Open access peer-reviewed chapter

Recent Advances in Classification and Histopathological Diagnosis of Ovarian Epithelial Malignant Tumours

Written By

Gabriela-Monica Stanc, Efthymia Souka and Christos Valavanis

Submitted: 10 July 2022 Reviewed: 13 July 2022 Published: 17 August 2022

DOI: 10.5772/intechopen.106545

From the Edited Volume

Recent Advances, New Perspectives and Applications in the Treatment of Ovarian Cancer

Edited by Michael Friedrich

Chapter metrics overview

410 Chapter Downloads

View Full Metrics

Abstract

Ovarian tumours are a heterogeneous group of neoplasms classified based on histopathologic type and grade of differentiation. They comprise a broad range of tumours from benign and borderline to malignant histotypes characterised by different histopathological, immunophenotypic and molecular features. The purpose of this chapter is to present an overview of the recent advances in the ovarian epithelial malignant tumours classification along with the histopathological, immunophenotypic and molecular diagnostic criteria highlighting areas of terminology discrepancies or changes and diagnostic challenges. These changes provide a better understanding of the ovarian tumours nature and lead to a more efficient therapeutic management of these pathological entities.

Keywords

  • ovarian cancer
  • histopathology
  • immunophenotype
  • molecular pathology

1. Introduction

Ovarian cancer is the most lethal malignancy of gynaecological cancer representing 23% of gynaecological neoplasms and the 5th most common leading cause of death in women. Most ovarian malignant neoplasms are diagnosed at an advanced stage with high recurrence rates and an overall 5-year survival rate of around 50% [1, 2, 3].

Ovarian tumours originate from ovarian or fallopian tube tissue and are divided into epithelial tumours (benign, border-line and malignant), germ cell tumours, sex cord-stromal tumours and mesenchymal tumours [4].

In this chapter, we will focus on the epithelial ovarian malignant tumours and will present an overview of the recent advances in the ovarian tumours classification along with their histopathological, immunophenotypic and molecular features.

Epithelial ovarian malignant tumours comprise a heterogeneous neoplastic disease with distinct histomorphologic features, pathogenesis, precursor lesions, immunophenotypic and molecular profiles, different biological behaviour and clinical outcomes [4]. According to the recent 2020 WHO classification based on histomorphology, immunophenotypic features and molecular alterations, epithelial ovarian malignant tumours are classified into five main types with different incidences: high-grade serous carcinoma (HGSC—70%), low-grade serous carcinoma (LGSC—5%), endometrioid (EC—10%), mucinous (MC—3%) and clear cell carcinomas (CCC—10%) [3, 4, 5, 6].

Rare histopathologic entities are seromucinous carcinoma, malignant Brenner tumour, mesonephric-like carcinoma, undifferentiated and dedifferentiated carcinomas, carcinosarcoma and mixed cell carcinoma [5]. Seromucinous carcinoma as a distinct entity, characterised by serous and endocervical-type mucinous epithelium with foci of clear cells and areas of endometrioid and squamous differentiation, has been removed from the recent 2020 WHO classification. It is now considered a subtype of endometrioid carcinoma based on immunohistochemical and molecular studies [5].

Traditional concepts of ovarian carcinogenesis assumed that ovarian cancer pathogenesis is due to Müllerian or non-Müllerian metaplasia of ovarian surface epithelium leading progressively to malignant transformation [7] or due to malignant transformation of endometriosis lesions and/or inclusion cysts [8]. Recent pathological observations and molecular studies have revealed that the majority of the high-grade serous carcinomas arise from a precursor dysplastic lesion in the fimbria of fallopian tubes, designated as STIC (Serous Tubal Intraepithelial Carcinoma), whereas low-grade serous carcinomas arise within the ovarian parenchyma from benign or borderline serous tumours [9, 10, 11]. In addition, the presence of genomic alterations in the BRCA1 and BRCA2 tumour suppressor genes and gene mutations in p53, p16, CCNE1, BRD4 and RSF1, and centrosome copy number abnormalities, in STIC and high-grade serous carcinoma, suggest a clonal histogenetic relationship between this precursor lesion and HGSC [12, 13, 14, 15]. A clonal histogenetic relationship has also been observed among endometriosis, precursor ovarian surface epithelium lesions and endometrioid and clear cell carcinomas based on mutations found in ARID1A, PIK3CA, KRAS and MET genes among these pathologic entities [16, 17, 18].

It is now well established that ovarian carcinogenesis is based on a dualistic model of pathogenesis that divides ovarian epithelial malignant tumours into two main categories, designated as type I and type II ovarian tumours [7].

Type I ovarian epithelial tumours arise from precursor lesions in the ovary such as cystadenoma or adenofibroma. These lesions can undergo malignant transformation through atypical proliferation or transformation to borderline tumours and eventually into invasive ovarian neoplasms. Type I ovarian epithelial tumours include low-grade serous carcinoma, endometrioid carcinoma, mucinous carcinoma, clear cell carcinoma and malignant Brenner tumour. They have a relatively indolent clinical course and are characterised by genomic stability, distinctive molecular profile for each histotype and low incidence of p53 mutations (10–13%), except of mucinous carcinoma, which displays high incidence of p53 mutations (64%) [19, 20].

Type II ovarian epithelial tumours arise from distal fallopian tube fimbria epithelium through dysplastic lesions (STIC) and finally towards invasive carcinomas. They show aggressive biological behaviour and comprise high-grade serous carcinoma, carcinosarcoma and dedifferentiated and undifferentiated carcinomas [621]. They are characterised by genomic instability, high incidence of p53 mutations and abnormal function of tumour suppressor genes BRCA1 and BRCA2 due to mutations or gene promoter hypermethylation [19, 20].

In the following sections, we are going to present the morphologic, immunophenotypic and molecular alterations that can be found in the five main histotypes of ovarian epithelial malignant tumours and the putative implications that may have in the clinical outcomes and targeted therapeutic interventions.

Advertisement

2. High-grade serous ovarian carcinomas (HGSCs)

High-grade serous carcinomas (HGSCs) account for 70% of ovarian carcinomas and represent the most aggressive and chemoresistant epithelial ovarian neoplasms. Most patients are postmenopausal women and admitted to the hospital at advanced clinical stage (80%) with extra-ovarian metastasis and thus with a high incidence of mortality around 70–80% globally [22].

Common predisposition factors are infertility, menopausal hormonal therapy and the hereditary breast and ovarian cancer (HBOC) syndrome characterised mostly by germline BRCA1/2 genomic alterations [23] and less frequently (2%) by germline genomic alterations in other homologous recombination repair (HRR) genes such as ATM, BRIP1, RAD51C and RAD51D [24, 25].

2.1 Pathology of ovarian high-grade serous carcinomas

These tumours are large, exophytic and usually bilateral with solid and papillary growth patterns and fluid-containing cysts. Necrosis is not uncommon and extraovarian affected sites can be observed. Occasionally a small tumour nodule can be found at the distal part of the fallopian tube fimbria.

Microscopically, HGSCs are heterogeneous tumours displaying solid, papillary, glandular and/or cribriform architectural patterns, sometimes with slit-like spaces. Recently, two histotypes have been observed, the classic type and the SET type (solid pseudoendometrioid and transitional variant) [5]. Classic type HGSCs exhibit papillary, micropapillary and solid growth patterns. The neoplastic cells demonstrate large pleomorphic nuclei with prominent nucleoli, high mitotic activity (>5 mitoses/mm2) including atypical mitoses and presence of multinucleated cells. SET type HGSCs are characterised by solid sheets of neoplastic cells mimicking endometrioid and/or transitional cell carcinomas, sometimes with bizarre cytologic features. Occasionally a micropapillary pattern can be seen and areas with geographical necrosis can be also found. These tumours are associated with a high number of tumour infiltrating lymphocytes (TILs) [5, 26]. SET pattern is more commonly correlated with germline and/or somatic BRCA1/2 mutations [26] and is more sensitive to chemotherapy and PARP inhibitors.

Based on BRCA mutation status, histomorphological patterns (classic vs. SET), STIC presence and clinical outcome, two categories of HGSC can be identified: (1) BRCA-mutated tumours exhibiting SET morphology, absence of STIC lesions, more common in young patients, more chemosensitive and responsive to PARP inhibitors with favourable prognosis and (2) tumours without BRCA genomic alterations displaying classic morphology, presence of STIC lesions, more common in old aged patients and less responsive to chemotherapy with unfavourable clinical outcome [26, 27].

The WHO 2020 classification introduces specific criteria for site assignment of HGSCs origin (fallopian tube, ovary, tubo-ovarian or peritoneal) Table 1 [5]. According to these criteria, 80% of the cases are of tubal origin, whereas peritoneal origin should be considered only after careful exclusion of STIC and absence of ovarian involvement.

Primary siteCriteria for diagnosis
Fallopian tubePresence of STIC
or
Presence of invasive fallopian tube HGSC
or
Part or entire fallopian tube length inseparable from tubo-ovarian tumor
OvaryBoth fallopian tubes separable from ovarian tumor
and
No STIC or invasive HGSC in either fallopian tube examined by SEE-FM (sectioning and extensively examining the tubal fimbria)
Tubo-ovarianFallopian tubes and ovaries not available for full examination
and
Pathological findings consistent with extrauterine HGSC
PeritonealBoth tubes and both ovaries fully examined
and
No gross or microscopic evidence of STIC or HGSC in tubes or ovaries

Table 1.

Criteria for assigning primary site in high grade serous carcinomas.

2.2 Immunophenotypic features of ovarian high-grade serous carcinomas

Immunohistochemical analysis should be performed in order to distinguish ovarian high-grade serous carcinoma from mesothelioma or other poorly differentiated carcinomas in cases of peritoneal carcinomatosis. It can also be useful in small biopsies and in post-therapy specimens.

Both classical and SET HGSC histotypes show positive immunoreactivity against CK7, p16, PAX8, WT1, ER (oestrogen receptor) and PgR (progesterone receptor), and this panel of antibodies is enough to establish an ovarian serous carcinoma diagnosis [5]. In addition, 30–50% of HGSCs can exhibit three different aberrant expression patterns for p53, strong, diffuse nuclear overexpression in >80% of cells associated with TP53 missense mutation, no expression implying p53 loss of function and diffuse cytoplasmic expression with weak nuclear intensity similar to wild-type immunoreactivity correlated with loss of function mutations disrupting the nuclear localization signal domain [28, 29]. Strong positive p53 immunoreactivity can also help to identify foci of intraepithelial carcinoma on the ovarian surface or in the fallopian tube epithelium (STIC). HGSCs also exhibit cytoplasmic and/or nuclear p16 expression along with a high Ki-67 (MIB-1) cell proliferation index (>75%) [30].

Ovarian HGSCs can be differentiated from mesotheliomas using a panel of various antibodies such as PAX8, Ber-EP4, MOC-31, ER (positive expression in ovarian serous carcinomas) and Calretinin, CK5/6 (both positive in mesotheliomas) [31].

Diagnostic problem can arise between HGSC of solid morphologic patterns and a poorly differentiated endometrioid carcinoma. In such a case serous carcinoma displays diffuse strong staining for WT1, p53 and p16 [32, 33].

Serous carcinoma of the endometrium shows negative or focally weakly positive WT1 immunoreactivity but p53 positive, therefore, metastatic serous carcinoma with this immunophenotype should be considered endometrial than ovarian origin [33]. WT1 immunopositivity is also observed in serous borderline tumours and in sex cord-stromal tumours [34, 35]. On the other hand, serous borderline and benign tumours show negative immunoreactivity for p53 [29, 33].

2.3 Molecular pathology of ovarian high-grade serous carcinomas

Massive parallel sequencing studies have revealed that ovarian HGSCs are characterised by somatic TP53 mutations more commonly in the DNA binding domain in high frequency (>95%) [36, 37]. They have also demonstrated genomic alterations in the homologous recombination repair (HRR) pathway leading to genomic instability and aneuploidy characterised by high copy number structural alterations (CNAs). CNAs can be recognised as oncogene amplifications such as CCNE1 (20%), MECOM, EMSY and MYC and deletions/breaks of tumour suppressor genes such as PTEN, RB1, RAD51B and NF1 [38, 39]. Additionally, recurrent mutations have been observed in a variety of genes such as NF1 (4%–6%), RB1 (2%–6%)and PTEN (<1%) along with structural alterations/deletions can result in genes inactivation in relatively high frequency such as 20%, 17% and 7%, respectively [38, 40]. Ovarian HGSCs display genomic alterations in BRCA1/2 genes that are involved in the homologous recombination repair (HRR) pathway. Almost 15% of HGSCs have BRCA1/2 germline mutations, 5% somatic mutations and 11% show BRCA1 promoter epigenetic silencing through CpG islands hypermethylation [38, 41]. Mutational alterations have also been observed in other HRR-related genes resulting in an HRR deficient phenotype in 50% of the cases and leading to high genomic instability [42]. These HRR-related genes include Fanconi anaemia genes (PALB2, FANCA, FANC1, FANCL, FANCC), RAD family genes (RAD50, RAD51, RAD51B, RAD51C, RAD54L), MRN complex genes [Mre11-Rad50-Nbs1(Nibrin)] and DNA damage response (DDR) genes (ATM, ATR, CHEK1, CHEK2) [42, 43]. Based on the HRR pathway status, HGSCs can be categorised into two morphologically distinct histotypes. HRR- proficient tumours demonstrate papillary, micropapillary and slit-like space architectural patterns with worse prognosis, while HRR-deficient (HRD) tumours show SET-like morphology (solid, endometrioid and transitional patterns) and improved progression-free survival due to beneficial responsiveness to platinum and poly ADP-ribose polymerase (PARP) inhibitors [26, 44]. HRR-proficient tumours are more likely to be resistant to these therapeutic interventions and are characterised by genomic alterations unrelated and mutually exclusive to BRCA1/2 pathway, such as CCNE1 gene amplification [45, 46].

According to NCCN guidelines HRD status should be tested in order to optimise the PARP inhibitor HGSCs treatment. Most of the HRD assays evaluate the status of germline or somatic HRR gene mutations and the presence of genomic instability by analysing the percentage of genomic loss of heterozygosity, telomeric allelic imbalance and genome-wide structural alterations (HRD mutation signatures) [47, 48, 49]. It should be noted that primary resistance to PARP inhibition can be observed in HGSCs with functional HRR, particularly in the presence of CCNE1 amplification. Additionally acquired resistance or decreased sensitivity to PARPi therapy can occur through HRR genes functional restoration by secondary mutations [50, 51]. In this setting, a functional assay can be used by evaluating the HRD status at RNA or protein levels [52, 53].

Other prognostic and treatment predictive biomarkers in HGSCs include tumour molecular subtyping based on transcriptional profiling that divides HGSCs into four categories (differentiated, immunoreactive, mesenchymal and proliferative), the former (differentiated and immunoreactive) with favourable biological behaviour and prognosis and the latter (mesenchymal and proliferative) with aggressive clinical course and worse prognosis [54, 55, 56]. In addition, promoter hypermethylation of TAP1 gene in 6p21.3 chromosomal locus confers an unfavourable prognosis, while an increased count of CD8+ tumour infiltrating T lymphocytes is associated with favourable outcome [57, 58, 59]. Protein expression studies on PD-L1, LAG3 and potential use of immunotherapeutic modalities on ovarian HGSCs have demonstrated modest therapeutic results and controversial prognosis [60, 61, 62]. Other dysregulated pathways in HGSCs are the PIK3CA/AKT and NOTCH pathways which can be therapeutically targeted by using PIK3CA or AKT inhibitors [63, 64], whereas HER2 overexpression/amplification (2–4% in HGSCs) has no significant impact on prognosis, albeit a finding that can be exploited for anti-HER2 targeted therapy [65, 66].

Advertisement

3. Low-grade serous ovarian carcinomas (LGSCs)

Low-grade serous carcinomas (LGSCs) are more common in younger women, with a median age of 43 years, have a better clinical course and prognosis than HGSCs and account for approximately 3–5% of epithelial ovarian tumours [67]. LSGCs originate from benign or borderline serous tumours and about 50% are associated with a borderline component. Progression of borderline serous tumour into low-grade serous carcinoma occurs in 6–7% of the cases and evolution to high grade serous carcinoma is rare. A more aggressive clinical course is associated with the presence of a borderline serous tumour component showing micropapillary histological pattern, microinvasions and bilateral ovarian presence [68].

3.1 Pathology of ovarian low-grade serous carcinomas

Macroscopically LGSCs are often bilateral and have a papillary appearance. Foci of calcification may be present. Microscopically, they display papillary, micropapillary, glandular, nested or inverted macropapillary architectural patterns- free-floating within unlined empty spaces, with a variety of invasion patterns. Neoplastic cells demonstrate low to moderate grade nuclear atypia with no pleomorphism (<3× size variation), distinct central nucleoli and relatively low mitotic activity (1–2 mitoses/mm2). Necrosit areas are rare and psammoma bodies are frequent. LGSCs are differentiated from their serous borderline tumour component by the presence of stromal invasive foci measuring >5 mm or 10 mm2 in size [5, 69].

3.2 Immunophenotypic features of ovarian low-grade serous carcinomas

LGSCs show positive immunoreactivity against CK7, PAX8, WT1, ER (oestrogen receptor) and PgR (progesterone receptor). Unlike HGSCs, they display patchy or negative expression for p16, low levels of Ki-67 proliferation index (less than 3%) and wild-type immunoreactivity for p53 [70].

3.3 Molecular pathology of ovarian low-grade serous carcinomas

LGSCs are characterised by genomic stability with low mutation rates, are not associated with BRCA germline genomic alterations and display low copy number genetic aberrations, like chromosomal loss of 1p36.33, 9p and homozygous deletions of the CDKN2A/2B locus in approximately 86% of LGSCs [71, 72].

The most common molecular alterations found in LGSCs have activated mutations of upstream regulators of MAPK signal transduction pathway like KRAS (25–54%), BRAF (8–33%), NRAS (8–26%) and ERBB2 (5–6%) [4, 73]. KRAS and BRAF mutations are early events in LGSCs evolution and can be found in 85% of benign cystadenomas and serous borderline tumours [74]. KRAS mutations are associated with aggressive biological behaviour and unfavourable prognosis whereas BRAF mutations are found in early clinical stage [75, 76]. Other driver mutations involved in the pathogenetic mechanisms of LGSCs have been observed in PIK3CA, FFAR1, MACF1 (11%), USP9X (11-27%), ARID1A (9%), NF2 (4%), DOT1L (6%), ASH1L (4%) and EIF1AX (15%) [77]. USPX9 and EIF1AX are downstream effectors of MAPK pathway and linked to the mTOR pathway. Therefore, mTOR inhibitors may be used for targeted therapies in chemoresistant recurrent tumours [78].

Advertisement

4. Ovarian endometrioid carcinomas (OECs)

Endometrioid ovarian carcinomas account for 10–15% of epithelial ovarian tumours are correlated with favourable prognosis and can be found in women ageing 30–80 years [79]. They arise mainly from endometriosis lesions and less frequently from benign or borderline endometrioid ovarian neoplasms, such as adenofibromas or endometrioid borderline tumours [80]. Atypical endometriosis is the precursor lesion for 40% of OECs. Risk factors for OECs are endometriosis lesions, Lynch syndrome, hereditary breast and ovarian cancer syndrome, late menopause and post-menopausal hormone therapy [80, 81].

4.1 Pathology of ovarian endometrioid carcinomas

Endometrioid carcinomas of the ovary present mainly as unilateral mass in the ovary and less frequently as bilateral (20%) [82]. They display a smooth external surface and a solid/cystic cut surface, sometimes with a residual endometriotic cyst at the periphery of the tumour [80]. Microscopically, these tumours show a variety of morphologic patterns such as glandular, cribriform, villoglandular or solid with characteristic back-to-back glands, areas of squamous differentiation and expansile rather than an infiltrative pattern of invasion [5]. Sometimes a destructive invasion pattern can be seen characterised by neoplastic cells infiltrating the stroma accompanied by a desmoplastic reaction. The neoplastic cells are tall columnar and focally mucinous with a mitotic activity of 5–10 mitoses per high power field [5, 82]. Histologic characteristics confirmatory of OECs are metaplastic features such as squamous, morular, hobnail or mucinous metaplasia, presence of endometriosis lesions, ovarian endometrioid adenofibroma or endometrioid borderline tumour, and presence of a synchronous uterine endometrioid carcinoma found in 15–20% of the cases [5]. Ovarian endometrioid carcinomas are divided according to the presence of solid growth pattern in grade 1 (less than 5% solid growth), grade 2 (5–50% solid growth) and grade 3 (more than 50% solid growth), excluding areas of squamous differentiation [5].

4.2 Immunophenotypic features of ovarian endometrioid carcinomas

OECs show diffuse immunopositivity for CK7, PAX8, ER and PgR [83]. Approximately 33% of OECs display membranous and/or cytoplasmic diffuse expression for vimentin [84]. Squamous morules of endometrioid tumours demonstrate strong CD10 cytoplasmic expression [85] and infrequently CDX2 immunopositive reaction [86]. Endometrioid ovarian carcinomas also exhibit β-catenin membranous and/or cytoplasmic expression. Nuclear β-catenin expression is correlated with CTNNB1 genomic alterations and favourable prognosis [77]. Differential diagnosis between high-grade endometrioid carcinomas with solid architectural patterns and high-grade serous carcinomas can be made based on the intense diffuse immunopositivity of WT1, p53 and p16 in HGSCs, whereas ECs display patchy expression for p16, wild-type or mutation-type expression for p53 and loss of WT1 immunoreactivity [87].

4.3 Molecular pathology of ovarian endometrioid carcinomas

OECs are characterised mainly by mutations in PIK3CA (15–40%), ARID1A (30–35%), a component of the SW1/SNF chromatin remodelling complex, KRAS (10–30%) and CTNNB1 (25–60%) involved in the WNT/β-catenin signal transduction pathway [88, 89, 90]. Borderline endometrioid ovarian tumours have CTNNB1 mutations in 90% of the cases [77, 90]. Other less common genomic alterations are mutations in PTEN (20–30%) with frequent loss of heterozygosity (45–75%), TP53 (10–25%) and POLE (3–10%) [91, 92]. Somatic or germline predisposition mutations in MMR genes (MLH1, MSH2, MSH6, PMS2) can be found in 10–20% of the cases, some of them associated with Lynch syndrome [93, 94]. POLE-mutated EOCs and MMR-deficient ovarian endometrioid tumours have favourable clinical outcomes [95, 96]. CTNNB1-mutated tumours show low genomic complexity and are correlated with low-grade tumours and good prognosis, unlike their uterine endometrioid counterparts, which demonstrate worse clinical outcomes [77, 97]. TP53 mutated EOCs have high genomic complexity with poor prognosis [98]. Synchronous presence of endometrial and ovarian endometrioid carcinomas can be encountered in 25% of the cases demonstrating a putative clonal relationship with favourable prognosis [99, 100]. Ovarian seromucinous carcinomas (mixed serous and endocervical-type mucinous carcinomas) are considered by the current WHO classification a subtype of endometrioid ovarian carcinomas based on their morphological and molecular overlapping features [5, 101, 102]. Based on their molecular features, EOCs are divided into four molecular subcategories: 1. hypermutated with microsatellite instability due to MMR deficiency (10–20%), 2. ultramutated due to POLE exonuclease domain mutations (3–10%), 3. TP53 mutated (10–25%) and 4. with no specific molecular signatures (60–70%) [95, 103]. Hypermutated and ultramutated EOCs display high mutation burden, a molecular finding that might be exploited for immunotherapeutic interventions. The genomic aberrations identified in EOCs might be used as targets for therapeutic interventions, such as targeting mutated ARID1A with HDAC inhibitors or targeting dysregulated MAPK or PI3K pathways using MEK or PIK3AC inhibitors, respectively [104, 105].

Advertisement

5. Ovarian mucinous carcinomas (OMCs)

Primary ovarian mucinous tumours are relatively rare, approximately 10–15% of all ovarian tumours and most of them (80%) are benign or borderline mucinous tumours. Mucinous carcinomas in the ovary represent 3% of ovarian carcinomas with relatively high prevalence in women below 40 years of age [106] and most of them are of metastatic origin, particularly from the gastrointestinal or pancreatobiliary tract associated with pseudomyxoma peritonei [5, 107]. Primary OMCs originate mainly from mucinous benign or borderline tumours, with a small percentage originating from mature cystic teratomas or Brenner tumours with gastrointestinal pattern component [106, 107]. They are more commonly unilateral of large size (>13 cm) with no ovarian surface involvement. On the other hand, metastatic mucinous carcinomas are bilateral, smaller in size and associated with pseudomyxoma peritonei and imaging findings from other organs, mainly gastrointestinal or pancreatobiliary tract. They have a favourable prognosis in early clinical stages, albeit in advanced stages they show chemoresistance with unfavourable clinical outcomes [108, 109].

5.1 Pathology of ovarian mucinous carcinomas

Ovarian mucinous carcinomas are usually unilateral masses, large in size (8–40 cm), with the presence of unilocular or multilocular cysts filled with mucinous content. These tumours may display a variety of lesions from cystadenoma areas to borderline mucinous tumour regions and carcinomatous components. Microscopically there are two major histotypes, the intestinal and the endocervical. The intestinal type is more common than the endocervical. They exhibit glandular, cribriform, papillary and solid patterns of growth with two different patterns of invasion (i) expansile/confluent (more common) with a back-to-back glands labyrinthine complex appearance and minimal stroma and (ii) infiltrative/destructive characterised by irregular malignant glands infiltrating desmoplastic stroma. Each pattern of invasion measures 5 mm or more in linear size and they may coexist [5, 108]. Rarely, mural nodules of anaplastic carcinoma or high-grade sarcomatous-like component may be seen in ovarian mucinous carcinomas [5, 110]. There is no standardised grading system for OMCs till now.

5.2 Immunophenotypic features of ovarian mucinous carcinomas

Immunohistochemistry of OMCs is characterised by diffuse intense positivity for CK7 and variable positivity for CA19-9, CEA, CK20 and CDX2 focal weak positivity for PAX8 can be seen in a subset of tumours [111, 112], whereas WT1, Napsin A, Vimentin, CA125, ER and PgR are mostly negative. SATB2 is rarely expressed in ovarian mucinous tumours (5–7%) and its expression is associated with the presence of mature teratoma [113] p53 may demonstrate wild-type or mutation-type immunoreactivity and p16 is usually negative or focally positive [114, 115].

5.3 Molecular pathology of ovarian mucinous carcinomas

OMCs are characterised by KRAS and TP53 mutations (64–66%), CDKN2A inactivation (76%) and HER2/neu gene amplification (20–26%) [107, 116]. HER2 gene amplification is almost mutually exclusive to KRAS mutations and is found in most of the cases with mutated TP53 [64%) [116]. OMCs can be developed from benign mucinous tumours through a progression tumour evolution model starting with KRAS or CDKN2A genomic alterations. Both KRAS and CDKN2A mutations along with extra genomic copy number aberrations have been found in mucinous borderline tumours and, therefore, are regarded as early molecular events [117, 118]. Chromosomal locus 9p13.3 amplification and TP53 mutations are identified at the final evolutionary steps of OMCs’ progressive carcinogenesis [118]. Other less frequently mutated genes in MOCs are PIK3CA, PTEN, BRAF, CTNNB1/APC (regulators of the β-catenin/Wnt signal transduction pathway), RNF43 and ARID1A (8–12%) [107]. About 34% of OMCs have neither KRAS nor HER2 gene alterations and are considered to be neoplasms arising from mature cystic teratomas and correlated with an increased risk of recurrence and poor clinical outcome [119]. OMCs with high number of genomic aberrations and mutational burden are associated with high grade and unfavourable prognosis [107]. Targeted therapeutic approaches against HER2 amplification and/or MAPK pathway mutations might be applied along with other inhibitors, such as HDAC inhibitor for ARID1A, for more effective tailored treatment of OMCs [120].

Advertisement

6. Ovarian clear cell carcinomas (OCCCs)

Ovarian clear cell carcinomas comprise 10–12% of ovarian carcinomas with relatively high prevalence in young women of East Asian origin [121]. They are frequently associated with endometriosis lesions (50–74% of the cases) and/or clear cell benign (adenofibroma) or borderline tumours, pathologic entities with PIK3CA and ARID1A precursor genetic alterations [122]. They present mostly as unilateral mass in clinical stage I or II [5] and are considered a high-grade malignancy, although stage I patients have a favourable prognosis. Advanced-stage patients are related to poor clinical outcomes due to chemoresistance. Predisposition risk factors are late menopause, Lynch syndrome and expression of the genetic locus HNF1B through epigenetic mechanisms [122]. Lynch syndrome-associated OCCCs or tumours with MMR deficiency are correlated with long survival due to putative tumour immunogenicity [123].

6.1 Pathology of ovarian clear cell carcinomas

Grossly, OCCCs are large (mean size 13 cm) unilateral masses, solid and cystic in appearance, frequently containing endometriosis lesions. Microscopically, they have solid, tubulocystic and papillary architectural patterns and are composed of neoplastic cells characterised as polygonal, cuboidal or hobnail-like cells with clear cytoplasm, atypical nuclei and distinct nucleoli, without prominent pleomorhism [5, 122]. Mitotic activity is less than 5 mitoses per 10 high power fields. The clear cytoplasm is glycogen rich, PAS positive and diastase sensitive. Stromal hyalinization and myxoid appearance can be seen frequently [5].

6.2 Immunophenotypic features of ovarian clear cell carcinomas

OCCCs are strongly immunopositive for Napsin A (80–85%) and HNF1β (80–92%) [124, 125] and negative for WT1, ER and PgR. In addition, OCCCs show a wild-type pattern of p53 expression [126]. Differential diagnosis of OCCCs should be made among ECs and HGSCs because in bothtumours clear cell change can be observed, HGSCs demonstrate positive immunoreaction for WT1 and ER while ECs show positivity for ER [127]. Additionally, both tumours (HGSCs and ECs) are negative for napsin and HNF1β [127].

6.3 Molecular pathology ovarian clear cell carcinomas

The genomic aberrations found in OCCCs involve PIK3CA activating mutations (40–50%), ARID1A loss of function mutations (50–75%), MET gene amplifications, mutations in ARID1B (10%), KRAS (15%), PPP2R1A (15%), TERT promoter (15%), SMARCA4, PTEN (1–5%), PIK3CA, PIK3R1, AKT2, TP53 (5–20%) and ZNF217transcription factor overexpression, which is associated with poor outcome [128, 129]. PIK3CA mutations commonly coexist with ARID1A genomic alterations and are more frequent in endometriosis-associated OCCCs [128]. Studies on genes involved in antioxidant cell machineries such as GPX3 (glutathione peroxidase 3), GLRX3 (glutaredoxin) and SOD3 (superoxide dismutase) have shown that these genes are highly expressed in CCOCs resulting in tumour chemotherapy resistance [129, 130]. HER2 gene amplification (15%) and Mismatch Repair (MMR) gene deficiency (2–3%) have also been identified [131]. MMR germline mutations can be found in 10% of OCCCs and may predispose them in developing OCCCs [131]. Therefore, MMR gene expression should be tested by immunohistochemical methods or by MSI testing in order to identify OCCCs associated with MMR deficiency and/or Lynch syndrome. This is important, taking into account that MMR deficient OCCCs are correlated with favourable clinical outcome even in advanced stages [132]. Mutations of the TP53 gene are usually rare, albeit abnormal p53 expression (7%) has been reported and is associated with adverse prognosis [132]. Molecules involved in the PIK3/AKT/mTOR pathway and loss of function of ARID1A gene might be targeted therapeutically by using mTOR inhibitors and through inhibition of EZH2 transcription factor, respectively [133].

References

  1. 1. Reid BM, Permuth JB, Sellers TA. Epidemiology of ovarian cancer: A review. Cancer Biology & Medicine. 2017;14(1):9-32
  2. 2. Torre LA, Trabert B, DeSantis CE, Miller KD, Samimi G, Runowicz CD, et al. Ovarian cancer statistics, 2018. CA: A Cancer Journal for Clinicians. 2018;68(4):284-296
  3. 3. Cancer statistics-Ovarian Cancer 2011-2017. Available from: https://seer.cancer.gov/statfacts/html/ovary.html
  4. 4. Romero I, Leskelä S, Mies BP, Velasco AP, Palacios J. Morphological and molecular heterogeneity of epithelial ovarian cancer: Therapeutic implications. EJC Supplements. 2020;15:1-15
  5. 5. WHO Classification of Tumors Editorial Board. WHO Classification of Tumours: Female Genital Tumours. 5th ed. Vol. 4. Lyon (France): International Agency for Research on Cancer; 2020
  6. 6. Prat J, D’Angelo E, Espinosa I. Ovarian carcinomas: At least five different diseases with distinct histological features and molecular genetics. Human Pathology. 2018;80:11-27
  7. 7. Kurman RJ, IeM S. The dualistic model of ovarian carcinogenesis: Revisited, revised, and expanded. The American Journal of Pathology. 2016;186(4):733-747
  8. 8. Králíčková M, Laganà AS, Ghezzi F, Vetvicka V. Endometriosis and risk of ovarian cancer: What do we know? Archives of Gynecology and Obstetrics. 2020;301(1):1-10
  9. 9. Hatina J, Boesch M, Sopper S, Kripnerova M, Wolf D, Reimer D, et al. Ovarian cancer stem cell heterogeneity. Advances in Experimental Medicine and Biology. 2019;1139:201-221
  10. 10. Shih IM, Wang Y, Wang TL. The origin of ovarian cancer species and precancerous landscape. The American Journal of Pathology. 2021;191(1):26-39
  11. 11. Li HX, Lu ZH, Shen K, Cheng WJ, Malpica A, Zhang J, et al. Advances in serous tubal intraepithelial carcinoma: Correlation with high grade serous carcinoma and ovarian carcinogenesis. International Journal of Clinical and Experimental Pathology. 2014;7(3):848-857
  12. 12. Kuhn E, Wang TL, Doberstein K, Bahadirli-Talbott A, Ayhan A, Sehdev AS, et al. CCNE1 amplification and centrosome number abnormality in serous tubal intraepithelial carcinoma: Further evidence supporting its role as a precursor of ovarian high-grade serous carcinoma. Modern Pathology. 2016;29(10):1254-1261
  13. 13. Soong TR, Howitt BE, Horowitz N, Nucci MR, Crum CP. The fallopian tube, “precursor escape” and narrowing the knowledge gap to the origins of high-grade serous carcinoma. Gynecologic Oncology. 2019;152(2):426-433
  14. 14. Rhyasen GW, Yao Y, Zhang J, Dulak A, Castriotta L, Jacques K, et al. BRD4 amplification facilitates an oncogenic gene expression program in high-grade serous ovarian cancer and confers sensitivity to BET inhibitors. PLoS One. 2018;13(7):e0200826
  15. 15. Sehdev AS, Kurman RJ, Kuhn E, IeM S. Serous tubal intraepithelial carcinoma upregulates markers associated with high-grade serous carcinomas including Rsf-1 (HBXAP), cyclin E and fatty acid synthase. Modern Pathology. 2010;23(6):844-855
  16. 16. Ayhan A, Mao TL, Seckin T, Wu CH, Guan B, Ogawa H, et al. Loss of ARID1A expression is an early molecular event in tumor progression from ovarian endometriotic cyst to clear cell and endometrioid carcinoma. International Journal of Gynecological Cancer. 2012;22(8):1310-1315
  17. 17. Bulun SE, Wan Y, Matei D. Epithelial mutations in endometriosis: Link to ovarian cancer. Endocrinology. 2019;160(3):626-638
  18. 18. Ruderman R, Pavone ME. Ovarian cancer in endometriosis: An update on the clinical and molecular aspects. Minerva Ginecologica. 2017;69(3):286-294
  19. 19. Ramalingam P. Morphologic, immunophenotypic, and molecular features of epithelial ovarian cancer. Oncology (Williston Park, N.Y.). 2016;30(2):166-176
  20. 20. Prahm KP, Karlsen MA, Høgdall E, Scheller NM, Lundvall L, Nedergaard L, et al. The prognostic value of dividing epithelial ovarian cancer into type I and type II tumors based on pathologic characteristics. Gynecologic Oncology. 2015;136(2):205-211
  21. 21. Koshiyama M, Matsumura N, Konishi I. Recent concepts of ovarian carcinogenesis: Type I and type II. BioMed Research International. 2014;2014:934261. DOI: 10.1155/2014/934261
  22. 22. Lisio MA, Fu L, Goyeneche A, Gao ZH, Telleria C. High-grade serous ovarian cancer: Basic sciences, clinical and therapeutic standpoints. International Journal of Molecular Sciences. 2019;20(4):952
  23. 23. Minion LE, Dolinsky JS, Chase DM, Dunlop CL, Chao EC, Monk BJ. Hereditary predisposition to ovarian cancer, looking beyond BRCA1/BRCA2. Gynecologic Oncology. 2015;137(1):86-92
  24. 24. Suszynska M, Ratajska M, Kozlowski P. BRIP1, RAD51C, and RAD51D mutations are associated with high susceptibility to ovarian cancer: Mutation prevalence and precise risk estimates based on a pooled analysis of ~30,000 cases. Journal of Ovarian Research. 2020;13(1):50
  25. 25. Chandrasekaran D, Sobocan M, Blyuss O, Miller RE, Evans O, Crusz SM, et al. Implementation of multigene germline and parallel somatic genetic testing in epithelial ovarian cancer: SIGNPOST study. Cancers (Basel). 2021;13(17):4344
  26. 26. Soslow RA, Han G, Park KJ, Garg K, Olvera N, Spriggs DR, et al. Morphologic patterns associated with BRCA1 and BRCA2 genotype in ovarian carcinoma. Modern Pathology. 2012;25(4):625-636
  27. 27. De Leo A, Santini D, Ceccarelli C, Santandrea G, Palicelli A, Acquaviva G, et al. What is new on ovarian carcinoma: Integrated morphologic and molecular analysis following the new 2020 world health organization classification of female genital tumors. Diagnostics (Basel). 2021;11(4):697
  28. 28. Manu V, Hein TA, Boruah D, Srinivas V. Serous ovarian tumors: Immunohistochemical profiling as an aid to grading and understanding tumorigenesis. Medical Journal, Armed Forces India. 2020;76(1):30-36
  29. 29. Köbel M, Piskorz AM, Lee S, Lui S, LePage C, Marass F, et al. Optimized p53 immunohistochemistry is an accurate predictor of TP53 mutation in ovarian carcinoma. The Journal of Pathology. Clinical Research. 2016;2(4):247-258
  30. 30. Chen M, Yao S, Cao Q , Xia M, Liu J, He M. The prognostic value of Ki67 in ovarian high-grade serous carcinoma: An 11-year cohort study of Chinese patients. Oncotarget. 2016;8(64):107877-107885
  31. 31. Ordóñez NG. Value of immunohistochemistry in distinguishing peritoneal mesothelioma from serous carcinoma of the ovary and peritoneum: A review and update. Advances in Anatomic Pathology. 2006;13(1):16-25
  32. 32. Köbel M, Ronnett BM, Singh N, Soslow RA, Gilks CB, McCluggage WG. Interpretation of P53 immunohistochemistry in endometrial carcinomas: Toward increased reproducibility. International Journal of Gynecological Pathology. 2019;38(1):S123-S131
  33. 33. Al-Hussaini M, Stockman A, Foster H, McCluggage WG. WT-1 assists in distinguishing ovarian from uterine serous carcinoma and in distinguishing between serous and endometrioid ovarian carcinoma. Histopathology. 2004;44(2):109-115
  34. 34. Hashi A, Yuminamochi T, Murata S, Iwamoto H, Honda T, Hoshi K. Wilms tumor gene immunoreactivity in primary serous carcinomas of the fallopian tube, ovary, endometrium, and peritoneum. International Journal of Gynecological Pathology. 2003;22(4):374-377
  35. 35. Waldstrøm M, Grove A. Immunohistochemical expression of Wilms tumor gene protein in different histologic subtypes of ovarian carcinomas. Archives of Pathology & Laboratory Medicine. 2005;129(1):85-88
  36. 36. Wojnarowicz PM, Oros KK, Quinn MC, Arcand SL, Gambaro K, Madore J, et al. The genomic landscape of TP53 and p53 annotated high grade ovarian serous carcinomas from a defined founder population associated with patient outcome. PLoS One. 2012;7(9):e45484
  37. 37. Vang R, Levine DA, Soslow RA, Zaloudek C, IeM S, Kurman RJ. Molecular alterations of TP53 are a defining feature of ovarian high-grade serous carcinoma: A rereview of cases lacking TP53 mutations in the cancer genome atlas ovarian study. International Journal of Gynecological Pathology. 2016;35(1):48-55
  38. 38. Otsuka I. Mechanisms of high-grade serous carcinogenesis in the fallopian tube and ovary: Current hypotheses, etiologic factors, and molecular alterations. International Journal of Molecular Sciences. 2021;22(9):4409
  39. 39. Brown LA, Irving J, Parker R, Kim H, Press JZ, Longacre TA, et al. Amplification of EMSY, a novel oncogene on 11q13, in high grade ovarian surface epithelial carcinomas. Gynecologic Oncology. 2006;100(2):264-270
  40. 40. Ballabio S, Craparotta I, Paracchini L, Mannarino L, Corso S, Pezzotta MG, et al. Multisite analysis of high-grade serous epithelial ovarian cancers identifies genomic regions of focal and recurrent copy number alteration in 3q26.2 and 8q24.3. International Journal of Cancer. 2019;145(10):2670-2681
  41. 41. Moschetta M, George A, Kaye SB, Banerjee S. BRCA somatic mutations and epigenetic BRCA modifications in serous ovarian cancer. Annals of Oncology. 2016;27(8):1449-1455
  42. 42. Takaya H, Nakai H, Takamatsu S, Mandai M, Matsumura N. Homologous recombination deficiency status-based classification of high-grade serous ovarian carcinoma. Scientific Reports. 2020;10(1):2757. DOI: 10.1038/s41598-020-59671-3
  43. 43. Pietragalla A, Arcieri M, Marchetti C, Scambia G, Fagotti A. Ovarian cancer predisposition beyond BRCA1 and BRCA2 genes. International Journal of Gynecological Cancer. 2020;30(11):1803-1810
  44. 44. Ritterhouse LL, Nowak JA, Strickland KC, Garcia EP, Jia Y, Lindeman NI, et al. Morphologic correlates of molecular alterations in extrauterine Müllerian carcinomas. Modern Pathology. 2016;29(8):893-903
  45. 45. Banerjee SN, Lord CJ. First-line PARP inhibition in ovarian cancer - Standard of care for all? Nature Reviews. Clinical Oncology. 2020;17(3):136-137
  46. 46. Jiang X, Li X, Li W, Bai H, Zhang Z. PARP inhibitors in ovarian cancer: Sensitivity prediction and resistance mechanisms. Journal of Cellular and Molecular Medicine. 2019;23(4):2303-2313
  47. 47. Miller RE, Leary A, Scott CL, Serra V, Lord CJ, Bowtell D, et al. ESMO recommendations on predictive biomarker testing for homologous recombination deficiency and PARP inhibitor benefit in ovarian cancer. Annals of Oncology. 2020;31:12, 1606-1622
  48. 48. Sztupinszki Z, Diossy M, Borcsok J, Prosz A, Cornelius N, Kjeldsen MK, et al. Comparative assessment of diagnostic homologous recombination deficiency-associated mutational signatures in ovarian cancer. Clinical Cancer Research. 2021;27(20):5681-5687
  49. 49. Stover EH, Fuh K, Konstantinopoulos PA, Matulonis UA, Liu JF. Clinical assays for assessment of homologous recombination DNA repair deficiency. Gynecologic Oncology. 2020;159(3):887-898
  50. 50. Weigelt B, Comino-Méndez I, de Bruijn I, Tian L, Meisel JL, García-Murillas I, et al. Diverse BRCA1 and BRCA2 reversion mutations in circulating cell-free DNA of therapy-resistant breast or ovarian cancer. Clinical Cancer Research. 2017;23(21):6708-6720
  51. 51. Norquist B, Wurz KA, Pennil CC, Garcia R, Gross J, Sakai W, et al. Secondary somatic mutations restoring BRCA1/2 predict chemotherapy resistance in hereditary ovarian carcinomas. Journal of Clinical Oncology. 2011;29(22):3008-3015
  52. 52. Sheta R, Bachvarova M, Plante M, Renaud MC, Sebastianelli A, Gregoire J, et al. Development of a 3D functional assay and identification of biomarkers, predictive for response of high-grade serous ovarian cancer (HGSOC) patients to poly-ADP ribose polymerase inhibitors (PARPis): targeted therapy. Journal of Translational Medicine. 2020;18(1):439
  53. 53. Fuh K, Mullen M, Blachut B, Stover E, Konstantinopoulos P, Liu J, et al. Homologous recombination deficiency real-time clinical assays, ready or not? Gynecologic Oncology. 2020;159(3):877-886
  54. 54. Konecny GE, Wang C, Hamidi H, Winterhoff B, Kalli KR, Dering J, et al. Prognostic and therapeutic relevance of molecular subtypes in high-grade serous ovarian cancer. Journal of the National Cancer Institute. 2014;106(10):dju249
  55. 55. Cook DP, Vanderhyden BC. Ovarian cancer and the evolution of subtype classifications using transcriptional profiling†. Biology of Reproduction. 2019;101(3):645-658
  56. 56. Mota A, Oltra S, S, Moreno-Bueno G. Insight updating of the molecular hallmarks in ovarian carcinoma. EJC Supplements. 2020;15:16-26
  57. 57. Wang C, Cicek MS, Charbonneau B, Kalli KR, Armasu SM, Larson MC, et al. Tumor hypomethylation at 6p21.3 associates with longer time to recurrence of high-grade serous epithelial ovarian cancer. Cancer Research. 2014;74(11):3084-3091
  58. 58. Bodelon C, Killian JK, Sampson JN, Anderson WF, Matsuno R, Brinton LA, et al. Molecular classification of epithelial ovarian cancer based on methylation profiling: Evidence for survival heterogeneity. Clinical Cancer Research. 2019;25(19):5937-5946
  59. 59. Li J, Wang J, Chen R, Bai Y, Lu X. The prognostic value of tumor-infiltrating T lymphocytes in ovarian cancer. Oncotarget. 2017;8(9):15621-15631
  60. 60. Wang L. Prognostic effect of programmed death-ligand 1 (PD-L1) in ovarian cancer: A systematic review, meta-analysis and bioinformatics study. Journal of Ovarian Research. 2019;12(1):37
  61. 61. Darb-Esfahani S, Kunze CA, Kulbe H, Sehouli J, Wienert S, Lindner J, et al. Prognostic impact of programmed cell death-1 (PD-1) and PD-ligand 1 (PD-L1) expression in cancer cells and tumor-infiltrating lymphocytes in ovarian high grade serous carcinoma. Oncotarget. 2016;7(2):1486-1499
  62. 62. Whitehair R, Peres LC, Mills AM. Expression of the immune checkpoints LAG-3 and PD-L1 in high-grade serous ovarian carcinoma: Relationship to tumor-associated lymphocytes and germline BRCA status. International Journal of Gynecological Pathology. 2020;39(6):558-566
  63. 63. Huang CC, Cheng SH, Wu CH, Li WY, Wang JS, Kung ML, et al. Delta-like 1 homologue promotes tumorigenesis and epithelial-mesenchymal transition of ovarian high-grade serous carcinoma through activation of Notch signaling. Oncogene. 2019;38(17):3201-3215
  64. 64. Chen S, Cavazza E, Barlier C, Salleron J, Filhine-Tresarrieu P, Gavoilles C, et al. Beside P53 and PTEN: Identification of molecular alterations of the RAS/MAPK and PI3K/AKT signaling pathways in high-grade serous ovarian carcinomas to determine potential novel therapeutic targets. Oncology Letters. 2016;12(5):3264-3272
  65. 65. Pfisterer J, Du Bois A, Bentz EK, Kommoss F, Harter P, Huober J, et al. Prognostic value of human epidermal growth factor receptor 2 (Her-2)/neu in patients with advanced ovarian cancer treated with platinum/paclitaxel as first-line chemotherapy: A retrospective evaluation of the AGO-OVAR 3 Trial by the AGO OVAR Germany. International Journal of Gynecological Cancer. 2009;19(1):109-115
  66. 66. Ersoy E, Cao QJ, Otis CN. HER2 protein overexpression and gene amplification in tubo-ovarian high-grade serous carcinomas. International Journal of Gynecological Pathology. 2022;41(4):313-319
  67. 67. Slomovitz B, Gourley C, Carey MS, Malpica A, Shih IM, Huntsman D, et al. Low-grade serous ovarian cancer: State of the science. Gynecologic Oncology. 2020;156(3):715-725
  68. 68. Longacre TA, McKenney JK, Tazelaar HD, Kempson RL, Hendrickson MR. Ovarian serous tumors of low malignant potential (borderline tumors): Outcome-based study of 276 patients with long-term (> or =5-year) follow-up. The American Journal of Surgical Pathology. 2005;29(6):707-723
  69. 69. Bell DA. Low-grade serous tumors of ovary. International Journal of Gynecological Pathology. 2014;33(4):348-356
  70. 70. O’Neill CJ, Deavers MT, Malpica A, Foster H, McCluggage WG. An immunohistochemical comparison between low-grade and high-grade ovarian serous carcinomas: Significantly higher expression of p53, MIB1, BCL2, HER-2/neu, and C-KIT in high-grade neoplasms. The American Journal of Surgical Pathology. 2005;29(8):1034-1041
  71. 71. Van Nieuwenhuysen E, Busschaert P, Laenen A, Moerman P, Han SN, Neven P, et al. Loss of 1p36.33 frequent in low-grade serous ovarian cancer. Neoplasia. 2019;21(6):582-590
  72. 72. Hunter SM, Anglesio MS, Ryland GL, Sharma R, Chiew YE, Rowley SM, et al. Molecular profiling of low grade serous ovarian tumours identifies novel candidate driver genes. Oncotarget. 2015;6(35):37663-37677
  73. 73. Anglesio MS, Arnold JM, George J, Tinker AV, Tothill R, Waddell N, et al. Mutation of ERBB2 provides a novel alternative mechanism for the ubiquitous activation of RAS-MAPK in ovarian serous low malignant potential tumors. Molecular Cancer Research. 2008;6(11):1678-1690
  74. 74. Ho CL, Kurman RJ, Dehari R, Wang TL, IeM S. Mutations of BRAF and KRAS precede the development of ovarian serous borderline tumors. Cancer Research. 2004;64(19):6915-6918
  75. 75. Tsang YT, Deavers MT, Sun CC, Kwan SY, Kuo E, Malpica A, et al. KRAS (but not BRAF) mutations in ovarian serous borderline tumour are associated with recurrent low-grade serous carcinoma. The Journal of Pathology. 2013;231(4):449-456
  76. 76. Grisham RN, Iyer G, Garg K, Delair D, Hyman DM, Zhou Q , et al. BRAF mutation is associated with early stage disease and improved outcome in patients with low-grade serous ovarian cancer. Cancer. 2013;119(3):548-554
  77. 77. Zyla RE, Olkhov-Mitsel E, Amemiya Y, Bassiouny D, Seth A, Djordjevic B, et al. CTNNB1 mutations and aberrant β-catenin expression in ovarian endometrioid carcinoma: Correlation with patient outcome. The American Journal of Surgical Pathology. 2021;45(1):68-76
  78. 78. Farley J, Brady WE, Vathipadiekal V, Lankes HA, Coleman R, Morgan MA, et al. Selumetinib in women with recurrent low-grade serous carcinoma of the ovary or peritoneum: An open-label, single-arm, phase 2 study. The Lancet Oncology. 2013;14(2):134-140
  79. 79. Nasioudis D, Latif NA, Simpkins F, Cory L, Giuntoli RL 2nd, Haggerty AF, et al. Adjuvant chemotherapy for early stage endometrioid ovarian carcinoma: An analysis of the National Cancer Data Base. Gynecologic Oncology. 2020;156(2):315-319
  80. 80. Terada T. Endometrioid adenocarcinoma of the ovary arising in atypical endometriosis. International Journal of Clinical and Experimental Pathology. 2012;5(9):924-927
  81. 81. Helder-Woolderink JM, Blok EA, Vasen HF, Hollema H, Mourits MJ, De Bock GH. Ovarian cancer in Lynch syndrome; a systematic review. European Journal of Cancer. 2016;55:65-73
  82. 82. Assem H, Rambau PF, Lee S, Ogilvie T, Sienko A, Kelemen LE, et al. High-grade endometrioid carcinoma of the ovary: A clinicopathologic study of 30 cases. The American Journal of Surgical Pathology. 2018;42(4):534-544
  83. 83. Santandrea G, Piana S, Valli R, Zanelli M, Gasparini E, De Leo A, et al. Immunohistochemical biomarkers as a surrogate of molecular analysis in ovarian carcinomas: A review of the literature. Diagnostics (Basel). 2021;11(2):199
  84. 84. Köbel M, Rahimi K, Rambau PF, Naugler C, Le Page C, Meunier L, et al. An immunohistochemical algorithm for ovarian carcinoma typing. International Journal of Gynecological Pathology. 2016;35(5):430-441
  85. 85. Chiarelli S, Buriticá C, Litta P, Ciani S, Guarch R, Nogales FF. An immunohistochemical study of morules in endometrioid lesions of the female genital tract: CD10 is a characteristic marker of morular metaplasia. Clinical Cancer Research. 2006;12(14 Pt 1):4251-4256
  86. 86. Houghton O, Connolly LE, McCluggage WG. Morules in endometrioid proliferations of the uterus and ovary consistently express the intestinal transcription factor CDX2. Histopathology. 2008;53(2):156-165
  87. 87. Lim D, Murali R, Murray MP, Veras E, Park KJ, Soslow RA. Morphological and immunohistochemical reevaluation of tumors initially diagnosed as ovarian endometrioid carcinoma with emphasis on high-grade tumors. The American Journal of Surgical Pathology. 2016;40(3):302-312
  88. 88. Wiegand KC, Shah SP, Al-Agha OM, Zhao Y, Tse K, Zeng T, et al. ARID1A mutations in endometriosis-associated ovarian carcinomas. The New England Journal of Medicine. 2010;363(16):1532-1543
  89. 89. Samartzis EP, Noske A, Dedes KJ, Fink D, Imesch P. ARID1A mutations and PI3K/AKT pathway alterations in endometriosis and endometriosis-associated ovarian carcinomas. International Journal of Molecular Sciences. 2013;14(9):18824-18849
  90. 90. Oliva E, Sarrió D, Brachtel EF, Sánchez-Estévez C, Soslow RA, Moreno-Bueno G, et al. High frequency of beta-catenin mutations in borderline endometrioid tumours of the ovary. The Journal of Pathology. 2006;208(5):708-713
  91. 91. Cybulska P, Paula ADC, Tseng J, Leitao MM Jr, Bashashati A, Huntsman DG, et al. Molecular profiling and molecular classification of endometrioid ovarian carcinomas. Gynecologic Oncology. 2019;154(3):516-523
  92. 92. Leskela S, Romero I, Rosa-Rosa JM, Caniego-Casas T, Cristobal E, Pérez-Mies B, et al. Molecular heterogeneity of endometrioid ovarian carcinoma: An analysis of 166 cases using the endometrial cancer subrogate molecular classification. The American Journal of Surgical Pathology. 2020;44(7):982-990
  93. 93. Leskela S, Romero I, Cristobal E, Pérez-Mies B, Rosa-Rosa JM, Gutierrez-Pecharroman A, et al. Mismatch repair deficiency in ovarian carcinoma: Frequency, causes, and consequences. The American Journal of Surgical Pathology. 2020;44(5):649-656
  94. 94. Bennett JA, Pesci A, Morales-Oyarvide V, Da Silva A, Nardi V, Oliva E. Incidence of mismatch repair protein deficiency and associated clinicopathologic features in a cohort of 104 ovarian endometrioid carcinomas. The American Journal of Surgical Pathology. 2019;43(2):235-243
  95. 95. Hollis RL, Thomson JP, Stanley B, Churchman M, Meynert AM, Rye T, et al. Molecular stratification of endometrioid ovarian carcinoma predicts clinical outcome. Nature Communications. 2020;11(1):4995
  96. 96. Chui MH, Ryan P, Radigan J, Ferguson SE, Pollett A, Aronson M, et al. The histomorphology of Lynch syndrome-associated ovarian carcinomas: Toward a subtype-specific screening strategy. The American Journal of Surgical Pathology. 2014;38(9):1173-1181
  97. 97. Wang L, Rambau PF, Kelemen LE, Anglesio MS, Leung S, Talhouk A, et al. Nuclear β-catenin and CDX2 expression in ovarian endometrioid carcinoma identify patients with favourable outcome. Histopathology. 2019;74(3):452-462
  98. 98. Brett MA, Atenafu EG, Singh N, Ghatage P, Clarke BA, Nelson GS, et al. Equivalent survival of p53 mutated endometrial endometrioid carcinoma grade 3 and endometrial serous carcinoma. International Journal of Gynecological Pathology. 2021;40(2):116-123
  99. 99. Anglesio MS, Wang YK, Maassen M, Horlings HM, Bashashati A, Senz J, et al. Synchronous endometrial and ovarian carcinomas: Evidence of clonality. Journal of the National Cancer Institute. 2016;108(6):djv428
  100. 100. Schultheis AM, Ng CK, De Filippo MR, Piscuoglio S, Macedo GS, Gatius S, et al. Massively parallel sequencing-based clonality analysis of synchronous endometrioid endometrial and ovarian carcinomas. Journal of the National Cancer Institute. 2016;108(6):djv427
  101. 101. Kurman RJ, Shih IM. Seromucinous tumors of the ovary. What’s in a Name? International Journal of Gynecological Pathology. 2016;35(1):78-81
  102. 102. Ben-Mussa A, McCluggage WG. Ovarian seromucinous cystadenomas and adenofibromas: First report of a case series. Histopathology. 2021;78(3):445-452
  103. 103. Parra-Herran C, Lerner-Ellis J, Xu B, Khalouei S, Bassiouny D, Cesari M, et al. Molecular-based classification algorithm for endometrial carcinoma categorizes ovarian endometrioid carcinoma into prognostically significant groups. Modern Pathology. 2017;30(12):1748-1759
  104. 104. Fukumoto T, Park PH, Wu S, Fatkhutdinov N, Karakashev S, Nacarelli T, et al. Repurposing Pan-HDAC inhibitors for ARID1A-mutated ovarian cancer. Cell Reports. 2018;22(13):3393-3400
  105. 105. Lee JM, Minasian L, Kohn EC. New strategies in ovarian cancer treatment. Cancer. 2019;125(Suppl. 24):4623-4629
  106. 106. Babaier A, Ghatage P. Mucinous cancer of the ovary: Overview and current status. Diagnostics (Basel). 2020;10(1):52
  107. 107. Cheasley D, Wakefield MJ, Ryland GL, Allan PE, Alsop K, Amarasinghe KC, et al. The molecular origin and taxonomy of mucinous ovarian carcinoma. Nature Communications. 2019;10(1):3935
  108. 108. Gouy S, Saidani M, Maulard A, Bach-Hamba S, Bentivegna E, Leary A, et al. Characteristics and prognosis of stage i ovarian mucinous tumors according to expansile or infiltrative type. International Journal of Gynecological Cancer. 2018;28(3):493-499
  109. 109. Hada T, Miyamoto M, Ishibashi H, Matsuura H, Sakamoto T, Kakimoto S, et al. Survival and biomarker analysis for ovarian mucinous carcinoma according to invasive patterns: Retrospective analysis and review literature. Journal of Ovarian Research. 2021;14(1):33
  110. 110. Provenza C, Young RH, Prat J. Anaplastic carcinoma in mucinous ovarian tumors: A clinicopathologic study of 34 cases emphasizing the crucial impact of stage on prognosis, their histologic spectrum, and overlap with sarcomalike mural nodules. The American Journal of Surgical Pathology. 2008;32(3):383-389
  111. 111. Vang R, Gown AM, Barry TS, Wheeler DT, Yemelyanova A, Seidman JD, et al. Cytokeratins 7 and 20 in primary and secondary mucinous tumors of the ovary: Analysis of coordinate immunohistochemical expression profiles and staining distribution in 179 cases. The American Journal of Surgical Pathology. 2006;30(9):1130-1139
  112. 112. Vang R, Gown AM, Wu LS, Barry TS, Wheeler DT, Yemelyanova A, et al. Immunohistochemical expression of CDX2 in primary ovarian mucinous tumors and metastatic mucinous carcinomas involving the ovary: Comparison with CK20 and correlation with coordinate expression of CK7. Modern Pathology. 2006;19(11):1421-1428
  113. 113. Schmoeckel E, Kirchner T, Mayr D. SATB2 is a supportive marker for the differentiation of a primary mucinous tumor of the ovary and an ovarian metastasis of a low-grade appendiceal mucinous neoplasm (LAMN): A series of seven cases. Pathology, Research and Practice. 2018;214(3):426-430
  114. 114. McCluggage WG. Immunohistochemistry in the distinction between primary and metastatic ovarian mucinous neoplasms. Journal of Clinical Pathology. 2012;65(7):596-600
  115. 115. Tabrizi AD, Kalloger SE, Köbel M, Cipollone J, Roskelley CD, Mehl E, et al. Primary ovarian mucinous carcinoma of intestinal type: Significance of pattern of invasion and immunohistochemical expression profile in a series of 31 cases. International Journal of Gynecological Pathology. 2010;29(2):99-107
  116. 116. Anglesio MS, Kommoss S, Tolcher MC, Clarke B, Galletta L, Porter H, et al. Molecular characterization of mucinous ovarian tumours supports a stratified treatment approach with HER2 targeting in 19% of carcinomas. The Journal of Pathology. 2013;229(1):111-120
  117. 117. Hunter SM, Gorringe KL, Christie M, Rowley SM, Bowtell DD, Australian Ovarian Cancer Study Group, et al. Pre-invasive ovarian mucinous tumors are characterized by CDKN2A and RAS pathway aberrations. Clinical Cancer Research. 2012;18(19):5267-5277
  118. 118. Mackenzie R, Kommoss S, Winterhoff BJ, Kipp BR, Garcia JJ, Voss J, et al. Targeted deep sequencing of mucinous ovarian tumors reveals multiple overlapping RAS-pathway activating mutations in borderline and cancerous neoplasms. BMC Cancer. 2015;15:415
  119. 119. Bouri S, Simon P, D’Haene N, Catteau X, Noël JC. P53 and PIK3CA Mutations in KRAS/HER2 negative ovarian intestinal-type mucinous carcinoma associated with mature teratoma. Case Reports in Obstetrics and Gynecology. 2020;2020:8863610
  120. 120. Gorringe KL, Cheasley D, Wakefield MJ, Ryland GL, Allan PE, Alsop K, et al. Therapeutic options for mucinous ovarian carcinoma. Gynecologic Oncology. 2020;156(3):552-560
  121. 121. Liu H, Xu Y, Ji J, Dong R, Qiu H, Dai X. Prognosis of ovarian clear cell cancer compared with other epithelial cancer types: A population-based analysis. Oncology Letters. 2020;19(3):1947-1957
  122. 122. Iida Y, Okamoto A, Hollis RL, Gourley C, Herrington CS. Clear cell carcinoma of the ovary: A clinical and molecular perspective. International Journal of Gynecological Cancer. 2021;31(4):605-616
  123. 123. Stewart CJ, Bowtell DD, Doherty DA, Leung YC. Long-term survival of patients with mismatch repair protein-deficient, high-stage ovarian clear cell carcinoma. Histopathology. 2017;70(2):309-313
  124. 124. Skirnisdottir I, Bjersand K, Akerud H, Seidal T. Napsin A as a marker of clear cell ovarian carcinoma. BMC Cancer. 2013;13:524
  125. 125. Li Q , Zeng X, Cheng X, Zhang J, Ji J, Wang J, et al. Diagnostic value of dual detection of hepatocyte nuclear factor 1 beta (HNF-1β) and napsin A for diagnosing ovarian clear cell carcinoma. International Journal of Clinical and Experimental Pathology. 2015;8(7):8305-8310
  126. 126. Heckl M, Schmoeckel E, Hertlein L, Rottmann M, Jeschke U, Mayr D. The ARID1A, p53 and ß-Catenin statuses are strong prognosticators in clear cell and endometrioid carcinoma of the ovary and the endometrium. PLoS One. 2018;13(2):e0192881
  127. 127. Köbel M, Kalloger SE, Carrick J, Huntsman D, Asad H, Oliva E, et al. A limited panel of immunomarkers can reliably distinguish between clear cell and high-grade serous carcinoma of the ovary. The American Journal of Surgical Pathology. 2009;33(1):14-21
  128. 128. Huang HN, Lin MC, Huang WC, Chiang YC, Kuo KT. Loss of ARID1A expression and its relationship with PI3K-Akt pathway alterations and ZNF217 amplification in ovarian clear cell carcinoma. Modern Pathology. 2014;27(7):983-990
  129. 129. Friedlander ML, Russell K, Millis S, Gatalica Z, Bender R, Voss A. Molecular profiling of clear cell ovarian cancers: identifying potential treatment targets for clinical trials. International Journal of Gynecological Cancer. 2016;26(4):648-654
  130. 130. Takano M, Tsuda H, Sugiyama T. Clear cell carcinoma of the ovary: Is there a role of histology-specific treatment? Journal of Experimental & Clinical Cancer Research. 2012;31(1):53
  131. 131. Bennett JA, Morales-Oyarvide V, Campbell S, Longacre TA, Oliva E. Mismatch repair protein expression in clear cell carcinoma of the ovary: Incidence and morphologic associations in 109 cases. The American Journal of Surgical Pathology. 2016;40(5):656-663
  132. 132. Parra-Herran C, Bassiouny D, Lerner-Ellis J, Olkhov-Mitsel E, Ismiil N, Hogen L, et al. Mismatch repair protein, and POLE abnormalities in ovarian clear cell carcinoma: An outcome-based clinicopathologic analysis. The American Journal of Surgical Pathology. 2019;43(12):1591-1599
  133. 133. Bitler BG, Aird KM, Garipov A, Li H, Amatangelo M, Kossenkov AV, et al. Synthetic lethality by targeting EZH2 methyltransferase activity in ARID1A-mutated cancers. Nature Medicine. 2015;21(3):231-238

Written By

Gabriela-Monica Stanc, Efthymia Souka and Christos Valavanis

Submitted: 10 July 2022 Reviewed: 13 July 2022 Published: 17 August 2022