Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 157
Filter
1.
Chirurgie (Heidelb) ; 94(10): 845-849, 2023 Oct.
Article in German | MEDLINE | ID: mdl-37432477

ABSTRACT

BACKGROUND: The gold standard in the treatment of mucinous intra-abdominal neoplasms is cytoreductive surgery (CRS) combined with hyperthermic intraperitoneal chemotherapy (HIPEC). Despite complete cytoreduction up to 45% of patients develop recurrences. METHOD: A search and analysis of the current literature were carried out. RESULTS: There is still controversy regarding the best treatment strategy for patients with recurrent pseudomyxoma peritonei (PMP) after CRS and HIPEC. The clinical management of these patients depends on many factors, such as the site and volume of recurrence, histological subtype and symptoms. Treatment options range from repeated surgery with curative intent with or without HIPEC to watch and wait strategies. In selected patients redo surgery is feasible and safe with low morbidity and mortality. Iterative complete CRS can result in a median 5­year overall survival of more than 80%. Debulking surgery leads to a prolonged survival and to symptom control fora period with of nearly 2 years. CONCLUSION: Repeated complete cytoreduction of recurrent PMP can result in long-term survival. Tumor debulking surgery may be particularly beneficial for symptomatic patients.


Subject(s)
Appendiceal Neoplasms , Hyperthermia, Induced , Neoplasms, Cystic, Mucinous, and Serous , Peritoneal Neoplasms , Pseudomyxoma Peritonei , Humans , Pseudomyxoma Peritonei/surgery , Pseudomyxoma Peritonei/drug therapy , Peritoneal Neoplasms/surgery , Peritoneal Neoplasms/drug therapy , Appendiceal Neoplasms/pathology , Appendiceal Neoplasms/therapy , Neoplasms, Cystic, Mucinous, and Serous/therapy
3.
Am J Surg Pathol ; 46(1): e27-e42, 2022 01 01.
Article in English | MEDLINE | ID: mdl-33417324

ABSTRACT

Appendiceal mucinous neoplasms show a range of morphologic features and biological risk. At one end of the spectrum, high-grade adenocarcinomas are cytologically malignant with infiltrative invasion, lymph node metastases, and behavior similar to that of extra-appendiceal mucinous adenocarcinomas. At the other end, mucinous neoplasms confined to the mucosa are uniformly benign. Some cases lying between these extremes have potential risk to metastasize within the abdomen despite a lack of malignant histologic features. They show "diverticulum-like," pushing invasion of mostly low-grade epithelium through the appendix with, or without, concomitant organizing intra-abdominal mucin. The latter condition, widely termed "pseudomyxoma peritonei," tends to pursue a relentless course punctuated by multiple recurrences despite cytoreductive therapy, culminating in death for many patients. The combination of bland histologic features and protracted behavior of peritoneal disease has led some authors to question whether these metastatic tumors even represent malignancies. The World Health Organization and its cadre of experts widely promote usage of "low-grade appendiceal mucinous neoplasm" as an umbrella term to encompass benign and malignant conditions, as well as those that have uncertain biological potential. Although this practice greatly simplifies tumor classification, it causes confusion and consternation among pathologists, clinical colleagues, and patients. It also increases the likelihood that at least some patients will undergo unnecessary surveillance for, and treatment of, benign neoplasms and non-neoplastic conditions. The purpose of this review is to critically evaluate the relevant literature and discuss a practical approach to classifying appendiceal mucinous neoplasms that more closely approximates their biological risk.


Subject(s)
Appendiceal Neoplasms/pathology , Appendix/pathology , Neoplasms, Cystic, Mucinous, and Serous/pathology , Pseudomyxoma Peritonei/pathology , Appendiceal Neoplasms/chemistry , Appendiceal Neoplasms/therapy , Appendix/chemistry , Biomarkers, Tumor/analysis , Biopsy , Clinical Decision-Making , Humans , Neoplasm Grading , Neoplasms, Cystic, Mucinous, and Serous/chemistry , Neoplasms, Cystic, Mucinous, and Serous/therapy , Predictive Value of Tests , Pseudomyxoma Peritonei/chemistry , Pseudomyxoma Peritonei/therapy , Risk Assessment , Risk Factors
4.
Inflammation ; 44(6): 2203-2216, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34091823

ABSTRACT

Gasdermins (GSDM) are members of a family of pore-forming effector proteins which lead to membrane permeabilization and pyroptosis, a lytic cell death with pro-inflammatory characteristics. Recently, two members of the gasdermin family, gasdermin B (GSDMB) and gasdermin E (GSDME), were shown to suppress tumor growth, through the involvement of cytotoxic lymphocytes. Other studies also reported the important functions of gasdermins in various cancer types including gastric cancer, hepatocarcinoma, and cervix and breast cancer. However, gasdermins have not been previously studied in the context of serous ovarian cancer. Here, we showed that gasdermin D (GSDMD) and gasdermin C (GSDMC) expression increases in serous ovarian cancer; in contrast, the expression of GSDME and PJVK (Pejvakin, DFNB59) is downregulated, compared to healthy ovaries, in multiple independent gene expression datasets. We found that copy number gains are highly frequent (present in approximately 50% of patients) in genes encoding GSDMD and GSDMC in ovarian cancer, in line with their upregulated expression in serous ovarian cancer. Moreover, we observed that the expression of GSDMB and GSDMD, but not of GSDME, is different among several histotypes of epithelial ovarian cancer. Therefore, we propose that differential expression and copy number variations of certain gasdermins might be associated with the development of serous ovarian cancer, in which different members of the family have distinct functions; however, further research is required in in vivo models to understand how changes in gasdermin family members mechanistically contribute to serous ovarian cancer.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Ovarian Epithelial/genetics , DNA Copy Number Variations , Gene Dosage , Neoplasms, Cystic, Mucinous, and Serous/genetics , Ovarian Neoplasms/genetics , Pyroptosis , Carcinoma, Ovarian Epithelial/mortality , Carcinoma, Ovarian Epithelial/pathology , Carcinoma, Ovarian Epithelial/therapy , Case-Control Studies , Databases, Genetic , Female , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Humans , Neoplasms, Cystic, Mucinous, and Serous/mortality , Neoplasms, Cystic, Mucinous, and Serous/pathology , Neoplasms, Cystic, Mucinous, and Serous/therapy , Nerve Tissue Proteins/genetics , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Phenotype , Phosphate-Binding Proteins/genetics , Pore Forming Cytotoxic Proteins/genetics , Progression-Free Survival
5.
J Pathol Clin Res ; 7(6): 548-555, 2021 11.
Article in English | MEDLINE | ID: mdl-34138519

ABSTRACT

Ovarian carcinoma histotypes are distinct diseases with variable clinical outcomes and response to treatment. There is a need for new subtype-specific treatment modalities, especially for women with widespread and chemo-resistant disease. Stimulator of interferon genes (STING) is a part of the cGAS-STING pathway that mediates innate immune defence against infectious DNA-containing pathogens and also detects tumour-derived DNA and generates intrinsic antitumour immunity. The STING signalling pathway is suppressed by several mechanisms in a variety of malignant diseases and, in some cancers that may be a requirement for cellular transformation. The aim of this study was to use immunohistochemistry to evaluate STING protein expression across normal tissue, paratubal and ovarian cysts, and ovarian tumour histotypes including ovarian carcinomas. Herein, we show that the fallopian tube ciliated cells express STING protein, whereas the secretory cells are negative. STING expression differs among ovarian cancer histotypes; low-grade serous ovarian carcinomas and serous borderline tumours have uniform high STING expression, while high-grade serous and endometrioid carcinomas have heterogeneous expression, and clear cell and mucinous carcinomas show low expression. As low-grade serous carcinomas are known to be genomically stable and typically lack a prominent host immune response, the consistently high STING expression is unexpected. High STING expression may reflect pathway activation or histogenesis and the mechanisms may be different in different ovarian carcinoma histotypes. Further studies are needed to determine whether the STING signalling pathway is active and whether these tumours would be candidates for therapeutic interventions that trigger innate immunity activation.


Subject(s)
Biomarkers, Tumor/analysis , Membrane Proteins/analysis , Neoplasms, Cystic, Mucinous, and Serous/chemistry , Ovarian Neoplasms/chemistry , Female , Humans , Immunity, Innate , Immunotherapy , Neoplasm Grading , Neoplasms, Cystic, Mucinous, and Serous/immunology , Neoplasms, Cystic, Mucinous, and Serous/pathology , Neoplasms, Cystic, Mucinous, and Serous/therapy , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Signal Transduction
8.
World Neurosurg ; 147: e32-e39, 2021 03.
Article in English | MEDLINE | ID: mdl-33321250

ABSTRACT

BACKGROUND: Brain metastases from endometrial cancer are rare and poorly described. We aimed to estimate the proportion of brain metastases at our institution that arose from endometrial cancer, and to detail clinicopathologic features and survival outcomes. METHODS: We retrospectively identified and reviewed the charts of 30 patients with brain metastases from endometrial cancer seen at Stanford Hospital from 2008 to 2018. RESULTS: Among all patients with brain metastases, the proportion arising from endometrial cancer was 0.84%. The median age at diagnosis was 62 years (range, 39-79 years), and the median overall survival from brain metastasis diagnosis was 6.8 months (range, 1.0-58.2 months). Most patients harbored endometrioid histology (53.3%), and some had concurrent metastases to lung (50.0%), bone (36.7%), and liver (20.0%). The median time from endometrial cancer diagnosis to brain metastasis development was 20.8 months (range, 1.4 months to 11.2 years), and the median number of brain metastases was 2 (range, 1-20). Patients with non-endometrioid histologies had more brain metastases than those with endometrioid histology (6.21 vs. 2.44, P = 0.029). There was no difference in overall survival by histology. CONCLUSIONS: We describe the largest cohort to date of patients with brain metastases originating from endometrial cancer. These patients represent a small fraction of all patients with brain metastases and have poor prognoses. These data enable providers caring for patients with brain metastases from endometrial cancer to appropriately counsel their patients.


Subject(s)
Brain Neoplasms/secondary , Carcinoma, Endometrioid/secondary , Carcinosarcoma/secondary , Endometrial Neoplasms/pathology , Neoplasms, Cystic, Mucinous, and Serous/secondary , Adult , Aged , Asymptomatic Diseases , Ataxia/physiopathology , Bone Neoplasms/secondary , Brain Neoplasms/physiopathology , Brain Neoplasms/therapy , Carcinoma, Endometrioid/physiopathology , Carcinoma, Endometrioid/therapy , Carcinosarcoma/physiopathology , Carcinosarcoma/therapy , Cranial Nerve Diseases/physiopathology , Female , Headache/physiopathology , Humans , Karnofsky Performance Status , Liver Neoplasms/secondary , Lung Neoplasms/secondary , Metastasectomy , Middle Aged , Muscle Weakness/physiopathology , Neoplasm Grading , Neoplasms, Cystic, Mucinous, and Serous/physiopathology , Neoplasms, Cystic, Mucinous, and Serous/therapy , Neurosurgical Procedures , Radiosurgery , Survival Rate , Time Factors
9.
Int Immunopharmacol ; 91: 107274, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33360087

ABSTRACT

Treatment of serous ovarian cancer (SOC) remains a clinical challenge. Classification of SOC based on immunogenomic profiling is important for establishing immunotherapy strategies. We extracted RNA-seq data of SOC from TCGA-OV. The samples were ultimately classified into high immune (Immunity_H) group and low immune (Immunity_L) group based on the immunogenomic profiling of 29 immune signatures by using unsupervised machine learning methods and modified by multifaceted characterization of immune response. High immune group showed the lower tumor purity and higher anti-tumor immune activity, and the higher expressions of PDCD1, CD274 and CTLA4. Furthermore, the overall survival time and the progression-free interval were significantly longer in high-immun group. The differentially expressed genes were mainly enriched in some immune response related functional terms and PI3K-AKT signaling pathway. According to ImmuCellAI, the abundance of various T cell subtypes in high immune group were significantly higher than those in low immune group. This novel immunotyping shows promise for prognostic and immunotherapeutic stratification in SOC patients.


Subject(s)
Biomarkers, Tumor/genetics , Gene Expression Profiling , Immunophenotyping , Neoplasms, Cystic, Mucinous, and Serous/genetics , Ovarian Neoplasms/genetics , Transcriptome , Tumor Microenvironment/immunology , Aged , Clinical Decision-Making , Computational Biology , Databases, Genetic , Female , Humans , Immunotherapy , Lymphocytes, Tumor-Infiltrating/immunology , Middle Aged , Neoplasms, Cystic, Mucinous, and Serous/classification , Neoplasms, Cystic, Mucinous, and Serous/immunology , Neoplasms, Cystic, Mucinous, and Serous/therapy , Ovarian Neoplasms/classification , Ovarian Neoplasms/immunology , Ovarian Neoplasms/therapy , Predictive Value of Tests , Progression-Free Survival , RNA-Seq , T-Lymphocytes/immunology
10.
Mod Pathol ; 34(3): 613-626, 2021 03.
Article in English | MEDLINE | ID: mdl-32759977

ABSTRACT

Mucinous ovarian tumors rarely harbor mural nodules, which have historically been classified as sarcoma-like, anaplastic carcinomatous, or sarcomatous on the basis of predominant morphologic features. The molecular relationship between mural nodules and associated mucinous ovarian tumors remains poorly characterized, as does the molecular pathogenesis of these mural nodules. Thus, we analyzed the morphological, immunohistochemical, and genetic features of 13 mucinous ovarian tumors and associated mural nodule(s). Three harbored sarcoma-like mural nodules and ten contained anaplastic carcinomatous nodules, including 1 tumor with spatially discrete anaplastic carcinomatous and sarcomatous nodules. Twelve of 13 cases showed genetic evidence of clonality between the mural nodule(s) and associated mucinous ovarian tumor, including all three tumors with sarcoma-like morphology. Mural nodules were genetically identical in the five cases in which there were multiple discrete mural nodules that were sequenced separately. MTAP and p53 immunohistochemistry confirmed the distribution of neoplastic cells in a subset of sarcoma-like and anaplastic carcinomatous nodules. No single recurrent genetic alteration was associated with mural nodule development. No recurrent genetic differences were identified between mural nodules with sarcoma-like, anaplastic carcinomatous, and sarcomatous morphology. Of 11 patients with clinical follow-up, three died of disease 3, 8, and 9 months after diagnosis, but no recurrent genetic events were associated with poor outcome. These molecular data suggest that sarcoma-like, anaplastic carcinomatous, and sarcomatous nodules represent a morphologic spectrum of clonal neoplasms arising in mucinous ovarian tumors rather than three discrete biological entities.


Subject(s)
Biomarkers, Tumor , High-Throughput Nucleotide Sequencing , Immunohistochemistry , Neoplasms, Cystic, Mucinous, and Serous , Ovarian Neoplasms , Adolescent , Adult , Aged , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Biopsy , Female , Humans , Microtubule-Associated Proteins/analysis , Microtubule-Associated Proteins/genetics , Middle Aged , Neoplasms, Cystic, Mucinous, and Serous/chemistry , Neoplasms, Cystic, Mucinous, and Serous/genetics , Neoplasms, Cystic, Mucinous, and Serous/pathology , Neoplasms, Cystic, Mucinous, and Serous/therapy , Ovarian Neoplasms/chemistry , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Predictive Value of Tests , Prognosis , Tumor Suppressor Protein p53/analysis , Tumor Suppressor Protein p53/genetics
11.
Mod Pathol ; 34(1): 194-206, 2021 01.
Article in English | MEDLINE | ID: mdl-32724153

ABSTRACT

TP53 mutations are implicated in the progression of mucinous borderline tumors (MBOT) to mucinous ovarian carcinomas (MOC). Optimized immunohistochemistry (IHC) for TP53 has been established as a proxy for the TP53 mutation status in other ovarian tumor types. We aimed to confirm the ability of TP53 IHC to predict TP53 mutation status in ovarian mucinous tumors and to evaluate the association of TP53 mutation status with survival among patients with MBOT and MOC. Tumor tissue from an initial cohort of 113 women with MBOT/MOC was stained with optimized IHC for TP53 using tissue microarrays (75.2%) or full sections (24.8%) and interpreted using established criteria as normal or abnormal (overexpression, complete absence, or cytoplasmic). Cases were considered concordant if abnormal IHC staining predicted deleterious TP53 mutations. Discordant tissue microarray cases were re-evaluated on full sections and interpretational criteria were refined. The initial cohort was expanded to a total of 165 MBOT and 424 MOC for the examination of the association of survival with TP53 mutation status, assessed either by TP53 IHC and/or sequencing. Initially, 82/113 (72.6%) cases were concordant using the established criteria. Refined criteria for overexpression to account for intratumoral heterogeneity and terminal differentiation improved concordance to 93.8% (106/113). In the expanded cohort, 19.4% (32/165) of MBOT showed evidence for TP53 mutation and this was associated with a higher risk of recurrence, disease-specific death, and all-cause mortality (overall survival: HR = 4.6, 95% CI 1.5-14.3, p = 0.0087). Within MOC, 61.1% (259/424) harbored a TP53 mutation, but this was not associated with survival (overall survival, p = 0.77). TP53 IHC is an accurate proxy for TP53 mutation status with refined interpretation criteria accounting for intratumoral heterogeneity and terminal differentiation in ovarian mucinous tumors. TP53 mutation status is an important biomarker to identify MBOT with a higher risk of mortality.


Subject(s)
Biomarkers, Tumor/genetics , DNA Mutational Analysis , Immunohistochemistry , Mutation , Neoplasms, Cystic, Mucinous, and Serous/genetics , Ovarian Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Adult , Australia , Female , Humans , Middle Aged , Neoplasms, Cystic, Mucinous, and Serous/mortality , Neoplasms, Cystic, Mucinous, and Serous/pathology , Neoplasms, Cystic, Mucinous, and Serous/therapy , North America , Observer Variation , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Predictive Value of Tests , Prognosis , Reproducibility of Results , Risk Assessment , Risk Factors , Tissue Array Analysis , United Kingdom
12.
J Pathol ; 253(1): 41-54, 2021 01.
Article in English | MEDLINE | ID: mdl-32901952

ABSTRACT

Low-grade serous ovarian carcinoma (LGSOC) is associated with a poor response to existing chemotherapy, highlighting the need to perform comprehensive genomic analysis and identify new therapeutic vulnerabilities. The data presented here represent the largest genetic study of LGSOCs to date (n = 71), analysing 127 candidate genes derived from whole exome sequencing cohorts to generate mutation and copy-number variation data. Additionally, immunohistochemistry was performed on our LGSOC cohort assessing oestrogen receptor, progesterone receptor, TP53, and CDKN2A status. Targeted sequencing identified 47% of cases with mutations in key RAS/RAF pathway genes (KRAS, BRAF, and NRAS), as well as mutations in putative novel driver genes including USP9X (27%), MACF1 (11%), ARID1A (9%), NF2 (4%), DOT1L (6%), and ASH1L (4%). Immunohistochemistry evaluation revealed frequent oestrogen/progesterone receptor positivity (85%), along with CDKN2A protein loss (10%) and CDKN2A protein overexpression (6%), which were linked to shorter disease outcomes. Indeed, 90% of LGSOC samples harboured at least one potentially actionable alteration, which in 19/71 (27%) cases were predictive of clinical benefit from a standard treatment, either in another cancer's indication or in LGSOC specifically. In addition, we validated ubiquitin-specific protease 9X (USP9X), which is a chromosome X-linked substrate-specific deubiquitinase and tumour suppressor, as a relevant therapeutic target for LGSOC. Our comprehensive genomic study highlighted that there is an addiction to a limited number of unique 'driver' aberrations that could be translated into improved therapeutic paths. © 2020 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma/genetics , Genomics , Neoplasms, Cystic, Mucinous, and Serous/genetics , Ovarian Neoplasms/genetics , Australia , Biomarkers, Tumor/analysis , Canada , Carcinoma/chemistry , Carcinoma/pathology , Carcinoma/therapy , DNA Copy Number Variations , DNA Mutational Analysis , Female , Gene Dosage , Genetic Predisposition to Disease , Humans , Immunohistochemistry , Mutation , Neoplasm Grading , Neoplasms, Cystic, Mucinous, and Serous/chemistry , Neoplasms, Cystic, Mucinous, and Serous/pathology , Neoplasms, Cystic, Mucinous, and Serous/therapy , Ovarian Neoplasms/chemistry , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Phenotype , Treatment Outcome , Ubiquitin Thiolesterase/genetics , Exome Sequencing
15.
Surg Oncol ; 33: 19-23, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32561085

ABSTRACT

BACKGROUND: Endometrial cancer is the most common malignancy of the female genital tract. For cancers detected at an advanced stage or intraperitoneal relapse, the prognosis is poor. Optimal cytoreductive surgery (CRS) is the most accepted treatment; however, patients with advanced intraperitoneal disease might benefit from hyperthermic intraoperative peritoneal chemotherapy (HIPEC). The aim of this study was to analyze recurrence-free survival (RFS) after CRS and HIPEC in a large series of patients with peritoneal metastases from endometrial cancer. METHODS: Patients with a diagnosis of endometrial cancer with primary or recurrent peritoneal dissemination were included. All patients underwent CRS plus HIPEC. Data were prospectively collected in the Spanish Group of Peritoneal Oncological Surgery (GECOP) database. RESULTS: Forty-three patients with endometrial cancer and peritoneal metastasis were included. Fifteen patients (35%) were diagnosed with G3 endometrioid carcinomas and 28 (65%) with other non-endometroid histologies. A completeness of cytoreduction score of CC-0 was achieved in 41 patients (95%). RFS at 5 years was 23%, being factors related to worse RFS: treatment with preoperative chemotherapy (p = 0.027), resection of more than three peritoneal areas (p = 0.010), cytoreduction of the upper abdominal space (p = 0.023), HIPEC treatment with paclitaxel (p = 0.013), and the presence of metastatic lymph nodes (p = 0.029). CONCLUSIONS: Better RFS rates after CRS and HIPEC were observed for patients with the following characteristics: cytoreductive surgery without preoperative chemotherapy, complete surgery performed with limited surgical maneuvers, treated with cisplatin, and no lymph node metastases. SYNOPSIS: Endometrial cancer has a poor prognosis when diagnosed at advance stage. Patients with intraperitoneal metastases from endometrial cancer may benefit from CRS plus HIPEC with improvement in the recurrence-free survival results.


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma, Endometrioid/therapy , Cisplatin/administration & dosage , Cytoreduction Surgical Procedures , Endometrial Neoplasms/therapy , Hyperthermic Intraperitoneal Chemotherapy , Neoplasms, Cystic, Mucinous, and Serous/therapy , Peritoneal Neoplasms/therapy , Aged , Carcinoma, Endometrioid/secondary , Disease-Free Survival , Endometrial Neoplasms/pathology , Female , Humans , Lymph Node Excision , Middle Aged , Neoplasms, Cystic, Mucinous, and Serous/secondary , Peritoneal Neoplasms/secondary
16.
Am J Surg Pathol ; 44(8): 1005-1016, 2020 08.
Article in English | MEDLINE | ID: mdl-32452870

ABSTRACT

Endocrine mucin-producing sweat gland carcinoma (EMPSGC) is a rare, low-grade adnexal neoplasm with predilection for the periorbital skin of older women. Histologically and immunophenotypically, EMPSGC is analogous to another neoplasm with neuroendocrine differentiation, solid papillary carcinoma of the breast. Both lesions are spatially associated with neuroendocrine mucinous adenocarcinomas of the skin and breast, respectively. EMPSGC is ostensibly a precursor of neuroendocrine-type mucinous sweat gland adenocarcinoma (MSC), a lesion of uncertain prognosis. Non-neuroendocrine MSC has been deemed locally aggressive with metastatic potential, and previous works speculated that EMPSGC-associated (neuroendocrine-type) MSC had similar recurrence and metastatic potential with implications for patient follow-up. Only 96 cases of EMPSGC have been reported (12 cases in the largest case series). Herein, we present 63 cases diagnosed as "EMPSGC" in comparison with aggregated results from known published EMPSGC cases. We aim to clarify the clinicopathologic features and prognostic significance of the neuroendocrine differentiation of EMPSGC and its associated adenocarcinoma and to determine the nosological relevance of EMPSGC association in the spectrum of MSC histopathogenesis. Results established an overall female predominance (66.7%) and average presenting age of 64 years. EMPSGC lesions were associated with adjacent MSC in 33.3% of cases. The recurrence rate for neuroendocrine-type MSC was ~21%, less than the reported 30% for non-neuroendocrine MSC. There were no cases of metastasis. EMPSGC and neuroendocrine-type MSC are distinct entities with more indolent behavior than previously reported, supporting a favorable prognosis for patients.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma/pathology , Mucins/analysis , Neoplasms, Cystic, Mucinous, and Serous/pathology , Sweat Gland Neoplasms/pathology , Aged , Aged, 80 and over , Carcinoma/chemistry , Carcinoma/epidemiology , Carcinoma/therapy , Female , Humans , Male , Middle Aged , Neoplasms, Cystic, Mucinous, and Serous/chemistry , Neoplasms, Cystic, Mucinous, and Serous/epidemiology , Neoplasms, Cystic, Mucinous, and Serous/therapy , North America , Prognosis , Retrospective Studies , Sweat Gland Neoplasms/chemistry , Sweat Gland Neoplasms/epidemiology , Sweat Gland Neoplasms/therapy
17.
Am J Surg Pathol ; 44(8): 1050-1060, 2020 08.
Article in English | MEDLINE | ID: mdl-32384321

ABSTRACT

The prevalence and significance of programmed death-1 ligand (PD-L1) expression in different types of tubo-ovarian carcinoma have not been well defined. We evaluated PD-L1 expression and CD8 tumor-infiltrating lymphocyte (TIL) density in whole tissue sections of 189 cases of tubo-ovarian carcinoma, including high-grade serous carcinoma (HGSC, n=100), clear cell carcinoma (CCC, n=24), endometrioid carcinoma (EmC, n=40), and mucinous carcinomas (MC, n=25). Using the tumor proportion score (TPS) with a 1% cutoff, PD-L1 expression was present in 21% of HGSC, 16.7% of CCC, 2.5% of EmC, and 4% of MC. Using the combined positive score (CPS) with a cutoff of 1, PD-L1 expression was present in 48% of HGSC, 25% of CCC, 20% of EmC, and 24% of MC. HGSC demonstrated significantly higher CD8 TIL density than CCC (P=0.013238), EmC (P=0.01341), or MC (P=0.004556). In HGSC, CD8 TIL density was directly correlated with PD-L1 positivity using either TPS (P=0.0008) or CPS (P=0.00011). Survival analysis of patients with high stage (stage III to IV) HGSC revealed PD-L1 positivity by TPS to be associated with improved progression-free survival (adjusted hazard ratio: 0.4912 vs. 2.036, P=0.0378). Although not statistically significant, a similar trend was observed in overall survival (adjusted hazard ratio: 0.3387 vs. 2.953, P=0.0548). In contrast, with CPS, no significant difference was identified between PD-L1-positive and negative groups in either progression-free survival (P=0.5086) or overall survival (P=0.7823). Neoadjuvant chemotherapy was associated with higher PD-L1 expression by TPS (P=0.00407) but not CPS. No significant difference in PD-L1 expression was detected in tumors from patients with germline BRCA1/2 mutations compared with germline mutation-negative tumors by either TPS or CPS. In conclusion, the prevalence of PD-L1 expression is variable in different types of tubo-ovarian carcinoma and is highest in HGSC. In high-stage HGSC, PD-L1 positivity in tumor cells is associated with an increased immune response and improved survival.


Subject(s)
Adenocarcinoma, Clear Cell/immunology , B7-H1 Antigen/analysis , Biomarkers, Tumor/analysis , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Endometrioid/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasms, Cystic, Mucinous, and Serous/immunology , Ovarian Neoplasms/immunology , Adenocarcinoma, Clear Cell/mortality , Adenocarcinoma, Clear Cell/pathology , Adenocarcinoma, Clear Cell/therapy , Adult , Aged , Aged, 80 and over , Carcinoma, Endometrioid/mortality , Carcinoma, Endometrioid/pathology , Carcinoma, Endometrioid/therapy , Chemotherapy, Adjuvant , Female , Humans , Middle Aged , Neoadjuvant Therapy , Neoplasm Grading , Neoplasm Staging , Neoplasms, Cystic, Mucinous, and Serous/mortality , Neoplasms, Cystic, Mucinous, and Serous/pathology , Neoplasms, Cystic, Mucinous, and Serous/therapy , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Progression-Free Survival , Time Factors , Young Adult
18.
Virchows Arch ; 477(5): 677-685, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32472195

ABSTRACT

The objective of the present study was to perform a quantitative analysis of cancer stem cell (CSC) marker expression in ovarian carcinoma effusions. The clinical role of SSEA1 in metastatic high-grade serous carcinoma (HGSC) was additionally analyzed. CD133, Nanog, SOX2, Oct3/4, SSEA1, and SSEA4 protein expressions were quantitatively analyzed using flow cytometry (FCM) in 24 effusions. SSEA1 expression by immunohistochemistry was analyzed in 384 HGSC effusions. Highly variable expression of CSC markers by FCM was observed, ranging from 0 to 78% of Ber-EP4-positive cells in the case of CD133, with the largest number of negative specimens seen for SSEA4. SSEA1 expression by immunohistochemistry was found in HGSC cells in 336/384 (89%) effusions, most commonly focally (< 5% of cells). SSEA1 was overexpressed in post-chemotherapy disease recurrence specimens compared with chemo-naïve HGSC effusions tapped at diagnosis (p = 0.029). In univariate survival analysis, higher SSEA1 expression was significantly associated with poor overall survival (p = 0.047) and progression-free survival (p = 0.018), though it failed to retain its prognostic role in Cox multivariate survival analysis in which it was analyzed with clinical parameters (p = 0.059 and p = 0.111 for overall and progression-free survival, respectively). In conclusion, CSC markers are variably expressed in ovarian carcinoma effusions. SSEA1 expression is associated with disease progression and poor survival in metastatic HGSC. Silencing this molecule may have therapeutic relevance in this cancer.


Subject(s)
Fucosyltransferases/analysis , Lewis X Antigen/analysis , Neoplasms, Cystic, Mucinous, and Serous/enzymology , Neoplastic Stem Cells/enzymology , Ovarian Neoplasms/enzymology , AC133 Antigen/analysis , Adult , Aged , Aged, 80 and over , Female , Flow Cytometry , Humans , Immunohistochemistry , Middle Aged , Neoplasm Grading , Neoplasm Recurrence, Local , Neoplasms, Cystic, Mucinous, and Serous/mortality , Neoplasms, Cystic, Mucinous, and Serous/secondary , Neoplasms, Cystic, Mucinous, and Serous/therapy , Neoplastic Stem Cells/pathology , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Predictive Value of Tests , Progression-Free Survival , Retrospective Studies , Risk Assessment , Risk Factors , Time Factors , Young Adult
19.
Eur J Surg Oncol ; 46(9): 1697-1702, 2020 09.
Article in English | MEDLINE | ID: mdl-32204935

ABSTRACT

INTRODUCTION: Endometrial cancer (EC) known prognostic factors are not sufficient to predict either outcome or recurrence rate/site: to investigate EC recurrence patterns according to ESMO-ESGO-ESTRO risk classes, could be beneficial for a more tailored adjuvant treatment and follow-up schedule. METHODS: 758 women diagnosed with EC, and a 5-years follow-up, were enrolled: they were divided into the ESMO-ESGO-ESTRO risk classes (low LR, intermediate IR, intermediate-high I-HR, and highrisk HR) and surgically treated as recommended, followed by adjuvants therapies when appropriate. RESULTS: Higher recurrence rate (RR) was significantly detected (p < 0,001) in the HR group (40,3%) compared to LR (9,6%), IR (16,7%) and I-HR (17,1%). Recurrences were detected more frequently at distant sites (64%) compared to pelvic (25,3%) and lymph nodes (10,7%) recurrences (p < 0,0001): only in LR group, no differences were detected between local and distant recurrences. 5-Year distant-free (LR 99%, IR 94%,I-HR 86%, HR 88%) and local-free survivals (LR 99%, IR 100%,I-HR 98%, HR 95%) significantly differ between groups (p < 0,0001 and p = 0,003, respectively). Adjuvant therapy modifies RRs only in LR group (p = 0,01). CONCLUSION: To identify biological factors to stratify patients at higher risk of relapse is needed. Distant site relapse could be the main reason of endometrial cancer failure follow-up, independently or in addition to their risk class prognosis.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Endometrioid/therapy , Endometrial Neoplasms/therapy , Lymph Nodes/pathology , Neoplasm Metastasis , Neoplasm Recurrence, Local/epidemiology , Adenocarcinoma, Clear Cell/pathology , Adenocarcinoma, Clear Cell/therapy , Adult , Aged , Aged, 80 and over , Anthracyclines/administration & dosage , Brachytherapy , Carcinoma, Adenosquamous/pathology , Carcinoma, Adenosquamous/therapy , Carcinoma, Endometrioid/pathology , Chemoradiotherapy, Adjuvant , Disease-Free Survival , Endometrial Neoplasms/pathology , Female , Humans , Hysterectomy , Laparoscopy , Lymph Node Excision , Lymphatic Metastasis , Middle Aged , Neoplasm Invasiveness , Neoplasms, Cystic, Mucinous, and Serous/pathology , Neoplasms, Cystic, Mucinous, and Serous/therapy , Omentum , Peritoneal Lavage , Platinum Compounds/administration & dosage , Radiotherapy, Adjuvant , Retrospective Studies , Risk Assessment , Robotic Surgical Procedures , Salpingo-oophorectomy , Taxoids/administration & dosage
20.
Am J Surg Pathol ; 44(5): 641-648, 2020 05.
Article in English | MEDLINE | ID: mdl-32205482

ABSTRACT

Endometrial carcinoma (EC), as described by Bokhman, has historically been classified as Type I (low-grade, hormone-dependant, young patients, good prognosis) or Type II (high-grade, hormone-independent, older patients, poor prognosis). This classification is no longer pragmatic, however, as EC is a much more heterogeneous disease. Four molecular subtypes of EC were identified by The Cancer Genome Atlas (TCGA), and subsequent studies have demonstrated its utility in predicting prognosis. While endometrial serous carcinoma (ESC), the prototypical Type II EC, largely occurs in older women, younger women with ESC were not accounted for in the Bokhman model and were underrepresented in the TCGA study. We hypothesized that a subset of ESCs in young patients do not represent bona fide serous carcinomas but rather high-grade endometrioid carcinomas mimicking a serous phenotype. We identified ESCs and mixed endometrioid/serous carcinomas in women <60 years (n=37), and analyzed their clinical, morphologic, immunohistochemical, and molecular characteristics. Sixteen percent showed mismatch repair deficiency (MMR-D) and 11% were diagnosed with Lynch syndrome. Additionally, 16% of cases tested harbored a hotspot POLE exonuclease domain mutation (POLE-EDM). Morphologically, 47% of tumors showed confirmatory endometrioid features, including atypical hyperplasia, a low-grade endometrioid carcinoma component, or squamous differentiation. Clinically, the overall survival in patients with MMR-D and POLE-EDM was significantly better than that of patients without these features (P=0.0329). In conclusion, ESCs in young patients comprise a heterogeneous group of tumors, demonstrating diverse clinical, immunohistochemical, morphologic, and molecular features which have implications for prognosis and adjuvant therapy.


Subject(s)
Biomarkers, Tumor/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , DNA Mismatch Repair , DNA Polymerase II/genetics , DNA Repair Enzymes/deficiency , Endometrial Neoplasms/genetics , Mutation , Neoplasms, Complex and Mixed/genetics , Neoplasms, Cystic, Mucinous, and Serous/genetics , Poly-ADP-Ribose Binding Proteins/genetics , Adult , Age Factors , Colorectal Neoplasms, Hereditary Nonpolyposis/enzymology , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , Colorectal Neoplasms, Hereditary Nonpolyposis/therapy , Endometrial Neoplasms/enzymology , Endometrial Neoplasms/pathology , Endometrial Neoplasms/therapy , Female , Genetic Predisposition to Disease , Humans , Middle Aged , Neoplasm Grading , Neoplasms, Complex and Mixed/enzymology , Neoplasms, Complex and Mixed/pathology , Neoplasms, Complex and Mixed/therapy , Neoplasms, Cystic, Mucinous, and Serous/enzymology , Neoplasms, Cystic, Mucinous, and Serous/pathology , Neoplasms, Cystic, Mucinous, and Serous/therapy , Phenotype , Risk Factors , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...