1) and 5470 (GMNN) formalin-fixed, paraffin-embedded ovarian neoplasms (4634 and 4185 HGSC, respectively) represented on tissue microarrays from the Ovarian Tumor Tissue Analysis consortium underwent immunohistochemical staining and scoring, then univariate and multivariate survival analysis. RESULTS: Consistent with mRNA, FOXJ1 protein expression exhibited a linear, increasing association with improved overall survival in HGSC patients. Women with >50% expression had the most favourable outcomes (HR = 0.78, 95% CI 0.67-0.91, p < 0.0001). GMNN protein expression was not significantly associated with overall HSGC patient survival. However, HGSCs with >35% GMNN expression showed a trend for better outcomes, though this was not significant. CONCLUSION: We provide foundational evidence for the prognostic value of FOXJ1 in HGSC, validating the prior mRNA-based prognostic association by immunohistochemistry.


Subject(s)
Cystadenocarcinoma, Serous , Ovarian Neoplasms , Humans , Female , Ovarian Neoplasms/pathology , Prognosis , Survival Analysis , RNA, Messenger/genetics , Cystadenocarcinoma, Serous/pathology , Biomarkers, Tumor/analysis , Forkhead Transcription Factors/genetics
14.
J Cancer Res Clin Oncol ; 149(7): 3361-3369, 2023 Jul.
Article in English | MEDLINE | ID: mdl-35939115

ABSTRACT

PURPOSE: Preoperative risk stratification of newly diagnosed endometrial carcinoma (EC) patients has been hindered by only moderate prediction performance for many years. Recently ENDORISK, a Bayesian network model, showed high predictive performance. It was the aim of this study to validate ENDORISK by applying the model to a population-based case series of EC patients. METHODS: ENDORISK was applied to a retrospective cohort of women surgically treated for EC from 2003 to 2013. Prediction accuracy for LNM as well as 5-year DSS was investigated. The model's overall performance was quantified by the Brier score, discriminative performance by area under the curve (AUC). RESULTS: A complete dataset was evaluable from 247 patients. 78.1% cases were endometrioid histotype. The majority of patients (n = 156;63.2%) had stage IA disease. Overall, positive lymph nodes were found in 20 (8.1%) patients. Using ENDORISK predicted probabilities, most (n = 156;63.2%) patients have been assigned to low or very low risk group with a false-negative rate of 0.6%. AUC for LNM prediction was 0.851 [95% confidence interval (CI) 0.761-0.941] with a Brier score of 0.06. For 5-year DSS the AUC was 0.698 (95% CI 0.595-0.800) as Brier score has been calculated 0.09. CONCLUSIONS: We were able to successfully validate ENDORISK for prediction of LNM and 5-year DSS. Next steps will now have to focus on ENDORISK performance in daily clinical practice. In addition, incorporating TCGA-derived molecular subtypes will be of key importance for future extended use. This study may support further promoting of data-based decision-making tools for personalized treatment of EC.


Subject(s)
Endometrial Neoplasms , Humans , Female , Prognosis , Retrospective Studies , Bayes Theorem , Endometrial Neoplasms/pathology , Risk Assessment , Lymph Nodes/pathology
15.
Am J Surg Pathol ; 46(8): 1142-1152, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35499168

ABSTRACT

The distinction between low-grade and high-grade endometrial stromal sarcomas (LGESS, HGESS) is increasingly defined by genetics. Recently, variant genomic alterations involving BCOR or BCORL1 have been reported in endometrial stromal sarcoma (ESS), although it remains unclear whether these justify a diagnosis of LGESS or HGESS. In this study, we describe clinicopathologic and molecular features of ESS with such alterations to help clarify their classification in the spectrum of ESS. We collected a cohort of 13 ESS harboring variant alteration involving BCOR (6 with internal tandem duplication, 1 with EP300::BCOR fusion, 1 with BCOR::LPP fusion) and BCORL1 ( 4 with JAZF1::BCORL1 fusion, 1 with EPC1::BCORL1 fusion). The median patient age at primary diagnosis was 51 years (range: 18 to 70 y). Median tumor size at primary diagnosis was 9.3 cm (range: 4.5 to 21 cm), and extrauterine disease spread (stage IIIB-C) was present in 27%. The tumors were composed of round to spindled cells with cellularity and cytologic atypia ranging from mild to marked and a median mitotic count of 18/10 HPFs (range: 2 to 85/10 HPFs). At least focally myopermeative growth was noted in 8/8 assessable cases. Of 12 patients with follow-up data (median: 25 mo), 4 patients died of disease and 3 were alive with recurrent disease. Unsupervised hierarchical clustering of DNA methylation data together with a large cohort of uterine mesenchymal tumors that included YWHAE::NUTM2 and Z C3H7B::BCOR HGESS and molecularly confirmed LGESS revealed a common methylation signature for all ESS with variant BCOR and BCORL1 alterations and HGESS with YWHAE::NUTM2 and ZC3H7B::BCOR gene fusion. Copy number analysis revealed amplifications of CDK4 and MDM2 , as well as homozygous deletions of CDKN2A/B and NF1 in a subset of tumors. Our results indicate that ESS with BCOR internal tandem duplication and variant BCOR and BCORL1 rearrangements clinically and molecularly resemble conventional HGESS.


Subject(s)
Endometrial Neoplasms , Proto-Oncogene Proteins , Repressor Proteins , Sarcoma, Endometrial Stromal , Sarcoma , Soft Tissue Neoplasms , Uterine Neoplasms , Cyclin-Dependent Kinase 4 , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Female , Gene Fusion , Humans , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-mdm2/genetics , Repressor Proteins/genetics , Sarcoma, Endometrial Stromal/genetics , Sarcoma, Endometrial Stromal/pathology , Uterine Neoplasms/pathology
16.
Basic Clin Pharmacol Toxicol ; 130(4): 542-552, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35040273

ABSTRACT

Pancreatic surgery is complicated by untreated fluid leakage, but no tenable techniques exist to detect and close leakage sites during surgery. A novel hydrogel called SmartPAN has been developed to meet this need and is here assessed for safety before trials on human patients. First, resazurin assays were used to test the cytotoxic effects of SmartPAN's active bromothymol blue (BTB) indicator and its solution of phosphate-buffered saline (PBS) on normal (HPDE: human pancreatic duct epithelial) or carcinomic (FAMPAC) human pancreatic cells. Cells incubated with BTB showed no significant reduction in cell viability below threshold safety levels. However, PBS had a mild cytotoxic effect on FAMPAC cells. Second, SmartPAN's pathological effects were evaluated in vivo by applying 4-ml SmartPAN to a porcine (Sus scrofa domesticus) model of pancreatic resection. There were no significant differences in macroscopic and microscopic pathologies between pigs treated with SmartPAN or saline. Third, measurements using HPLC-MS/MS demonstrate that BTB does not cross into the bloodstream and was eliminated from the body within 2 days of surgery. Overall, SmartPAN appears safe in the short term and ready for first-in-human trials because its components are either biocompatible or quickly neutralized by dilution and drainage.


Subject(s)
Pancreatic Fistula , Tandem Mass Spectrometry , Drainage/adverse effects , Drainage/methods , Humans , Pancreas/surgery , Pancreatic Fistula/complications , Pancreatic Fistula/etiology , Postoperative Complications/diagnosis , Postoperative Complications/etiology , Risk Factors
17.
Int J Gynecol Pathol ; 41(4): 417-422, 2022 07 01.
Article in English | MEDLINE | ID: mdl-34166279

ABSTRACT

We report a spindle cell sarcoma arising in the uterine corpus of a 26-yr-old patient. The patient underwent a simple hysterectomy and the uterine corpus contained a 9 cm tumor showing nodular and "finger-like" myometrial invasion. Histologically, the tumor was composed of a monomorphic population of atypical spindle cells which contained widespread foci of cytologically benign adipocytes. Immunohistochemistry revealed diffuse strong positivity for CD10, CD56, and CD99 and diffuse weak positive staining with ER and WT1 while smooth muscle markers, S100, TLE1, and cyclin D1 were negative. Total RNA sequencing identified an in-frame fusion between exon 6 of MEIS1 and exon 12 of NCOA2. Copy number analysis revealed few aberrations with no deletions or amplifications identified. No adjuvant therapy was given and the patient is disease-free 9 yr after initial diagnosis. This case represents the second report of a uterine sarcoma harboring a MEIS1-NCOA2/1 gene fusion and expands the morphologic spectrum of recently reported spindle cell sarcomas arising in the genitourinary tract harboring MEIS1-NCOA2/1 gene fusions. This is the first reported case of such tumors with an adipocytic component.


Subject(s)
Sarcoma , Soft Tissue Neoplasms , Uterine Neoplasms , Adult , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Female , Gene Fusion , Humans , Immunohistochemistry , Metaplasia , Myeloid Ecotropic Viral Integration Site 1 Protein/genetics , Nuclear Receptor Coactivator 2/genetics , Sarcoma/genetics , Sarcoma/pathology , Soft Tissue Neoplasms/pathology , Uterine Neoplasms/genetics
18.
Virchows Arch ; 480(4): 855-871, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34782936

ABSTRACT

Tubo-ovarian high-grade serous carcinomas (HGSC) are highly proliferative neoplasms that generally respond well to platinum/taxane chemotherapy. We recently identified minichromosome maintenance complex component 3 (MCM3), which is involved in the initiation of DNA replication and proliferation, as a favorable prognostic marker in HGSC. Our objective was to further validate whether MCM3 mRNA expression and possibly MCM3 protein levels are associated with survival in patients with HGSC. MCM3 mRNA expression was measured using NanoString expression profiling on formalin-fixed and paraffin-embedded tissue (N = 2355 HGSC) and MCM3 protein expression was assessed by immunohistochemistry (N = 522 HGSC) and compared with Ki-67. Kaplan-Meier curves and the Cox proportional hazards model were used to estimate associations with survival. Among chemotherapy-naïve HGSC, higher MCM3 mRNA expression (one standard deviation increase in the score) was associated with longer overall survival (HR = 0.87, 95% CI 0.81-0.92, p < 0.0001, N = 1840) in multivariable analysis. MCM3 mRNA expression was highest in the HGSC C5.PRO molecular subtype, although no interaction was observed between MCM3, survival and molecular subtypes. MCM3 and Ki-67 protein levels were significantly lower after exposure to neoadjuvant chemotherapy compared to chemotherapy-naïve tumors: 37.0% versus 46.4% and 22.9% versus 34.2%, respectively. Among chemotherapy-naïve HGSC, high MCM3 protein levels were also associated with significantly longer disease-specific survival (HR = 0.52, 95% CI 0.36-0.74, p = 0.0003, N = 392) compared to cases with low MCM3 protein levels in multivariable analysis. MCM3 immunohistochemistry is a promising surrogate marker of proliferation in HGSC.


Subject(s)
Cystadenocarcinoma, Serous , Minichromosome Maintenance Complex Component 3 , Ovarian Neoplasms , Biomarkers, Tumor/analysis , Cell Proliferation , Cystadenocarcinoma, Serous/pathology , Female , Humans , Ki-67 Antigen , Minichromosome Maintenance Complex Component 3/genetics , Ovarian Neoplasms/pathology , RNA, Messenger , Survival Rate
19.
Mod Pathol ; 34(12): 2122-2129, 2021 12.
Article in English | MEDLINE | ID: mdl-34312479

ABSTRACT

Undifferentiated mesenchymal tumors arising from the inner lining (intima) of large arteries are classified as intimal sarcomas (ISA) with MDM2 amplification as their molecular hallmark. Interestingly, undifferentiated pleomorphic sarcomas (UPS) of the heart have recently been suggested to represent the cardiac analog of ISA due to morphological overlap and high prevalence of MDM2 amplifications in both neoplasms. However, little is known about ISAs and cardiac UPS without MDM2 amplifications and molecular data supporting their common classification is sparse. Here, we report a series of 35 cases comprising 25 ISAs of the pulmonary artery, one ISA of the renal artery and 9 UPS of the left atrium. Tumors were analyzed utilizing the Illumina Infinium MethylationEPIC BeadChip array, enabling copy number profile generation and unsupervised DNA methylation analysis. DNA methylation patterns were investigated using t-distributed stochastic neighbor embedding (t-SNE) analysis. Histologically, all ISAs and UPS of the left atrium resembled extra-cardiac UPS. All cases exhibited highly complex karyotypes with overlapping patterns between ISA and UPS. 29/35 cases showed mutually exclusive amplifications in the cell-cycle associated oncogenes MDM2 (25/35), MDM4 (2/35), and CDK6 (2/35). We further observed recurrent co-amplifications in PDGFRA (21/35), CDK4 (15/35), TERT (11/35), HDAC9 (9/35), and CCND1 (4/35). Sporadic co-amplifications occurred in MYC, MYCN, and MET (each 1/35). The tumor suppressor CDKN2A/B was frequently deleted (10/35). Interestingly, DNA methylation profiling (t-SNE) revealed an overlap of ISA and cardiac UPS. This "ISA" methylation signature was distinct from potential histologic and molecular mimics. In conclusion, our data reveal MDM4 and CDK6 amplifications in ISAs and UPS of the left atrium, lacking MDM2 amplification. We further report novel co-amplifications of various oncogenes, which may have therapeutic implications. Finally, the genetic and epigenetic concordance of ISAs and UPS of the left atrium further supports a shared pathogenesis and common classification.


Subject(s)
Cell Cycle Proteins/genetics , Cyclin-Dependent Kinase 6/genetics , DNA Methylation/genetics , DNA, Neoplasm/genetics , Heart Neoplasms/genetics , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins/genetics , Sarcoma/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Cell Differentiation , DNA Copy Number Variations , Female , Gene Amplification , Genome-Wide Association Study , Heart Neoplasms/pathology , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , Neoplasm Proteins/genetics , Sarcoma/pathology , Tunica Intima/pathology , Young Adult
20.
J Dtsch Dermatol Ges ; 19(6): 842-851, 2021 Jun.
Article in German | MEDLINE | ID: mdl-34139087

ABSTRACT

HINTERGRUND UND ZIELE: Systeme künstlicher Intelligenz (durch "deep learning" faltende neuronale Netzwerke; engl. convolutional neural networks, CNN) erreichen inzwischen bei der Klassifikation von Hautläsionen vergleichbar gute Ergebnisse wie Dermatologen. Allerdings müssen die Limitationen solcher Systeme vor flächendeckendem klinischem Einsatz bekannt sein. Daher haben wir den Einfluss des "dunklen Rand-Artefakts" (engl. dark corner artefact; DCA) in dermatoskopischen Bildern auf die diagnostische Leistung eines CNN mit Marktzulassung zur Klassifikation von Hautläsionen untersucht. PATIENTEN UND METHODEN: Ein Datensatz aus 233 Bildern von Hautläsionen (60 maligne und 173 benigne) ohne DCA (Kontrolle) wurde digital so modifiziert, dass kleine, mittlere oder große DCA zu sehen waren. Alle 932 Bilder wurden dann mittels CNN mit Marktzulassung (Moleanalyzer-Pro® , FotoFinder Systems) auf Malignitätsscores hin analysiert. Das Spektrum reichte von 0-1; ein Score von > 0,5 wurde als maligne klassifiziert. ERGEBNISSE: In der Kontrollserie ohne DCA erreichte das CNN eine Sensitivität von 90,0 % (79,9 %-95,3 %), eine Spezifität von 96,5 % (92,6 %-98,4 %) sowie eine Fläche unter der Kurve (AUC, area under the curve) der "receiver operating characteristic" (ROC) von 0,961 (0,932-0,989). In den Datensätzen mit kleinen beziehungsweise mittleren DCA war die diagnostische Leistung vergleichbar. In den Bildersätzen mit großen DCA wurden allerdings signifikant höhere Malignitätsscores erzielt. Dies führte zu einer signifikant verminderten Spezifität (87,9 % [82,2 %-91,9 %], P < 0,001) sowie einer nicht signifikant erhöhten Sensitivität (96,7 % [88,6 %-99,1 %]). Die ROC-AUC blieb mit 0,962 (0,935-0,989) unverändert. SCHLUSSFOLGERUNGEN: Die Klassifizierung mittels des CNN war bei dermatoskopischen Bildern mit kleinen oder mittleren DCA nicht beeinträchtigt, das System zeigte jedoch Schwächen bei großen DCA. Wenn Ärzte solche Bilder zur Klassifikation mittels CNN einreichen, sollten sie sich dieser Grenzen der Technologie bewusst sein.

SELECTION OF CITATIONS
SEARCH DETAIL