Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 377
Filter
1.
Cochrane Database Syst Rev ; 9: CD015896, 2024 09 23.
Article in English | MEDLINE | ID: mdl-39312297

ABSTRACT

OBJECTIVES: This is a protocol for a Cochrane Review (prognosis). The objectives are as follows: To evaluate the predictive value of the prognostic factor HRD status, as determined by various clinically validated HRD assays at the time of staging laparotomy, compared to BRCA1/2 mutation status for progression-free survival and overall survival in patients with tubo-ovarian high-grade serous carcinoma treated in the first-line setting with a combination of surgery and platinum-based chemotherapy and/or maintenance with PARP inhibitors.


Subject(s)
Ovarian Neoplasms , Humans , Ovarian Neoplasms/genetics , Ovarian Neoplasms/mortality , Female , Fallopian Tube Neoplasms/genetics , Fallopian Tube Neoplasms/mortality , Fallopian Tube Neoplasms/pathology , Fallopian Tube Neoplasms/drug therapy , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/mortality , Prognosis , Progression-Free Survival , Genes, BRCA2 , Genes, BRCA1 , Mutation
2.
J Pathol ; 264(1): 42-54, 2024 09.
Article in English | MEDLINE | ID: mdl-38956451

ABSTRACT

Ovarian high-grade serous carcinoma (HGSC) originates in the fallopian tube, with secretory cells carrying a TP53 mutation, known as p53 signatures, identified as potential precursors. p53 signatures evolve into serous tubal intraepithelial carcinoma (STIC) lesions, which in turn progress into invasive HGSC, which readily spreads to the ovary and disseminates around the peritoneal cavity. We recently investigated the genomic landscape of early- and late-stage HGSC and found higher ploidy in late-stage (median 3.1) than early-stage (median 2.0) samples. Here, to explore whether the high ploidy and possible whole-genome duplication (WGD) observed in late-stage disease were determined early in the evolution of HGSC, we analysed archival formalin-fixed paraffin-embedded (FFPE) samples from five HGSC patients. p53 signatures and STIC lesions were laser-capture microdissected and sequenced using shallow whole-genome sequencing (sWGS), while invasive ovarian/fallopian tube and metastatic carcinoma samples underwent macrodissection and were profiled using both sWGS and targeted next-generation sequencing. Results showed highly similar patterns of global copy number change between STIC lesions and invasive carcinoma samples within each patient. Ploidy changes were evident in STIC lesions, but not p53 signatures, and there was a strong correlation between ploidy in STIC lesions and invasive ovarian/fallopian tube and metastatic samples in each patient. The reconstruction of sample phylogeny for each patient from relative copy number indicated that high ploidy, when present, occurred early in the evolution of HGSC, which was further validated by copy number signatures in ovarian and metastatic tumours. These findings suggest that aberrant ploidy, suggestive of WGD, arises early in HGSC and is detected in STIC lesions, implying that the trajectory of HGSC may be determined at the earliest stages of tumour development. © 2024 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
Cystadenocarcinoma, Serous , Fallopian Tube Neoplasms , Ovarian Neoplasms , Tumor Suppressor Protein p53 , Humans , Female , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/pathology , Fallopian Tube Neoplasms/genetics , Fallopian Tube Neoplasms/pathology , Tumor Suppressor Protein p53/genetics , Carcinoma in Situ/genetics , Carcinoma in Situ/pathology , Neoplasm Grading , DNA Copy Number Variations , Mutation , Genomics/methods , Whole Genome Sequencing , Ploidies , Middle Aged , Biomarkers, Tumor/genetics , Disease Progression
4.
Gynecol Oncol ; 187: 241-248, 2024 08.
Article in English | MEDLINE | ID: mdl-38833993

ABSTRACT

BACKGROUND: Endometrial serous carcinoma (ESC) and tubo-ovarian high-grade serous carcinoma (HGSC) are characterized by late-stage presentation and high mortality. Current guidelines for prevention recommend risk-reducing salpingo-oophorectomy (RRSO) in patients with hereditary mutations in cancer susceptibility genes. However, HGSC displays extensive genetic heterogeneity with alterations in 168 genes identified in TCGA study, but current germline testing panels are often limited to the handful of recurrently mutated genes, leaving families with rare hereditary gene mutations potentially at-risk. OBJECTIVE: To determine if there are rare germline mutations that may aid in early identification of more patients at-risk for ESC and/or HGSC by evaluating patients with concurrent ESC, HGSC or precursor lesions, and endometrial atypical hyperplasia (CAH) or low-grade endometrial endometrioid adenocarcinoma (LGEEA). METHODS: We performed targeted next-generation sequencing using TSO 500, a 523 gene panel, on formalin-fixed paraffin-embedded tumor and matched benign non-tumor tissue blocks from 5 patients with concurrent ESC, HGSC or precursor lesions, and CAH or LGEEA. RESULTS: We identified germline pathogenic, likely pathogenic or uncertain significance variants in cancer susceptibility genes in 4 of 5 patients - affected genes included GLI1, PIK3R1, FOXP1, FANCD2, INPP4B and H3F3C. Notably, none of these genes were included in the commercially available germline testing panels initially used to evaluate the patients at the time of their diagnoses. CONCLUSION: Comprehensive germline testing of patients with concurrent LGEEA or CAH and ESC, HGSC or precursor lesions may aid in early identification of relatives at-risk for cancer who may be candidates for RRSO with hysterectomy.


Subject(s)
Carcinoma, Endometrioid , Endometrial Neoplasms , Germ-Line Mutation , High-Throughput Nucleotide Sequencing , Ovarian Neoplasms , Humans , Female , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Carcinoma, Endometrioid/genetics , Carcinoma, Endometrioid/pathology , Middle Aged , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/pathology , Aged , Fallopian Tube Neoplasms/genetics , Fallopian Tube Neoplasms/pathology , Genetic Predisposition to Disease , Adult
5.
Gynecol Oncol ; 187: 113-119, 2024 08.
Article in English | MEDLINE | ID: mdl-38759517

ABSTRACT

OBJECTIVE: The majority of high-grade serous carcinomas (HGSC) of the ovary, fallopian tube, and peritoneum arise from the precursor lesion called serous tubal intraepithelial carcinoma (STIC). It has been postulated that cells from STICs exfoliate into the peritoneal cavity and give rise to peritoneal HGSC several years later. While co-existent STICs and HGSCs have been reported to share similarities in their mutational profiles, clonal relationship between temporally distant STICs and HGSCs have been infrequently studied and the natural history of STICs remains poorly understood. METHODS: We performed focused searches in two national databases from the Netherlands and identified a series of BRCA1/2 germline pathogenic variant (GPV) carriers (n = 7) who had STIC, and no detectable invasive carcinoma, at the time of their risk-reducing salpingo-oophorectomy (RRSO), and later developed peritoneal HGSC. The clonal relationship between these STICs and HGSCs was investigated by comparing their genetic mutational profile by performing next-generation targeted sequencing. RESULTS: Identical pathogenic mutations and loss of heterozygosity of TP53 were identified in the STICs and HGSCs of five of the seven patients (71%), confirming the clonal relationship of the lesions. Median interval for developing HGSC after RRSO was 59 months (range: 24-118 months). CONCLUSION: Our results indicate that cells from STIC can shed into the peritoneal cavity and give rise to HGSC after long lag periods in BRCA1/2 GPV carriers, and argues in favor of the hypothesis that STIC lesions may metastasize.


Subject(s)
Cystadenocarcinoma, Serous , Fallopian Tube Neoplasms , Ovarian Neoplasms , Salpingo-oophorectomy , Humans , Female , Fallopian Tube Neoplasms/genetics , Fallopian Tube Neoplasms/pathology , Fallopian Tube Neoplasms/surgery , Fallopian Tube Neoplasms/prevention & control , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/pathology , Cystadenocarcinoma, Serous/surgery , Cystadenocarcinoma, Serous/prevention & control , Middle Aged , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Ovarian Neoplasms/surgery , Ovarian Neoplasms/prevention & control , Adult , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/pathology , Carcinoma in Situ/genetics , Carcinoma in Situ/pathology , Carcinoma in Situ/surgery , Germ-Line Mutation , Genes, BRCA2 , BRCA2 Protein/genetics , BRCA1 Protein/genetics , Genes, BRCA1
6.
Gynecol Oncol ; 186: 110-116, 2024 07.
Article in English | MEDLINE | ID: mdl-38640774

ABSTRACT

OBJECTIVE: Recent evidence suggests that the fimbriated end of the fallopian tube harbors the precursor cells for many high-grade ovarian cancers, opening the door for development of better screening methods that directly assess the fallopian tube in women at risk for malignancy. Previously we have shown that the karyometric signature is abnormal in the fallopian tube epithelium in women at hereditary risk of ovarian cancer. In this study, we sought to determine whether the karyometric signature in serous tubal intraepithelial carcinoma (STIC) is significantly different from normal, and whether an abnormal karyometric signature can be detected in histologically normal tubal epithelial cells adjacent to STIC lesions. METHODS: The karyometric signature was measured in epithelial cells from the proximal and fimbriated portion of the fallopian tube in fallopian tube specimens removed from women at: 1) average risk for ovarian cancer undergoing surgery for benign gynecologic indications (n = 37), 2) hereditary risk of ovarian cancer (germline BRCA alterations) undergoing risk-reducing surgery (n = 44), and 3) diagnosed with fimbrial STICs (n = 17). RESULTS: The karyometric signature in tubes with fimbrial STICs differed from that of tubes with benign histology. The degree of karyometric alteration increased with increasing proximity to fimbrial STICs, ranging from moderate in the proximal portion of the tube, to greatest in both normal appearing fimbrial cells near STICs as well as in fimbrial STIC lesions. CONCLUSION: These data demonstrate an abnormal karyometric signature in STICs that may extend beyond the STIC, potentially providing an opportunity for early detection of fallopian tube neoplasia.


Subject(s)
Carcinoma in Situ , Fallopian Tube Neoplasms , Fallopian Tubes , Humans , Female , Fallopian Tube Neoplasms/pathology , Fallopian Tube Neoplasms/genetics , Carcinoma in Situ/pathology , Carcinoma in Situ/genetics , Fallopian Tubes/pathology , Cystadenocarcinoma, Serous/pathology , Cystadenocarcinoma, Serous/genetics , Middle Aged , Adult , Ovarian Neoplasms/pathology , Ovarian Neoplasms/genetics , Karyotype
7.
Am J Surg Pathol ; 48(8): 1017-1023, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38639044

ABSTRACT

Most extrauterine high-grade serous carcinomas (HGSCs) are thought to develop first in the distal fallopian tube. Most models of HGSC assume origin from relatively stable, noninvasive serous tubal intraepithelial carcinomas. However, widespread tumor involvement in the absence of a serous tubal intraepithelial carcinoma could occur after catastrophic genomic events (CGEs; such as chromothripsis or polyploidy). Twenty-six HGSCs assigned to fallopian tube (n = 9, group 1) and/or ovary (n = 9, group 2), and primary peritoneal (n = 8, group 3) were assessed by microarray (Oncoscan). CGEs were identified in 15/26 (57.7%); chromothripsis-like pattern in 13/26 (50.0%) and polyploidy in 6/26 (23.1%). CGE was seen in 4/9 (44.4%), 9/9 (100%), and 2/8 (25%) cases in groups 1. 2, and 3, respectively. Overall, CGEs were seen in 9/9 (100%) cases with grossly evident ovarian parenchymal involvement versus 6/17 (35.3%) without ( P = 0.0024). Ovarian size (measured on the long axis) correlated with CGE positivity ( P = 0.016). CGEs are significantly more common in HGSCs with ovarian parenchymal involvement compared with those limited to the fallopian tube and/or extraovarian tissues. These associations suggest geographically different tumor growth patterns and support the subdivision of HGSCs according to not only the stage but also tumor distribution. They have implications for clinical and pathologic presentation, trajectory of tumor evolution, and in the case of primary peritoneal HGSCs, potentially unique precursors to tumor transitions that could inform or influence cancer prevention efforts.


Subject(s)
Chromothripsis , Cystadenocarcinoma, Serous , Fallopian Tube Neoplasms , Neoplasm Grading , Ovarian Neoplasms , Peritoneal Neoplasms , Polyploidy , Humans , Female , Fallopian Tube Neoplasms/pathology , Fallopian Tube Neoplasms/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/pathology , Cystadenocarcinoma, Serous/pathology , Cystadenocarcinoma, Serous/genetics , Middle Aged , Aged , Adult , Oligonucleotide Array Sequence Analysis , Aged, 80 and over
8.
J Gynecol Oncol ; 35(4): e55, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38330378

ABSTRACT

OBJECTIVE: To determine the useful biomarker for predicting the effects of poly-(ADP ribose)-polymerase (PARP) inhibitors in Japanese patients with ovarian cancer. METHODS: We collected clinical information and performed molecular biological analysis on 42 patients with ovarian, fallopian tube, and primary peritoneal carcinomas who received PARP inhibitors. RESULTS: Among the analyzed patients with ovarian cancer, 23.8% had germline BRCA mutation (gBRCAm), 42.9% had homologous recombination repair-related gene mutation (HRRm), and 61.1% had a genomic instability score (GIS) of ≥42. Patients with HRRm had a significantly longer progression-free survival (PFS) than those without HRRm (median PFS 35.6 vs. 7.9 months; p=0.009), with a particularly marked increase in PFS in patients with gBRCAm (median PFS 42.3 months). Similarly, among patients with recurrent ovarian cancer, those with HRRm had a longer PFS than those without HRRm (median PFS 42.3 vs. 7.7 months; p=0.040). Multivariate Cox proportional hazards regression analysis found that performance status and gBRCAm status were independent factors associated with prolonged PFS with PARP inhibitors. In recurrent ovarian cancer, multivariate regression analysis identified platinum-free interval (PFI) in addition to performance status as a significant predictor of PFS. On the contrary, no significant association was observed between PFS and a GIS of ≥42 used in clinical practice. CONCLUSION: We found that HRRm can be a useful biomarker for predicting the effects of PARP inhibitors in treating ovarian cancer and that the PFI can also be useful in recurrent ovarian cancer.


Subject(s)
Biomarkers, Tumor , Ovarian Neoplasms , Poly(ADP-ribose) Polymerase Inhibitors , Progression-Free Survival , Humans , Female , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/mortality , Middle Aged , Aged , Biomarkers, Tumor/genetics , Adult , Germ-Line Mutation , Genomic Instability , Aged, 80 and over , Fallopian Tube Neoplasms/drug therapy , Fallopian Tube Neoplasms/genetics , Fallopian Tube Neoplasms/mortality , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/drug therapy , Peritoneal Neoplasms/drug therapy , Peritoneal Neoplasms/genetics , Recombinational DNA Repair/genetics , Recombinational DNA Repair/drug effects
9.
Pathol Res Pract ; 255: 155183, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38364651

ABSTRACT

Epithelial ovarian cancers (EOC) associated with germline or somatic BRCA pathogenetic variants have a significantly higher rate of TP53aberrations. The majority of TP53 mutations are detectable by immunohistochemistry and several studies demonstrated that an abnormal p53 pattern characterized high-grade EOCs. An abnormal p53 immunohistochemical staining in fallopian tube (serous tubal intraepithelial carcinoma (STIC) and "p53 signature" is considered as a precancerous lesion of high-grade EOCs and it is often found in fallopian tube tissues of BRCA germline mutated patients suggesting that STIC is an early lesion and the TP53 mutation is an early driver event of BRCA mutated high-grade EOCs. No relevant data are present in literature about the involvement of p53 abnormal pattern in EOC carcinogenesis of patients negative for germline BRCA variants. We describe TP53 mutation results in relationship to the immunohistochemical pattern of p53 expression in a series of EOCs negative for BRCA1 and BRCA2 germline mutations. In addition, we also investigated STIC presence and "p53 signature" in fallopian tube sampling of these EOCs. Our results demonstrate that TP53 alterations are frequent and early events in sporadic EOCs including also low-grade carcinomas. Also in this series, STIC is associated with an abnormal p53 pattern in fallopian tubes of high-grade EOCs. In summary, TP53 aberrations are the most frequent and early molecular events in EOC carcinogenesis independently from BRCA mutation status.


Subject(s)
Cystadenocarcinoma, Serous , Fallopian Tube Neoplasms , Ovarian Neoplasms , Humans , Female , Carcinoma, Ovarian Epithelial/genetics , Carcinoma, Ovarian Epithelial/pathology , BRCA1 Protein/analysis , Germ-Line Mutation , Ovarian Neoplasms/pathology , Tumor Suppressor Protein p53/metabolism , BRCA2 Protein/analysis , Fallopian Tubes/chemistry , Fallopian Tubes/metabolism , Fallopian Tubes/pathology , Fallopian Tube Neoplasms/genetics , Fallopian Tube Neoplasms/metabolism , Fallopian Tube Neoplasms/pathology , Cystadenocarcinoma, Serous/pathology , Mutation , Carcinogenesis/pathology , Germ Cells/pathology
10.
Am J Surg Pathol ; 48(4): 475-486, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38298022

ABSTRACT

Serous tubal intraepithelial carcinoma (STIC) is the fallopian tube precursor lesion for most cases of pelvic high-grade serous carcinoma (HGSC). To date, the morphologic, molecular, and clinical heterogeneity of STIC and a less atypical putative precursor lesion, termed serous tubal intraepithelial lesion, has not been well characterized. Better understanding of precursor heterogeneity could impact the clinical management of women with incidental STICs (without concurrent carcinoma) identified in cases of prophylactic or opportunistic salpingectomy. This study analyzed morphologic and molecular features of 171 STICs and 21 serous tubal intraepithelial lesions. We assessed their histologic features, Ki-67 and p53 staining patterns, and genome-wide DNA copy number alterations. We classified all precursor lesions into 2 morphologic subtypes, one with a flat surface (Flat) and the other characterized by budding, loosely adherent, or detached (BLAD) morphology. On the basis of pathology review by a panel of 8 gynecologic pathologists, we found 87 BLAD, 96 Flat, and 9 indeterminate lesions. As compared with Flat lesions, BLAD lesions were more frequently diagnostic of STIC ( P <0.0001) and were found concurrently with HGSC ( P <0.0001). BLAD morphology was also characterized by higher Ki-67 proliferation index ( P <0.0001), presence of epithelial stratification ( P <0.0001), and increased lymphocyte density ( P <0.0001). BLAD lesions also exhibited more frequent DNA copy number gain/amplification at the CCNE1 or CMYC loci canonical to HGSCs ( P <0.0001). Both BLAD morphology and STIC diagnoses are independent risk factors for an elevated Ki-67 proliferation index. No correlation was observed between BLAD and Flat lesions with respect to patient age, presence of germline BRCA1/2 mutation, or p53 staining pattern. These findings suggest that tubal precursor lesions are morphologically and molecularly heterogeneous, laying the foundation for further studies on the pathogenesis of HGSC initiation and identifying histologic features predictive of poor patient outcomes.


Subject(s)
Adenocarcinoma in Situ , Carcinoma in Situ , Carcinoma , Cystadenocarcinoma, Serous , Fallopian Tube Neoplasms , Ovarian Neoplasms , Female , Humans , BRCA1 Protein , Carcinoma in Situ/genetics , Carcinoma in Situ/pathology , Ovarian Neoplasms/pathology , Ki-67 Antigen , Tumor Suppressor Protein p53/genetics , BRCA2 Protein , Fallopian Tube Neoplasms/genetics , Fallopian Tube Neoplasms/pathology , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/pathology , DNA
11.
Clin Cancer Res ; 30(3): 600-615, 2024 02 01.
Article in English | MEDLINE | ID: mdl-38048050

ABSTRACT

PURPOSE: Serous tubal intraepithelial carcinoma (STIC) is now recognized as the main precursor of ovarian high-grade serous carcinoma (HGSC). Other potential tubal lesions include p53 signatures and tubal intraepithelial lesions. We aimed to investigate the extent and pattern of aneuploidy in these epithelial lesions and HGSC to define the features that characterize stages of tumor initiation and progression. EXPERIMENTAL DESIGN: We applied RealSeqS to compare genome-wide aneuploidy patterns among the precursors, HGSC (cases, n = 85), and histologically unremarkable fallopian tube epithelium (HU-FTE; control, n = 65). On the basis of a discovery set (n = 67), we developed an aneuploidy-based algorithm, REAL-FAST (Repetitive Element AneupLoidy Sequencing Fallopian Tube Aneuploidy in STIC), to correlate the molecular data with pathology diagnoses. We validated the result in an independent validation set (n = 83) to determine its performance. We correlated the molecularly defined precursor subgroups with proliferative activity and histology. RESULTS: We found that nearly all p53 signatures lost the entire Chr17, offering a "two-hit" mechanism involving both TP53 and BRCA1 in BRCA1 germline mutation carriers. Proliferatively active STICs harbor gains of 19q12 (CCNE1), 19q13.2, 8q24 (MYC), or 8q arm, whereas proliferatively dormant STICs show 22q loss. REAL-FAST classified HU-FTE and STICs into 5 clusters and identified a STIC subgroup harboring unique aneuploidy that is associated with increased proliferation and discohesive growth. On the basis of a validation set, REAL-FAST showed 95.8% sensitivity and 97.1% specificity in detecting STIC/HGSC. CONCLUSIONS: Morphologically similar STICs are molecularly distinct. The REAL-FAST assay identifies a potentially "aggressive" STIC subgroup harboring unique DNA aneuploidy that is associated with increased cellular proliferation and discohesive growth. REAL-FAST offers a highly reproducible adjunct technique to assist the diagnosis of STIC lesions.


Subject(s)
Carcinoma in Situ , Cystadenocarcinoma, Serous , Fallopian Tube Neoplasms , Ovarian Neoplasms , Humans , Female , Tumor Suppressor Protein p53/genetics , Ovarian Neoplasms/pathology , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/pathology , Fallopian Tubes/pathology , Fallopian Tube Neoplasms/genetics , Carcinoma in Situ/pathology
12.
Virchows Arch ; 484(2): 339-351, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38099957

ABSTRACT

Gynecologic pathology has moved, within only a few years, from being a diagnostic area devoid of molecular testing into a diagnostic discipline in which such analyses are becoming routine. The direct relevance of molecular characterization to the choice of treatment of patients with carcinomas originating in both the uterus and adnexae makes it likely that such testing will only expand along with our understanding of the molecular make-up of these tumors. As a consequence, gynecologic pathologists have become an integral part of patient management, rather than lab personnel providing external services.In parallel, molecular testing is expanding as a tool for diagnosing rare tumors affecting these organs, including soft tissue tumors, sex cord-stromal tumors and germ cell tumors, as well as other rare entities. Increased knowledge in this area bears directly on the ability to diagnose these tumors in a reproducible manner, as well as recognize and consult on genetic diseases. Hopefully, despite the inherent difficulty in studying rare cancers, it will also translate into new therapeutic options for the malignant ones among these rare cancers.


Subject(s)
Carcinoma , Fallopian Tube Neoplasms , Ovarian Neoplasms , Humans , Female , Fallopian Tubes/pathology , Carcinoma/pathology , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/genetics , Ovarian Neoplasms/therapy , Endometrium/pathology , Fallopian Tube Neoplasms/diagnosis , Fallopian Tube Neoplasms/genetics , Fallopian Tube Neoplasms/therapy
13.
Cell Death Dis ; 14(9): 644, 2023 09 30.
Article in English | MEDLINE | ID: mdl-37775701

ABSTRACT

Ovarian cancer is the leading cause of death from gynecologic cancer worldwide. High-grade serous carcinoma (HGSC) is the most common and deadliest subtype of ovarian cancer. While the origin of ovarian tumors is still debated, it has been suggested that HGSC originates from cells in the fallopian tube epithelium (FTE), specifically the epithelial cells in the region of the tubal-peritoneal junction. Three main lesions, p53 signatures, STILs, and STICs, have been defined based on the immunohistochemistry (IHC) pattern of p53 and Ki67 markers and the architectural alterations of the cells, using the Sectioning and Extensively Examining the Fimbriated End Protocol. In this study, we performed an in-depth proteomic analysis of these pre-neoplastic epithelial lesions guided by mass spectrometry imaging and IHC. We evaluated specific markers related to each preneoplastic lesion. The study identified specific lesion markers, such as CAVIN1, Emilin2, and FBLN5. We also used SpiderMass technology to perform a lipidomic analysis and identified the specific presence of specific lipids signature including dietary Fatty acids precursors in lesions. Our study provides new insights into the molecular mechanisms underlying the progression of ovarian cancer and confirms the fimbria origin of HGSC.


Subject(s)
Cystadenocarcinoma, Serous , Fallopian Tube Neoplasms , Ovarian Neoplasms , Female , Humans , Fallopian Tubes , Fallopian Tube Neoplasms/genetics , Fallopian Tube Neoplasms/chemistry , Fallopian Tube Neoplasms/pathology , Tumor Suppressor Protein p53 , Proteomics , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/pathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology
14.
J Gynecol Obstet Hum Reprod ; 52(8): 102642, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37573025

ABSTRACT

OBJECTIVE: Serous tubal intraepithelial carcinoma (STIC) is a precursor lesion which is located in the distal fallopian tube and causes high grade serous ovarian carcinoma (HGSOC). The incidence of STIC for women underwent risk reducing salpingo-oophorectomy for BRCA mutation varies from 0.6 to 7% and its clinical outcomes are still unclear. The aim of this study was to demonstrate the incidence of STIC and HGSOC in BRCA1/2 mutation carriers after risk reducing salpingo-oophorectomy (RRSO) and the clinical outcomes of these patients. MATERIAL AND METHODS: We retrospectively reviewed the records of 48 BRCA1 and/or 2 mutation carriers who underwent prophylactic salpingo-oophorectomy with or without hysterectomy at the Department of Obstetrics and Gynecology, Bursa Uludag University between January 2000 and January 2022. INCLUSION CRITERIA: BRCA 1 and/or 2 mutation carriers diagnosed by genetic testing, asymptomatic patients with no abnormal findings on pelvic examination. EXCLUSION CRITERIA: patients with no abnormal findings on pelvic examination and a presence of a personal history of ovarian, fallopian tube or peritoneal cancer. RESULTS: A total of 48 BRCA 1 and/or 2 mutation carriers underwent RRSO. STIC was diagnosed in 1 (2,0%) patient and restaging surgery was not performed. Primary peritoneal carcinoma (PPC) did not develop during the 20 months follow-up period. One (2.0%) patient was diagnosed with occult ovarian cancer. Restaging surgery was performed and chemotherapy treatments were given after surgery. A pelvic recurrence developed 25 months after the occult cancer diagnosis in the follow up period. One (2.0%) patient with normal histopathological findings after RRSO was diagnosed with peritoneal cancer 57 months after the operation. CONCLUSION: The risk of PPC continues after RRSO. Therefore, close follow-up procedure is very important for early diagnosis and effective treatment of patients with PPC after RRSO.


Subject(s)
Cystadenocarcinoma, Serous , Fallopian Tube Neoplasms , Peritoneal Neoplasms , Humans , Female , Salpingo-oophorectomy , BRCA1 Protein/genetics , Retrospective Studies , Clinical Relevance , BRCA2 Protein/genetics , Fallopian Tube Neoplasms/genetics , Fallopian Tube Neoplasms/surgery , Fallopian Tube Neoplasms/pathology , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/pathology , Peritoneal Neoplasms/prevention & control , Mutation
15.
Mol Carcinog ; 62(9): 1417-1427, 2023 09.
Article in English | MEDLINE | ID: mdl-37265438

ABSTRACT

Incessant ovulation is believed to be a potential cause of epithelial ovarian cancer (EOC). Our previous investigations have shown that insulin-like growth factor (IGF2) and hepatocyte growth factor (HGF) in the ovulatory follicular fluid (FF) contributed to the malignant transformation initiated by p53 mutations. Here we examined the individual and synergistic impacts of IGF2 and HGF on enhancing the malignant properties of high-grade serous carcinoma (HGSC), the most aggressive type of EOC, and its precursor lesion, serous tubal intraepithelial carcinoma (STIC). In a mouse xenograft co-injection model, we observed that FF co-injection induced tumorigenesis of STIC-mimicking cells, FE25. Co-injection with IGF2 or HGF partially recapitulated the tumorigenic effects of FF, but co-injection with both resulted in a higher tumorigenic rate than FF. We analyzed the different transformation phenotypes influenced by these FF growth signals through receptor inhibition. The IGF signal was necessary for clonogenicity, while the HGF signal played a crucial role in the migration and invasion of STIC and HGSC cells. Both signals were necessary for the malignant phenotype of anchoring-independent growth but had little impact on cell proliferation. The downstream signals responsible for these HGF activities were identified as the tyrosine-protein kinase Met (cMET)/mitogen-activated protein kinase and cMET/AKT pathways. Together with the previous finding that the FF-IGF2 could mediate clonogenicity and stemness activities via the IGF-1R/AKT/mammalian target of rapamycin and IGF-1R/AKT/NANOG pathways, respectively, this study demonstrated the cooperation of the FF-sourced IGF and HGF growth signals in the malignant transformation and progression of HGSC through both common and distinct signaling pathways. These findings help develop targeted prevention of HGSC.


Subject(s)
Cystadenocarcinoma, Serous , Fallopian Tube Neoplasms , Ovarian Neoplasms , Female , Humans , Mice , Animals , Fallopian Tubes/metabolism , Fallopian Tubes/pathology , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Follicular Fluid/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Ovarian Neoplasms/pathology , Tumor Suppressor Protein p53/genetics , Epithelial Cells/metabolism , Carcinogenesis/pathology , Carcinoma, Ovarian Epithelial/pathology , Cystadenocarcinoma, Serous/metabolism , Fallopian Tube Neoplasms/genetics , Fallopian Tube Neoplasms/metabolism , Fallopian Tube Neoplasms/pathology , Cell Transformation, Neoplastic/pathology , Mammals/metabolism
16.
Cancer Cell ; 41(6): 1103-1117.e12, 2023 06 12.
Article in English | MEDLINE | ID: mdl-37207655

ABSTRACT

Ovarian high-grade serous carcinoma (HGSC) is typically diagnosed at an advanced stage, with multiple genetically heterogeneous clones existing in the tumors long before therapeutic intervention. Herein we integrate clonal composition and topology using whole-genome sequencing data from 510 samples of 148 patients with HGSC in the prospective, longitudinal, multiregion DECIDER study. Our results reveal three evolutionary states, which have distinct features in genomics, pathways, and morphological phenotypes, and significant association with treatment response. Nested pathway analysis suggests two evolutionary trajectories between the states. Experiments with five tumor organoids and three PI3K inhibitors support targeting tumors with enriched PI3K/AKT pathway with alpelisib. Heterogeneity analysis of samples from multiple anatomical sites shows that site-of-origin samples have 70% more unique clones than metastatic tumors or ascites. In conclusion, these analysis and visualization methods enable integrative tumor evolution analysis to identify patient subtypes using data from longitudinal, multiregion cohorts.


Subject(s)
Cystadenocarcinoma, Serous , Fallopian Tube Neoplasms , Ovarian Neoplasms , Female , Humans , Ovarian Neoplasms/pathology , Phosphatidylinositol 3-Kinases/genetics , Prospective Studies , Cystadenocarcinoma, Serous/metabolism , Fallopian Tube Neoplasms/genetics
17.
J Clin Oncol ; 41(14): 2523-2535, 2023 05 10.
Article in English | MEDLINE | ID: mdl-36809028

ABSTRACT

PURPOSE: To investigate the prevalence of and clinical factors associated with high-grade serous carcinoma (HGSC) at risk-reducing salpingo-oophorectomy (RRSO) in asymptomatic BRCA1/2-pathogenic variant (PV) carriers. PATIENTS AND METHODS: We included BRCA1/2-PV carriers who underwent RRSO between 1995 and 2018 from the Hereditary Breast and Ovarian cancer in the Netherlands study. All pathology reports were screened, and histopathology reviews were performed for RRSO specimens with epithelial abnormalities or where HGSC developed after normal RRSO. We then compared clinical characteristics, including parity and oral contraceptive pill (OCP) use, for women with and without HGSC at RRSO. RESULTS: Of the 2,557 included women, 1,624 had BRCA1, 930 had BRCA2, and three had both BRCA1/2-PV. The median age at RRSO was 43.0 years (range: 25.3-73.8) for BRCA1-PV and 46.8 years (27.6-77.9) for BRCA2-PV carriers. Histopathologic review confirmed 28 of 29 HGSCs and two further HGSCs from among 20 apparently normal RRSO specimens. Thus, 24 (1.5%) BRCA1-PV and 6 (0.6%) BRCA2-PV carriers had HGSC at RRSO, with the fallopian tube identified as the primary site in 73%. The prevalence of HGSC in women who underwent RRSO at the recommended age was 0.4%. Among BRCA1/2-PV carriers, older age at RRSO increased the risk of HGSC and long-term OCP use was protective. CONCLUSION: We detected HGSC in 1.5% (BRCA1-PV) and 0.6% (BRCA2-PV) of RRSO specimens from asymptomatic BRCA1/2-PV carriers. Consistent with the fallopian tube hypothesis, we found most lesions in the fallopian tube. Our results highlight the importance of timely RRSO with total removal and assessment of the fallopian tubes and show the protective effects of long-term OCP.


Subject(s)
Carcinoma , Fallopian Tube Neoplasms , Ovarian Neoplasms , Female , Humans , Salpingo-oophorectomy , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Prevalence , Mutation , Genetic Predisposition to Disease , Ovarian Neoplasms/epidemiology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/prevention & control , Ovariectomy , Fallopian Tube Neoplasms/epidemiology , Fallopian Tube Neoplasms/genetics , Fallopian Tube Neoplasms/prevention & control
18.
Taiwan J Obstet Gynecol ; 62(1): 107-111, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36720520

ABSTRACT

OBJECTIVE: Serous tubal intraepithelial carcinoma (STIC) is a known precursor of high-grade serous ovarian cancer (HGSOC). This study aimed to evaluate the proportion of STIC in patients with HGSOC and analyze the STIC-related prognosis in patients with HGSOC. MATERIALS AND METHODS: All pathology reports at our institution that included bilateral salpingectomies of patients with HGSOC from January 2013 to December 2018 were reviewed by two experienced pathologists. The specimens from the ovaries and the salpinx including fimbria were examined. We analyzed the correlation between STIC and HGSOC and compared the clinical characteristics and STIC-related prognostic outcomes in patients with HGSOC. RESULTS: Eleven of the 76 cases were STIC. BRCA mutations were found in 16.9% of patients with HGSOC. STIC was observed in 30.0% of patients with BRCA mutations and in 14.3% of patients without BRCA mutations. The incidence of STIC in patients with BRCA mutations was approximately twice that in patients without BRCA mutations; however, the difference was not statistically significant (P = 0.231). Further, the 5-year survival rate of patients without STIC appeared to be high; nevertheless, the difference was not statistically significant (59.7% vs. 47.4%, P = 0.633). Moreover, there was no significant difference in disease-free survival rate according to STIC (36.4% vs. 33.1%, P = 0.956). CONCLUSION: STIC was identified in patients with HGSOC, and STIC incidence was prominent in HGSOC related to BRCA mutation. Although low frequency, STIC was detected in patients without BRCA mutation. Therefore, prophylactic salpingectomy may be useful for prevention of HGSOC.


Subject(s)
Adenocarcinoma in Situ , Carcinoma in Situ , Cystadenocarcinoma, Serous , Fallopian Tube Neoplasms , Ovarian Neoplasms , Humans , Female , Fallopian Tube Neoplasms/genetics , Fallopian Tube Neoplasms/surgery , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/surgery , Cystadenocarcinoma, Serous/pathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/surgery , Carcinoma in Situ/surgery , Carcinoma in Situ/pathology
19.
Int J Gynecol Pathol ; 42(1): 89-92, 2023 Jan 01.
Article in English | MEDLINE | ID: mdl-35149617

ABSTRACT

The RAD51D gene codes a protein-paralog of the RAD51 DNA recombinase, which catalyzes DNA strand exchange during homologous recombination. Similar to BRCA1 / BRCA2 , mutations in RAD51D both predispose to ovarian carcinoma and impart sensitivity to poly (ADP-ribose) polymerase inhibitors in preclinical studies. Based on cancer risk prediction models, RAD51D mutations pose a moderate-to-high risk for ovarian cancer warranting consideration for risk-reducing surgery. We report a case of serous tubal intraepithelial carcinoma in a patient undergoing risk-reducing total hysterectomy with bilateral salpingo-oophorectomy for a RAD51D pathogenic variant. The histopathologic and p53-immunophenotypic features of this lesion are similar to those reported previously in BRCA1 / BRCA2 mutation carriers and those of serous tubal intraepithelial carcinoma associated with sporadic high-grade serous carcinomas. These features include marked increase in nuclear-to-cytoplasmic ratio, loss of cell polarity, absence of ciliation, prominent nucleoli, mitotic activity, epithelial stratification, surface exfoliative changes, and complete loss of p53 staining. Although familial ovarian cancers with mutations in RAD51D -or other genes in the Fanconi anemia pathway-are much less common those with BRCA1 / BRCA2 mutations, our findings support a common phenotype for early serous cancers in this pathway.


Subject(s)
Carcinoma in Situ , Cystadenocarcinoma, Serous , Fallopian Tube Neoplasms , Ovarian Neoplasms , Female , Humans , Tumor Suppressor Protein p53/genetics , Fallopian Tube Neoplasms/genetics , Fallopian Tube Neoplasms/surgery , Fallopian Tube Neoplasms/pathology , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/surgery , Cystadenocarcinoma, Serous/pathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/surgery , Ovarian Neoplasms/pathology , Mutation , Carcinoma in Situ/genetics , Carcinoma in Situ/surgery , Carcinoma in Situ/pathology , DNA-Binding Proteins/genetics
20.
Cancer Res ; 82(24): 4528-4541, 2022 12 16.
Article in English | MEDLINE | ID: mdl-36206311

ABSTRACT

Elucidating the earliest pathogenic steps in cancer development is fundamental to improving its early detection and prevention. Ovarian high-grade serous carcinoma (HGSC), a highly aggressive cancer, mostly originates from the fallopian tube epithelium through a precursor stage, serous tubal intraepithelial carcinoma (STIC). In this study, we performed spatial transcriptomic analysis to compare STICs, carcinoma, and their matched normal fallopian tube epithelium. Several differentially expressed genes in STICs and carcinomas were involved in cancer metabolism and detected in a larger independent transcriptomic dataset of ovarian HGSCs. Among these, insulin-like growth factor binding protein-2 (IGFBP2) was found to undergo DNA hypomethylation and to be increased at the protein level in STICs. Pyrosequencing revealed an association of IGFBP2 expression with the methylation state of its proximal enhancer, and 5-azacytidine treatment increased IGFBP2 expression. In postmenopausal fallopian tubes, where most STICs are detected, IGFBP2 immunoreactivity was detected in all 38 proliferatively active STICs but was undetectable in morphologically normal tubal epithelia, including those with TP53 mutations. In premenopausal fallopian tubes, IGFBP2 expression was limited to the secretory epithelium at the proliferative phase, and estradiol treatment increased IGFBP2 expression levels. IGFBP2 knockdown suppressed the growth of IGFBP2-expressing tubal epithelial cells via inactivation of the AKT pathway. Taken together, demethylation of the proximal enhancer of IGFBP2 drives tumor development by maintaining the increased IGFBP2 required for proliferation in an otherwise estrogen-deprived, proliferation-quiescent, and postmenopausal tubal microenvironment. SIGNIFICANCE: Molecular studies of the earliest precursor lesions of ovarian cancer reveal a role of IGFBP2 in propelling tumor initiation, providing new insights into ovarian cancer development.


Subject(s)
Carcinoma in Situ , Carcinoma , Cystadenocarcinoma, Serous , Fallopian Tube Neoplasms , Ovarian Neoplasms , Humans , Female , Transcriptome , Carcinoma in Situ/genetics , Tumor Suppressor Protein p53/genetics , Fallopian Tube Neoplasms/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/pathology , Fallopian Tubes/pathology , Carcinoma/pathology , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL