Open access peer-reviewed chapter

Ovarian Cancer: Molecular Classification and Targeted Therapy

Written By

Febina Ravindran and Bibha Choudhary

Submitted: 06 December 2020 Reviewed: 11 January 2021 Published: 05 February 2021

DOI: 10.5772/intechopen.95967

From the Edited Volume

Ovarian Cancer - Updates in Tumour Biology and Therapeutics

Edited by Gwo-Yaw Ho and Kate Webber

Chapter metrics overview

2,407 Chapter Downloads

View Full Metrics

Abstract

Ovarian cancer is the deadliest gynecological cancer among women with an overall 5-year survival rate below 50% due to its asymptomatic nature, diagnosis at advanced stages, and a high recurrence rate after standard therapy in 70% of cases. Ovarian cancers are heterogenous cancers where each subtype possesses a varied morphology and biologic behavior. Accumulating evidence has identified each of these subtypes characterized with specific pathways activated in each along with specific gene alterations. For example, high-grade serous ovarian cancer is characterized by universal TP53 mutation, mucinous ovarian cancer with KRAS mutation and clear cell or endometrioid ovarian cancers with ARID1A mutations. With the current focus of molecular-targeted therapies for cancer, such druggable markers serve as excellent targets for precision therapy and combination therapy. This chapter, provides an overview of the critical molecular pathways activated in the ovarian cancer subtypes with its druggable targets studied in ovarian cancer. We also highlight the implications of miRNAs in chemoresistance and sensitivity in the regulation of ovarian cancer.

Keywords

  • ovarian cancer subtypes
  • targeted therapy
  • miRNAs in ovarian cancers

1. Introduction

Ovaries are the prime female reproductive organ that produces the oocyte or the egg cell for fertilization. It is also an endocrine gland that produces the female sex hormones estrogen and progesterone responsible for ovulation and pregnancy maintenance. Some of the diseases that affect the ovaries are ovarian cysts, primary ovarian insufficiency, ovarian torsion and more recently ovarian cancer (OC). OC was first detected in the 1950s and is now one of the deadliest gynecological cancers among women [1, 2]. According to the latest Global Cancer Observatory: CANCER TODAY (GLOBOCAN 2018), the incidence and mortality rates of OC vary globally and ranks at the 8th and 7th position respectively [3]. The highest mortality rates are reported in Oceania and Europe and the lowest are from Latin America, the Caribbean and Asia [3]. OCs are also prevalent in countries with a high human development index (HDI) but with lower mortality rates due to increased diagnostic and therapeutic support [4].

Most OCs manifest post menopause and the increased incidence is reported in women older than 65 years [5]. Considering the ethnicity, non-Hispanic white women are reported to have the highest incidence and mortality rates [6]. OCs are heterogeneous cancer, hence the risk factors for each histological subtype vary. In general, some of the major risk factors for OC include Hereditary Breast and Ovarian Cancer (HBOC) syndrome [7], Lynch syndrome [8], menopausal hormonal therapy [9, 10], endometriosis [11], IVF treatment [12], use of fertility drugs [13], late menopause [14] and null parity [15]. Interestingly, high parity [16], hysterectomy [17] and usage of hormonal contraceptive pills for prolonged periods [18] are reported to have a protective effect since these conditions confer in the suppression of ovulatory cycles [19]. The sterilization treatment, tubal ligation is also reported to reduce the risk of OCs [17, 20]. Recently reported other emerging risk factors for OCs are the use of talc powders [21], asbestos exposure [22] and pelvic inflammatory disease [23].

OCs are difficult to detect;therefore almost 60% of OC cases are diagnosed at advanced stages [24]. It is often called the “whispering cancer” or “silent cancer” due to its asymptomatic nature and late presentation [25, 26]. Late-stage OC symptoms are very nonspecific and diffuse but may include abdominal bloating or swelling, pelvic pain, increased urinary urgency, weight loss, or fatigue [27, 28]. Although a biopsy is the only reliable diagnosis for OC, screening for serum cancer antigen 125 (CA-125) levels combined with ultrasound imaging are used for women with increased risk [29]. The emerging technique of liquid biopsy is being explored for identifying serum biomarkers for early detection of OCs. It holds great promise being non-invasive and is utilized to diagnose, prognose and predict surgical outcomes. One such serum biomarker identified is the Human Epididymis Protein 4 (HE4) which is reported to have high specificity for OCs [30, 31]. 2011 FDA approved, ROMA index (risk of ovarian malignancy algorithm) deduced from HE4, CA-125 and the menopausal status is used for diagnosis and prognosis of OCs with a specificity of 90% [32, 33, 34]. Another recent 2016 FDA approved serum-based screening test, Overa also uses HE4 levels along with other serum proteins is reported to show a sensitivity of 94% along with pathological diagnosis [35]. The mutational status of multiple cancer-causing genes are also being developed as screening tests for various cancers like PapSEEK and CancerSEEK and are reported to detect OC with a specificity of 63% and 98%, respectively [36, 37].

According to the World cancer report 2020, OC five-year survival rate is below 30% [38]. This is mainly because this cancer gets diagnosed at stage III or IV with metastasis and the recurrence rate high despite standard therapy. Cytoreductive surgery followed by chemotherapy based on cancer’s surgical stage remains the gold standard treatment for OCs. The most commonly administered chemotherapy drugs are platinum derivatives e.g. cisplatin and carboplatin and are often combined with taxane-based drugs like paclitaxel or docetaxel. These drugs induce apoptosis in the tumor cells by creating double-stranded breaks in the DNA [39]. Despite chemotherapy being effective for advanced cancers in the initial phases, cancer relapses in 70% of cases due to drug resistance [40]. In the case of recurrent OCs, the second line of the chemotherapy treatment regimen is based on the platinum-free interval and the tumor’s molecular profile [41]. Furthermore, the treatment options include combinations of carboplatin with gemcitabine, topotecan, vinorelbine, trabectedin, belotecan or pegylated liposomal doxorubicin [42].

Despite intensive combination chemotherapy, the survival rate decreases with chemoresistance and subsequent OC metastasis. The lack of anatomical barrier around the ovaries facilitates the dissemination of OC cells into the peritoneal cavity, metastasizing onto abdominal organs resulting in bowel obstruction, which is the major cause of OC morbidity and mortality [43, 44]. Currently, there are no preventive measures for OCs, and options for the high-risk category are prophylactic surgeries like hysterectomy (removal of the uterus) combined with bilateral salpingo-oophorectomy (removal of both ovaries and fallopian tube) or bilateral salpingectomy (removal of both fallopian tubes) [45]. Women with average risk can opt for oral contraceptive treatment [46].

Presently, there is no effective cure for advanced OC. Though these cancers vary histologically, clinical treatment therapies neglect these differences and are treated as a single disease. Each OC subtype is characterized by specific genetic mutations that deregulate specific signaling pathways that should be utilized for personalized or tailored therapeutics. Precision therapy is the need of the hour for OC treatment in improving the current survival rate. In the following sections of the chapter, we describe the various OC subtypes, their histological classification and the key molecular pathways activated in each subtype along with its druggable targets.

Advertisement

2. Ovarian cancer subtypes

OC neoplasms arise from distinct regions of the ovary. They are termed heterogeneous as each OC subtype is unique with varied morphology, biologic behavior and even prognosis. High throughput sequencing technologies have identified each OC subtype as distinct even on a molecular level with unique genomic characteristics. OCs are broadly classified into epithelial and non-epithelial cancers. Non-epithelial cancer comprises germ cell cancer, stromal cell cancer, and the rare small cell carcinoma. The origin of the various subtypes of OCs and the sub-classifications are depicted in Figure 1.

Figure 1.

Origin of the various ovarian cancer subtypes and their sub-classifications.

2.1 Epithelial ovarian cancer (EOC)

Epithelial ovarian cancers (EOCs) comprise 90% of all OCs and are among the most well-characterized forms of OC. EOCs are thought to arise from the epithelium, the outer lining of the ovary. EOC is an age-related disease and is considered mainly a postmenopausal disease. Based on tumor cell morphology, they are further subdivided into high grade serous ovarian carcinoma (HGSOC), low grade serous ovarian carcinoma (LGSOC), mucinous ovarian carcinoma (MOC), endometrioid carcinoma (EC), and clear-cell carcinoma (CCC). The histological image, epidemiology, molecular alterations and pathways affecting each EOC variant are outlined in Figure 2.

Figure 2.

EOC subtypes: histology, epidemiology, and molecular alterations. Histology images courtesy [47].

2.1.1 High grade serous ovarian carcinoma (HGSOC)

High grade serous ovarian carcinomas (HGSOCs) are the most lethal forms of OCs and account 75% of all EOCs [48]. They are the most aggressive and chemoresistant forms of EOCs responsible for 70–80% of OC related deaths. HGSOCs are thought to be derived from the fallopian tube [49]. These cancers are mainly diagnosed in postmenopausal women and due to its asymptomatic character presents themselves in advanced stages. Familial HBOC syndrome, and menopausal hormonal therapy predispose women towards this cancer [25, 50].

HGSOCs are characterized by a high frequency (90%) of somatic TP53 mutations. These mutations are present in the DNA binding domain of TP53 which render its tumor-suppressive function inactive, leading to enhanced cell proliferation and metastasis. The drug APR-246 targeting TP53 resulting in its wild type stabilization is under clinical trial and has shown favorable results [51]. Another drug, nutlin-3a targeting MDM2, a negative regulator of TP53, has also entered clinical trials with positive outcomes [52]. Moreover, combination therapy using nutlin-3 and RG7388 (another MDM2-TP53 antagonist) have reported cytotoxic effects in various OC cell lines [53].

15–20% of HGSOC patients harbor germline mutations in BRCA1 or BRCA2 [48]. The BRCA genes are involved in the repair of double-strand DNA breaks through homologous recombination (HR). Besides, most HGSOCs with the germline BRCA mutation are also reported to harbor somatic mutations in other HR-related genes conferring an HR deficient (HRD) phenotype [54]. The Cancer Genome Atlas Research Network (TCGA) has reported almost 50% HGSOCs cases as HR deficient [55]. HRD conferring genes besides BRCA1/2 include Fanconi anemia genes (PALB2, FANCA, FANCI, FANCL, FANCC), RAD family genes (RAD50, RAD51, RAD51C, RAD54L), MRN complex genes (Mre11-Rad50-Nbs1), and also DNA damage response genes (ATM, ATR, CHEK1, CHEK2) [54, 56]. This manifestation of inactivating BRCA gene mutations and other HRD genes confer a DNA repair-deficient phenotype leading to genomic instability [57].

One of the most remarkable developments for OC therapy has been the PARP (poly (ADP-ribose) polymerase) inhibitors. PARP is an excision repair enzyme involved in the repair of single DNA strand breaks. PARP inhibitor treatment in BRCA-deficient cancer induces synthetic lethality and cell death [58]. The PARP inhibitor olaparib has been reported to show increased progression-free survival (PFS) and is currently approved as first-line maintenance therapy for BRCA-mutant individuals [59, 60]. Another PARP inhibitor, niraparib, improved PFS regardless of BRCA or HRD status is also approved for first-line maintenance of advanced OCs [61]. CDK4/6 inhibitors (palbociclib, ribociclib and abemaciclib) are also under clinical trials as maintenance and combination therapy for HGSOCs [62]. Cyclin-dependent kinase 4 and 6 (CDK4/6) are key kinases that regulate the cell cycle. CDK4/6 inhibitors hinder G1-S transition inducing cell cycle arrest at the G1 phase. PI3K/AKT and NOTCH pathways are reported to be deregulated in HGSOCs which could also be targeted via combination therapies using PI3K inhibitors or the AKT inhibitor, afuresertib [63].

One of the first targeted therapy used to treat advanced OCs is Bevacizumab, an anti-angiogenic agent that targets vascular endothelial growth factor (VEGF) [64]. Angiogenesis plays a pivotal role in tumor progression and metastasis in many malignant cancers. This drug acts by neutralizing VEGF-A, thereby inhibiting tumor growth and invasion. Bevacizumab is currently approved as a combination therapy along with platinum/taxane drugs for advanced HGSOCs and has been reported to show a significant improvement in progression-free survival [57].

2.1.2 Low grade serous ovarian carcinoma (LGSOC)

As the name suggests, LGSOCs are indolent and less aggressive tumors with relatively better prognosis than HGSOC. They are prevalent in younger women with a median age of 55 years and constitute less than 5% of all OCs [65]. Though LGSOCs are chemoresistant they are treated the same way as HGSOCs with platinum/taxane drugs. The increased survival rate in LGSOCs is attributed to its longer disease trajectory and complete resection of the tumor post-primary cytoreductive surgery [66].

LGSOCs are characterized by activation of the mitogen-activated protein kinase (MAPK) pathway in 80% cases. KRAS (54%), BRAF (33%), NRAS (26%), and ERBB2, the upstream regulators of MAPK pathways are reported to be mutated, with mutations in BRAF/KRAS considered as good prognostic markers [67]. Due to the high prevalence of activated MAPK pathway in LGSOCs, MEK inhibitors (Trametinib, Selumetinib, Pimasertib, Binimetinib) are among the druggable targets for these cancers and some are under evaluation [65]. Recurrent mutations in PIK3CA, FFAR1, USP9X (11%) and EIF1AX (15%) are reported as driver mutations [68]. USP9X and EIF1AX are regulators of the mTOR pathway which are downstream effectors of the MAPK pathway. The use of Metformin, an inhibitor of the mTOR pathway, along with MEK inhibitor (Trametinib) has been reported to show an inhibitory effect in various LGSOCs cell lines [69]. Taken together, MEK inhibitors and Metformin are potential candidates for targeted therapies. CDK4/6 inhibitors, (ribociclib and abemaciclib) are under clinical trials for LGSOCs [65]. Endocrine therapy using letrozole, anastrozole or tamoxifen used as maintenance therapy has been reported to be beneficial in LGSOCs due to estrogen and progesterone receptors expressions [70].

2.1.3 Endometrioid carcinomas

Endometrioid carcinomas (ECs) are the second most common EOCs representing 10% of all OCs [71]. They are diagnosed in women in the age range of 40–70 years and are associated with a good prognosis. As its name suggests they are associated with endometriosis and are thought to be derived from the endometrium [72]. Endometriosis, menopausal hormone therapy, HBOC syndrome, Lynch syndrome and late menopause are some of the risk factors associated with ECs [14, 73].

One of the most mutated genes reported in ECs is ARID1A at a frequency of 30%. ARID1A is a component of the SWI/SNF chromatin remodeling complex. Targeting ARID1A with HDAC inhibitors have been reported to be effective in mice models harboring ARID1A tumor mutation [74]. CTNNB1, of the β-catenin signaling is also reported to be mutated at a rate of 25–60%. β-catenin signaling is a conserved pathway involved in development implicated in other epithelial cancers but its oncogenic role is less understood [75]. Other less frequent mutations are KRAS/BRAF (20%), which are regulators of MAPK pathways, PIK3CA (12%), and TP53 (25%) [76]. PTEN mutations with frequent loss of heterozygosity (45–75%) is also reported [52]. PTEN is a tumor-suppressor gene that is a negative regulator of the PI3K pathway and is also the most mutated in the related endometrial cancers [77]. The multiple mutational spectra of ECs warrants the investigation of combination therapy using MEK inhibitors (trametinib, MEK162), TP53 activators (APR-246), and PI3K inhibitors (idelalisib, voxtalisib). Only 14% of EC cases are reported to be BRCA mutation carriers [78], and HBOC syndrome being one of the risk factors for ECs, PARP inhibitors are a viable option for targeted therapies.

2.1.4 Mucinous ovarian carcinomas

Mucinous carcinomas (MOCs) are a rare subset of EOCs accounting for 2–3% of all OCs. They are histologically characterized by high levels of intracellular mucin. MOCs are more prevalent in women below 40 years and unlike other EOC types, the only risk factor identified is smoking [14, 79]. Early-stage MOCs have an excellent prognosis and beyond stage II, they are addressed by standard chemotherapeutic agents with poor outcomes, as these tumors are chemoresistant.

Though rare, MOCs have been well characterized. The predominant mutation present in MOCs is KRAS mutations reported in 66% of cases [79, 80]. A recent large cohort study identified many other mutations in MOCs besides KRAS in varying degrees which are TP53 mutation, HER2 amplification (a member of the epidermal growth factor receptor family), PIK3CA/PTEN (regulator of PI3K-PTEN-AKT pathway), BRAF mutation, CTNNB1/APC mutations (regulator of Wnt-signaling pathway), and ARID1A mutation (a member of the SWI/SNF family) [79, 80]. One of the potential drugs for the treatment of MOCs is 5-fluorouracil. MOCs and mucinous colorectal cancer (CRC) share a similar mutational profile with unfavorable outcome [81]. 5-fluorouracil, which is currently utilized for CRC treatment has been effective in various MOC cell lines in combination with oxaliplatin [82]. Moreover, the multiple mutational spectra reported in MOCs are a great avenue for identifying the most potent target for tailored therapies. Some targeted drugs like BRAF inhibitors, PI3K inhibitors are already being investigated in various other cancer types. Combinatorial therapy using dual inhibitors is warranted for MOC treatment due to its varied mutational landscape.

2.1.5 Clear-cell carcinomas

Clear cell carcinomas (CCCs) of the ovary constitute >5% of all OCs and 10% of all EOCs [83]. The incidence rates of CCCs vary by ethnicity; the majority of the cases are reported in East Asian countries (mainly Japan) for unknown reasons [84]. They are mostly diagnosed in younger women with an option of fertility-sparing surgery before standard chemotherapy. These are chemoresistant tumors with a poor prognosis if diagnosed at an advanced stage, but most of these cases are diagnosed early with a good prognosis [83]. They are a distinct class of EOCs thought to arise from endometriosis or clear cell adenofibroma, hence they are associated with endometriosis which is thought to be the precursor for CCC manifestation and this association is considered a good prognosis [85]. Late menopause and endometriosis are considered to be the highest risk factors for developing CCCs.

The most common genomic alterations identified in CCCs are activating mutations in PIK3CA, a regulator of the PI3K-PTEN-AKT pathway (50%), and loss of function in ARID1A, component of SWI/SNF chromatin remodeling complex (50%) [86]. Other mutations reported in varying degrees are MET gene amplification, mutations in ARID1B, SMARCA4, ERBB2, PIK3CA, PIK3R1, AKT2, PTEN, KRAS, PPP2R1A, TP53, TERT promoter, and ZNF217 overexpression [85, 87]. Antioxidant genes like Glutathione peroxidase 3 (GPX3), glutaredoxin (GLRX), and superoxide dismutase 2 (SOD2) are reported to be highly expressed in CCCs rendering them resistant to chemotherapy [88]. A recent report on the pharmacological inhibition of EZH2 for loss of function of ARID1A has shown considerable promise in treating CCCs [89]. The overexpression of the transcription factor ZNF217 is a poor prognostic marker. In-vitro studies in ZNF217-overexpressing cells treated with triciribine, a DNA synthesis inhibitor, have shown inhibitory effects suggesting ZNF217 be a druggable target [90]. Targeting PI3K/AKT/mTOR pathways using PI3K inhibitor (idelalisib, Voxtalisib) or mTOR pathway inhibitor (Metformin) are other viable options.

2.2 Sex cord-stromal tumors (SCSTs)

The rare ovarian sex cord-stromal tumors (SCSTs) constitute 8% of all OCs and are diagnosed in broad age groups with mixed prognosis [91]. These neoplasms originate from the stromal cells and/or the sex chord cells of the ovary, which are involved in the endocrine function of producing the female sex hormones, therefore unlike EOCs, they present with hormone-related disorders. Certain hereditary cancer syndromes predispose patients towards SCST. Based on the WHO classification of OCs, the various subtypes of SCSTs with their incidence, risk factors, prognosis, and molecular alterations are outlined in Table 1 [92].

SCST subtypesIncidence ratesIncident age groupsRisk factorsPrognosisChromatic alteration
Stromal tumors
Fibroma4% of all OCs~ 40 yearsMeigs’ syndromeGood
Thecoma0.5–1% of all OCs26–86 yearsPoorFOXL2 (~21%)
Fibrosarcoma20–73 yearsPoor
Leydig cell tumor0.1% of all SCSTPost-menopausal womenGood
Steroid cell tumor0.1% of all SCST~ 43 yearsCushing syndromeGood
Sclerosing stromal tumor>0.1% of all SCST<30 yearsGood
Sex-chord tumors
Adult granulosa cell tumor5% of all OCs, 70% of all SCSTs24–84 yearsPeutz Jeghers syndrome, Potters syndromePoorFOXL2 mutation (> 95%), TERT mutations (~40%), AKT1 amplification (~60%)
Juvenile granulosa tumor5% of all GCTs8–45 yearsOllier disease, Maffucci diseaseGoodAKT1 amplification (~60%), GNAS mutations (~30%)
Sertoli cell tumor2–76 yearsPeutz Jeghers syndromeGood
Sex chord tumor with annular tubules1.4% of all SCST5–39 yearsPeutz Jeghers syndromeFavorable
Mixed sex chord-stromal tumors
Sertoli-Leydig cell tumor0.5% of all OCs>30 yearsDicer syndromeGoodGermline and somatic DICER1 mutations (60%)

Table 1.

Sex cord-stromal tumors subtypes: epidemiology, and molecular alterations.

Due to the rarity of these tumor types, the molecular characteristics of only a few of these subtypes are reported. The cancers arising in the ovary’s granulosa cells are the most common in this group comprising 2–5% of all OCs [93]. Granulosa cells are somatic cells involved in folliculogenesis and ovulation, the variant adult granulosa cell tumors (AGCTs), which are estradiol producing are the most common in this group constituting 70% of all SCSTs [94]. Inhibin, a gonadal hormone secreted by granulosa cells, is reported to be elevated in GCT patients [95]. Inhibin level and CA-125 are utilized as a diagnostic biomarker to assess disease progression in GCTs [96]. 97% of AGCTs are characterized by the ubiquitous presence of FOXL2 mutations, a component of the TGFβ pathway [95]. The pleiotropic TGFβ pathway is reported to be deregulated in many cancers conferring chemoresistance and metastasis [97]. Moreover, TERT promoter mutations are reported in 40% of recurrent AGCT cases with poor prognosis [98]. Few small cohort studies of AGCTs, and juvenile granulosa cell tumors (JGCTs), have reported amplification in AKT leading to possible dysregulations in PI3K/AKT pathways [99, 100]. Activating GNAS mutations involved in tumor invasion are reported in 30% of JGCTs with aggressive nature [101]. The notch signaling pathway is also reported to be altered in GCTs [102]. Estrogen producing thecomas, composed of pure stromal cells are also reported to harbor FOXL2 mutation at a rate of 21% [103]. Sertoli-Leydig cell tumors (SLCTs), which belong to mixed-sex chord and stromal cells are androgen-secreting tumors that induce varying degrees of virilization (male physical characteristics) [104]. Mutation in DICER1, an endoribonuclease involved in microRNA biogenesis, is reported with a high frequency of 88% in undifferentiated SLCTs [105].

Targeting Activin A of the TGFβ pathway and aromatase, a downstream target of FOXL2 has been reported promising for targeted therapies [106, 107]. TERT promoter mutations are present in various cancer types and are reported to activate the oncogenic MAPK pathway; targeting this pathway using MEK inhibitors (trametinib, MEK162) are potential treatment options [108]. Besides, other druggable pathways for GCTs include PI3K and NOTCH pathways. Identifying drugs targeting DICER1 is warranted which could provide novel modalities for tailored therapies for SLCTs.

2.3 Ovarian germ cell tumors (OGCTs)

Ovarian germ cell tumors (OGCTs) of the ovary are rare ovarian neoplasms comprising 2–3% of all OCs [109]. These histologically variant heterogeneous neoplasms arise in the egg or ovum, the ovary’s primordial germ cell. They primarily manifest in young and adolescent women with excellent prognosis if diagnosed in earlier stages [110]. These tumors are chemosensitive allowing fertility-sparing surgery in most cases [111]. A recent small cohort study reported a low mutational burden in OGCTs explaining their chemosensitive disposition [112]. OGCTs are classified into dysgerminomas, immature teratomas, yolk sac tumors, and mixed germ cell tumors in order of their frequency. Embryonal carcinomas, choriocarcinomas, and malignant struma ovarii tumors are other very rare forms of OGCTs [113]. The understudied, very rare mixed germ cell tumors are the only aggressive OGCT subtype with poor prognosis [114]. There are no risk factors identified for OGCTs but certain genetic diseases like Turner’s syndrome, Triple X syndrome, and Swyer syndrome are reported to be high-risk factors for dysgerminomas [115]. The incidence rate, prognosis, risk factors, and their molecular characteristics are outlined in Table 2.

OGCT subtypesIncidence ratesIncident age groupsPrognosisChromatic alteration
Dysgerminoma40% of OGCTs19–23 yearsGoodKIT mutation (30–50%), 12p amplifications harboring KRAS (80%)
Immature teratoma~35% of OGCTs18–36 yearsGood
Yolk sac tumors15% of OGCTs15–40 yearsGoodPIK3CA or AKT1 mutation (72%), 12p amplifications harboring KRAS (60%)
Mixed germ cell tumors5% of OGCTs<20 yearsPoor12p amplifications harboring KRAS (~40%)

Table 2.

Ovarian germ cell tumors subtypes: epidemiology and molecular alterations.

The most frequent mutations reported in OGCTs are KIT mutations and 12p amplification, which harbor KRAS [112]. The OGCT subtype, dysgerminomas harbor 12p amplification and KIT mutation at a frequency of 80% and 30–50%, respectively [116]. KIT is a proto-oncogene involved in PI3K/AKT/mTOR, JAK/STAT and MAPK pathways [117], whereas the oncogene KRAS is involved in the tumor development pathway of Ras/Raf/MEK/ERK pathway [118]. The aneuploid, yolk sac tumors are reported to harbor PI3K and AKT1 mutations, besides KRAS altering PI3K/AKT/mTOR pathway. The TGFβ/BMP and Wnt/β-catenin signaling pathways are also reported to be activated in yolk sac tumors [116]. Few druggable targets of these pathways like AKT inhibitor (afuresertib) and MEK inhibitor (trametinib) are already under clinical trials for various OCs [119].

2.4 Small cell carcinoma of the ovary (SCCO)

Small cell carcinoma of the ovary (SCCO) is a group of extremely rare OCs accounting for <1% of all OCs [120]. Their biology is poorly understood as their cellular lineage is unknown. Based on histologic characterization, SCCO is classified into hypercalcemic type (SCCO-HT), which is chemoresistant and pulmonary type (SCCO-PT) which is chemo-sensitive. These are highly malignant cancers with an average survival of 5.7 years. The incidence rate, prognosis, risk factors and molecular characteristics are outlined in Table 3.

SCCO subtypesIncidence ratesIncident age groupsPrognosisChromatic alteration
SCCO-hypercalcemic type<1% of all OCs<40 yearsPoorSMARCA mutation (90%)
SCCO- pulmonary type<1% of all OCs<59 yearsPoorNone reported

Table 3.

Small cell carcinoma of the ovary subtypes: Epidemiology, and molecular alterations.

One of the significant mutations identified in 90% of cases of SCCOHT is germline or somatic mutations of SMARCA4 [121]. SMARCA4 mutation is considered one of the hallmarks of SCCOHT, it is a key component of the switching/sucrose non-fermenting (SWI/SNF) chromatin remodeling complex [122, 123]. The loss of function of SMARCA4 leads to the upregulation of EZH2, the catalytic subunit of the PRC2 complex which is utilized as a druggable target for SCCOHT [124]. Targeting EZH2 using tazemetostat has reported antiproliferative and antitumor effects in SCCOHT cell lines [125]. Moreover, a recent study has reported oncolytic viruses’ effect on SCCOHT derived cell line BIN-1 in reducing its proliferation >75%, which holds promise in developing targeted therapies [126]. Some of the broad categories of drugs being investigated for SCCOHT and some of which are already in clinical trials, include tyrosine kinase inhibitors, immune checkpoint inhibitors and HDAC inhibitors [127]. There are no studies reported on the molecular characterization and pathogenesis of SCCO-PT due to its rarity.

Advertisement

3. Potential drugs for targeted therapies in OCs

Presently, targeted therapy is employed only to improve the efficacy of standard therapy in OC treatment with drugs such as bevacizumab, an anti-angiogenic agent which is licensed for use as front-line therapy for advanced OCs [57] and olaparib, a PARP inhibitor which is now approved for first-line maintenance therapy for patients with relapsed BRCA-mutated OCs [128]. Very recently, the combination of bevacizumab and olaparib is FDA approved for first-line maintenance treatment in advanced OCs with HRD positive status [129].

Generic drugs being investigated for a variety of OC types are receptor tyrosine kinase (RTK) inhibitors. RTK inhibitors have been reported to be efficacious in treating a variety of malignant cancers by inhibiting tumor cell proliferation via blocking the signal transduction cascade. For e.g., Ponatinib is a multi-tyrosine kinase inhibitor that targets pathways like EGFR, FGFR, PDGFR, and VEGFR all of which are aberrantly activated in various cancer types [130]. Other RTK inhibitors being investigated for OCs are Palbociclib, Abemaciclib and Ribociclib [131, 132]. Likewise, immunotherapy using immune checkpoint inhibitors like Pembrolizumab and Nivolumab has been revolutionary in oncology research. These are monoclonal antibodies that trigger the immune T-cell activation to attack the cancer cells and Pembrolizumab is already under clinical trial for various cancer types [133, 134]. Epigenetic abnormalities being the hallmarks of cancers, epigenetic modulators like HDAC inhibitors have shown great promise as anti-cancer drugs. HDAC inhibitors like Vorinostat, Panobinostat, Quisinostat, and Trichostatin are under investigation for targeted therapies for OCs [126, 135].

Advertisement

4. Role of miRNAs in ovarian cancer

miRNAs are single-stranded RNA nucleotides that regulate gene expression. In the human body, they are reported to be involved in regulating around 60% of genes affecting various cellular and biological processes. Each miRNA has multiple gene targets or multiple miRNAs can act on one target gene. They can function either as an oncogene or a tumor suppressor and their expressions in cancer cells are deregulated [136]. The miRNA expression profile for each OC subtype is reported to be distinct, with a subset of miRNAs downregulated or upregulated [137]. The miRNA signatures identified in various cancer types are being investigated for their utility as cancer biomarkers in tumor diagnosis, prognosis and therapeutic outcome.

The sensitivity of a cancer drug profoundly affects treatment efficacy and prognosis. miRNAs are involved in conferring chemo-sensitive or chemoresistant phenotype by regulating the drug-resistance related genes [138]. Therefore, manipulating the expression levels of specific miRNAs can aid in drug sensitivity. As previously mentioned, the sensitivity for platinum drugs varies among each OC subtype, and this profoundly affects the treatment efficacy and prognosis. Though still in its infancy, targeting miRNA holds great promise for a more customized therapeutic approach. Here, we highlight the key miRNAs reported in recent literature, which are deregulated in the various OC subtypes (Table 4).

Ovarian cancer subtypeUpregulatedDownregulatedReferences
Serous Ovarian cancermiR-429, miR-141, miR-200c, miR-93, miR-16, miR-20a, miR-21, miR-27a, miR-200a, miR-200b, miR-200c, miR-203, miR-205, miR-375, miR-145miR-320c, miR-383, let-7b, miR-99a, miR-125b, miR-145, miR-100, miR-31, miR-137, miR-132, miR-26a[138, 139]
Clear-cell carcinomasmiR-93, miR-126, miR-338, miR-200a, miR-200b, miR-30a, miR-141, miR-182, miR-200a, miR-510, miR-509,miR-383, miR-424, miR-127, miR-155, miR-99b[138, 139]
Mucinous ovarian carcinomamiR-192, miR-194[137]
Endometrioid carcinomasmiR-7, miR-429, miR-21, miR-29a, miR-92, miR-30c1, miR-126, miR-126, miR-29amiR-342, miR-181a, miR-450b, miR-155, miR-25, miR-93, miR-127, miR-99b[136, 137, 138]
Ovarian Germ cell tumorsmiR-373-3p, miR-372-3p and miR-302c-3p, mir-302–367 cluster, mir-371–373 cluster, miR-146b, miR-155, miR-182miR-199a-5p, miR-214-5p and miR-202-3p, Let-7[139, 140]

Table 4.

Deregulated miRNAs in ovarian cancer subtypes.

Advertisement

5. Conclusion

The global incidence rate for OC is expected to increase by 47% by 2040 [141]. Except for the emergence of PARP inhibitors in women with HRD HGSOC tumors, the conventional treatment protocol for other OC subtypes has remained the same since the 1980s, with no significant impact on survival rates. Screening for high-grade OCs remains a challenge. With the advances in the high throughput screening technologies, the focus is warranted to shift towards translational research to treat each OC subtype for their underlying genomic aberrations.

References

  1. 1. Swerdlow M. Mesothelioma of the pelvic peritoneum resembling papillary cystadenocarcinoma of the ovary; case report. Am J Obstet Gynecol 1959; 77: 197-200
  2. 2. Jacobs IJ, Menon U. Progress and challenges in screening for early detection of ovarian cancer. Mol Cell Proteomics 2004; 3: 355-366
  3. 3. Ferlay J, Colombet M, Soerjomataram I, et al. Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. Int J Cancer 2019; 144: 1941-1953
  4. 4. Shabir S, Gill PK. Global scenario on ovarian cancer – Its dynamics, relative survival, treatment, and epidemiology. AUJMSR 2020; 2: 17-25
  5. 5. Mohammadian M, Ghafari M, Khosravi B, et al. Variations in the Incidence and Mortality of Ovarian Cancer and Their Relationship with the Human Development Index in European Countries in 2012. Biomedical Research and Therapy 2017; 4: 1541
  6. 6. Torre LA, Trabert B, DeSantis CE, et al. Ovarian cancer statistics, 2018. CA Cancer J Clin 2018; 68: 284-296
  7. 7. Nielsen FC, van Overeem Hansen T, Sørensen CS. Hereditary breast and ovarian cancer: new genes in confined pathways. Nat Rev Cancer 2016; 16: 599-612
  8. 8. Nakamura K, Banno K, Yanokura M, et al. Features of ovarian cancer in Lynch syndrome (Review). Mol Clin Oncol 2014; 2: 909-916
  9. 9. Collaborative Group On Epidemiological Studies Of Ovarian Cancer, Beral V, Gaitskell K, et al. Menopausal hormone use and ovarian cancer risk: individual participant meta-analysis of 52 epidemiological studies. Lancet 2015; 385: 1835-1842
  10. 10. Trabert B, Wentzensen N, Yang HP, et al. Ovarian cancer and menopausal hormone therapy in the NIH-AARP diet and health study. Br J Cancer 2012; 107: 1181-1187
  11. 11. Pearce CL, Templeman C, Rossing MA, et al. Association between endometriosis and risk of histological subtypes of ovarian cancer: a pooled analysis of case-control studies. Lancet Oncol 2012; 13: 385-394
  12. 12. D Farhud D, Zokaei S, Keykhaei M, et al. Strong Evidences of the Ovarian Carcinoma Risk in Women after IVF Treatment: A Review Article. Iran J Public Health 2019; 48: 2124-2132
  13. 13. Practice Committee of the American Society for Reproductive Medicine. Electronic address: ASRM@asrm.org, Practice Committee of the American Society for Reproductive Medicine. Fertility drugs and cancer: a guideline. Fertil Steril 2016; 106: 1617-1626
  14. 14. Wentzensen N, Poole EM, Trabert B, et al. Ovarian Cancer Risk Factors by Histologic Subtype: An Analysis From the Ovarian Cancer Cohort Consortium. J Clin Oncol 2016; 34: 2888-2898
  15. 15. Lundberg FE, Iliadou AN, Rodriguez-Wallberg K, et al. The risk of breast and gynecological cancer in women with a diagnosis of infertility: A nationwide population-based study. Eur J Epidemiol 2019; 34: 499-507
  16. 16. McGuire V, Hartge P, Liao LM, et al. Parity and Oral Contraceptive Use in Relation to Ovarian Cancer Risk in Older Women. Cancer Epidemiol Biomarkers Prev 2016; 25: 1059-1063
  17. 17. Rice MS, Murphy MA, Tworoger SS. Tubal ligation, hysterectomy and ovarian cancer: A meta-analysis. J Ovarian Res 2012; 5: 1-16
  18. 18. Michels KA, Brinton LA, Pfeiffer RM, et al. Oral Contraceptive Use and Risks of Cancer in the NIH-AARP Diet and Health Study. Am J Epidemiol 2018; 187: 1630-1641
  19. 19. Fathalla MF. Incessant ovulation and ovarian cancer - a hypothesis re-visited. Facts Views Vis Obgyn 2013; 5: 292-297
  20. 20. Madsen C, Baandrup L, Dehlendorff C, et al. Tubal ligation and salpingectomy and the risk of epithelial ovarian cancer and borderline ovarian tumors: a nationwide case-control study. Acta Obstet Gynecol Scand 2015; 94: 86-94
  21. 21. Kadry Taher M, Farhat N, Karyakina NA, et al. Critical review of the association between perineal use of talc powder and risk of ovarian cancer. Reprod Toxicol 2019; 90: 88-101
  22. 22. Steffen JE, Tran T, Yimam M, et al. Serous Ovarian Cancer Caused by Exposure to Asbestos and Fibrous Talc in Cosmetic Talc Powders-A Case Series. J Occup Environ Med 2020; 62: e65–e77
  23. 23. Zhou Z, Zeng F, Yuan J, et al. Pelvic inflammatory disease and the risk of ovarian cancer: a meta-analysis. Cancer Causes Control 2017; 28: 415-428
  24. 24. Siegel RL, Miller KD, Jemal A. Cancer Statistics, 2017. CA Cancer J Clin 2017; 67: 7-30
  25. 25. Momenimovahed Z, Tiznobaik A, Taheri S, et al. Ovarian cancer in the world: epidemiology and risk factors. Int J Womens Health 2019; 11: 287-299
  26. 26. Jasen P. From the ‘silent killer’ to the ‘whispering disease’: ovarian cancer and the uses of metaphor. Med Hist 2009; 53: 489-512
  27. 27. Gajjar K, Ogden G, Mujahid MI, et al. Symptoms and risk factors of ovarian cancer: a survey in primary care. ISRN Obstet Gynecol 2012; 2012: 754197
  28. 28. Ebell MH, Culp MB, Radke TJ. A Systematic Review of Symptoms for the Diagnosis of Ovarian Cancer. Am J Prev Med 2016; 50: 384-394
  29. 29. Menon U, Karpinskyj C, Gentry-Maharaj A. Ovarian Cancer Prevention and Screening. Obstet Gynecol 2018; 131: 909-927
  30. 30. Dochez V, Caillon H, Vaucel E, et al. Biomarkers and algorithms for diagnosis of ovarian cancer: CA125, HE4, RMI and ROMA, a review. J Ovarian Res 2019; 12: 28
  31. 31. Shen Y, Zhao L, Lu S. Diagnostic performance of HE4 and ROMA among Chinese women. Clin Chim Acta 2020; 500: 42-46
  32. 32. Kadija S, Stefanovic A, Jeremic K, et al. The utility of human epididymal protein 4, cancer antigen 125, and risk for malignancy algorithm in ovarian cancer and endometriosis. Int J Gynecol Cancer 2012; 22: 238-244
  33. 33. Zhang L, Chen Y, Wang K. Comparison of CA125, HE4, and ROMA index for ovarian cancer diagnosis. Curr Probl Cancer 2019; 43: 135-144
  34. 34. Cui R, Wang Y, Li Y, et al. Clinical value of ROMA index in diagnosis of ovarian cancer: meta-analysis. Cancer Manag Res 2019; 11: 2545-2551
  35. 35. Shulman LP, Francis M, Bullock R, et al. Clinical Performance Comparison of Two In-Vitro Diagnostic Multivariate Index Assays (IVDMIAs) for Presurgical Assessment for Ovarian Cancer Risk. Adv Ther 2019; 36: 2402-2413
  36. 36. Wang Y, Li L, Douville C, et al. Evaluation of liquid from the Papanicolaou test and other liquid biopsies for the detection of endometrial and ovarian cancers. Sci Transl Med; 10. Epub ahead of print 21 March 2018. DOI: 10.1126/scitranslmed.aap8793
  37. 37. Cohen JD, Li L, Wang Y, et al. Detection and localization of surgically resectable cancers with a multi-analyte blood test. Science 2018; 359: 926-930
  38. 38. Wild CP, Weiderpass E, Stewart BW, editors (2020). World Cancer Report: Cancer Research for Cancer Prevention. Lyon, France: International Agency for Research on Cancer. Available from: http://publications.iarc.fr/586. Licence: CC BY-NC-ND 3.0 IGO
  39. 39. Chen X, Wu Y, Dong H, et al. Platinum-based agents for individualized cancer treatment. Curr Mol Med 2013; 13: 1603-1612
  40. 40. Targeted therapy of ovarian cancer including immune check point inhibitor. Korean J Intern Med. DOI: 10.3904/kjim.2017.008
  41. 41. Corrado G, Salutari V, Palluzzi E, et al. Optimizing treatment in recurrent epithelial ovarian cancer. Expert Rev Anticancer Ther 2017; 17: 1147-1158
  42. 42. Monk BJ, Coleman RL. Changing the paradigm in the treatment of platinum-sensitive recurrent ovarian cancer: from platinum doublets to nonplatinum doublets and adding antiangiogenesis compounds. Int J Gynecol Cancer 2009; 19 Suppl 2: S63–S67
  43. 43. Motohara T, Masuda K, Morotti M, et al. An evolving story of the metastatic voyage of ovarian cancer cells: cellular and molecular orchestration of the adipose-rich metastatic microenvironment. Oncogene 2018; 38: 2885-2898
  44. 44. Mitra AK. Ovarian Cancer Metastasis: A Unique Mechanism of Dissemination. Tumor Metastasis. Epub ahead of print 2016. DOI: 10.5772/64700
  45. 45. Temkin SM, Bergstrom J, Samimi G, et al. Ovarian Cancer Prevention in High-risk Women. Clin Obstet Gynecol 2017; 60: 738-757
  46. 46. Grimbizis GF, Tarlatzis BC. The use of hormonal contraception and its protective role against endometrial and ovarian cancer. Best Pract Res Clin Obstet Gynaecol 2010; 24: 29-38
  47. 47. WebPathology, https://www.webpathology.com/contact.asp (accessed 6 December 2020)
  48. 48. Lheureux S, Gourley C, Vergote I, et al. Epithelial ovarian cancer. Lancet 2019; 393: 1240-1253
  49. 49. Karst AM, Levanon K, Drapkin R. Modeling high-grade serous ovarian carcinogenesis from the fallopian tube. Proc Natl Acad Sci U S A 2011; 108: 7547-7552
  50. 50. George SHL, Garcia R, Slomovitz BM. Ovarian Cancer: The Fallopian Tube as the Site of Origin and Opportunities for Prevention. Front Oncol 2016; 6: 108
  51. 51. Zhang Q , Bykov VJN, Wiman KG, et al. APR-246 reactivates mutant p53 by targeting cysteines 124 and 277. Cell Death Dis 2018; 9: 439
  52. 52. Moufarrij S, Dandapani M, Arthofer E, et al. Epigenetic therapy for ovarian cancer: promise and progress. Clin Epigenetics 2019; 11: 7
  53. 53. Zanjirband M, Gentles L, Curtin NJ, et al. 55P Evaluation of MDM2-p53 antagonists, nutlin-3 and RG7388, combined with the PARPi rucaparib in primary cultures of ovarian cancer. Annals of Oncology 2020; 31: S1234
  54. 54. Frey MK, Pothuri B. Homologous recombination deficiency (HRD) testing in ovarian cancer clinical practice: a review of the literature. Gynecol Oncol Res Pract 2017; 4: 4
  55. 55. Network TCGAR, The Cancer Genome Atlas Research Network. Integrated genomic analyses of ovarian carcinoma. Nature 2011; 474: 609-615
  56. 56. Konstantinopoulos PA, Ceccaldi R, Shapiro GI, et al. Homologous Recombination Deficiency: Exploiting the Fundamental Vulnerability of Ovarian Cancer. Cancer Discov 2015; 5: 1137-1154
  57. 57. Gadducci A, Guarneri V, Peccatori FA, et al. Current strategies for the targeted treatment of high-grade serous epithelial ovarian cancer and relevance of BRCA mutational status. J Ovarian Res 2019; 12: 1-8
  58. 58. Dziadkowiec KN, Gąsiorowska E, Nowak-Markwitz E, et al. PARP inhibitors: review of mechanisms of action and BRCA1/2 mutation targeting. Prz Menopauzalny 2016; 15: 215-219
  59. 59. Kaufman B, Shapira-Frommer R, Schmutzler RK, et al. Olaparib monotherapy in patients with advanced cancer and a germline BRCA1/2 mutation. J Clin Oncol 2015; 33: 244-250
  60. 60. Banerjee SN, Lord CJ. First-line PARP inhibition in ovarian cancer — standard of care for all? Nature Reviews Clinical Oncology 2020; 17: 136-137
  61. 61. Ethier J-L, Lheureux S, Oza AM. The role of niraparib for the treatment of ovarian cancer. Future Oncology 2018; 14: 2565-2577
  62. 62. Iyengar M, O’Hayer P, Cole A, et al. CDK4/6 inhibition as maintenance and combination therapy for high grade serous ovarian cancer. Oncotarget 2018; 9: 15658-15672
  63. 63. Lisio M-A, Fu L, Goyeneche A, et al. High-Grade Serous Ovarian Cancer: Basic Sciences, Clinical and Therapeutic Standpoints. Int J Mol Sci; 20. Epub ahead of print 22 February 2019. DOI: 10.3390/ijms20040952
  64. 64. Pfisterer J, Shannon CM, Baumann K, et al. Bevacizumab and platinum-based combinations for recurrent ovarian cancer: a randomised, open-label, phase 3 trial. Lancet Oncol 2020; 21: 699-709
  65. 65. Gadducci A, Cosio S. Therapeutic Approach to Low-Grade Serous Ovarian Carcinoma: State of Art and Perspectives of Clinical Research. Cancers; 12. Epub ahead of print 23 May 2020. DOI: 10.3390/cancers12051336
  66. 66. Grisham RN, Iyer G. Low-Grade Serous Ovarian Cancer: Current Treatment Paradigms and Future Directions. Curr Treat Options Oncol 2018; 19: 496
  67. 67. Kaldawy A, Segev Y, Lavie O, et al. Low-grade serous ovarian cancer: A review. Gynecol Oncol 2016; 143: 433-438
  68. 68. Hunter SM, Anglesio MS, Ryland GL, et al. Molecular profiling of low grade serous ovarian tumours identifies novel candidate driver genes. Oncotarget 2015; 6: 37663-37677
  69. 69. Mert I, Chhina J, Allo G, et al. Synergistic effect of MEK inhibitor and metformin combination in low grade serous ovarian cancer. Gynecologic Oncology 2017; 146: 319-326
  70. 70. Gershenson DM, Bodurka DC, Coleman RL, et al. Hormonal Maintenance Therapy for Women With Low-Grade Serous Cancer of the Ovary or Peritoneum. Journal of Clinical Oncology 2017; 35: 1103-1111
  71. 71. Nasioudis D, Latif NA, Simpkins F, et al. Adjuvant chemotherapy for early stage endometrioid ovarian carcinoma: An analysis of the National Cancer Data Base. Gynecol Oncol 2020; 156: 315-319
  72. 72. Terada T. Endometrioid adenocarcinoma of the ovary arising in atypical endometriosis. Int J Clin Exp Pathol 2012; 5: 924-927
  73. 73. Helder-Woolderink JM, Blok EA, Vasen HFA, et al. Ovarian cancer in Lynch syndrome; a systematic review. Eur J Cancer 2016; 55: 65-73
  74. 74. Fukumoto T, Park PH, Wu S, et al. Repurposing Pan-HDAC Inhibitors for ARID1A-Mutated Ovarian Cancer. Cell Rep 2018; 22: 3393-3400
  75. 75. Kim S, Jeong S. Mutation Hotspots in the β-Catenin Gene: Lessons from the Human Cancer Genome Databases. Mol Cells 2019; 42: 8-16
  76. 76. Elsherif S, Javadi S, Viswanathan C, et al. Low-grade epithelial ovarian cancer: what a radiologist should know. Br J Radiol 2019; 92: 20180571
  77. 77. Papa, Papa, Pandolfi. The PTEN–PI3K Axis in Cancer. Biomolecules 2019; 9: 153
  78. 78. Lynch HT, Casey MJ, Snyder CL, et al. Hereditary ovarian carcinoma: heterogeneity, molecular genetics, pathology, and management. Mol Oncol 2009; 3: 97-137
  79. 79. Babaier A, Ghatage P. Mucinous Cancer of the Ovary: Overview and Current Status. Diagnostics 2020; 10: 52
  80. 80. Gorringe KL, Cheasley D, Wakefield MJ, et al. Therapeutic options for mucinous ovarian carcinoma. Gynecol Oncol 2020; 156: 552-560
  81. 81. Kelemen LE, Köbel M. Mucinous carcinomas of the ovary and colorectum: different organ, same dilemma. The Lancet Oncology 2011; 12: 1071-1080
  82. 82. Sato S, Itamochi H, Kigawa J, et al. Combination chemotherapy of oxaliplatin and 5-fluorouracil may be an effective regimen for mucinous adenocarcinoma of the ovary: a potential treatment strategy. Cancer Sci 2009; 100: 546-551
  83. 83. Liu H, Xu Y, Ji J, et al. Prognosis of ovarian clear cell cancer compared with other epithelial cancer types: A population-based analysis. Oncology Letters. Epub ahead of print 2020. DOI: 10.3892/ol.2020.11252
  84. 84. Fujiwara K, Shintani D, Nishikawa T. Clear-cell carcinoma of the ovary. Ann Oncol 2016; 27 Suppl 1: i50–i52
  85. 85. Iida Y, Okamoto A, Hollis RL, et al. Clear cell carcinoma of the ovary: a clinical and molecular perspective. Int J Gynecol Cancer. Epub ahead of print 18 September 2020. DOI: 10.1136/ijgc-2020-001656
  86. 86. Huang H-N, Lin M-C, Huang W-C, et al. Loss of ARID1A expression and its relationship with PI3K-Akt pathway alterations and ZNF217 amplification in ovarian clear cell carcinoma. Mod Pathol 2014; 27: 983-990
  87. 87. Matsuzaki S, Yoshino K, Ueda Y, et al. Potential targets for ovarian clear cell carcinoma: a review of updates and future perspectives. Cancer Cell Int 2015; 15: 117
  88. 88. 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; 31. Epub ahead of print 2012. DOI: 10.1186/1756-9966-31-53
  89. 89. Bitler BG, Aird KM, Garipov A, et al. Synthetic lethality by targeting EZH2 methyltransferase activity in ARID1A-mutated cancers. Nat Med 2015; 21: 231-238
  90. 90. Littlepage LE, Adler AS, Kouros-Mehr H, et al. The transcription factor ZNF217 is a prognostic biomarker and therapeutic target during breast cancer progression. Cancer Discov 2012; 2: 638-651
  91. 91. Fuller PJ, Leung D, Chu S. Genetics and genomics of ovarian sex cord-stromal tumors. Clin Genet 2017; 91: 285-291
  92. 92. Horta M, Cunha TM. Sex cord-stromal tumors of the ovary: a comprehensive review and update for radiologists. Diagn Interv Radiol 2015; 21: 277-286
  93. 93. Khosla D, Dimri K, Pandey AK, et al. Ovarian granulosa cell tumor: clinical features, treatment, outcome, and prognostic factors. N Am J Med Sci 2014; 6: 133-138
  94. 94. Hodeib M, Tsui I, Sinno A, et al. Adult Granulosa Cell Tumor of the Ovary: Initial Evaluation and Current Treatment Paradigm. Journal of Cancer Science and Clinical Therapeutics 2018; 01: 6-18
  95. 95. Kottarathil VD, Antony MA, Nair IR, et al. Recent advances in granulosa cell tumor ovary: a review. Indian J Surg Oncol 2013; 4: 37-47
  96. 96. Robertson DM, Pruysers E, Jobling T. Inhibin as a diagnostic marker for ovarian cancer. Cancer Letters 2007; 249: 14-17
  97. 97. Colak S, ten Dijke P. Targeting TGF-β Signaling in Cancer. Trends in Cancer 2017; 3: 56-71
  98. 98. Alexiadis M, Rowley SM, Chu S, et al. Mutational Landscape of Ovarian Adult Granulosa Cell Tumors from Whole Exome and Targeted TERT Promoter Sequencing. Molecular Cancer Research 2019; 17: 177-185
  99. 99. Caburet S, Anttonen M, Todeschini A-L, et al. Combined comparative genomic hybridization and transcriptomic analyses of ovarian granulosa cell tumors point to novel candidate driver genes. BMC Cancer 2015; 15: 1-11
  100. 100. Auguste A, Bessière L, Todeschini A-L, et al. Molecular analyses of juvenile granulosa cell tumors bearing AKT1 mutations provide insights into tumor biology and therapeutic leads. Hum Mol Genet 2015; 24: 6687-6698
  101. 101. Kalfa N, Ecochard A, Patte C, et al. Activating mutations of the stimulatory g protein in juvenile ovarian granulosa cell tumors: a new prognostic factor? J Clin Endocrinol Metab 2006; 91: 1842-1847
  102. 102. Li J, Bao R, Peng S, et al. The molecular mechanism of ovarian granulosa cell tumors. J Ovarian Res 2018; 11: 1-8
  103. 103. Shah SP, Köbel M, Senz J, et al. Mutation of FOXL2 in granulosa-cell tumors of the ovary. N Engl J Med 2009; 360: 2719-2729
  104. 104. Abu-Zaid A, Azzam A, Alghuneim LA, et al. Poorly Differentiated Ovarian Sertoli-Leydig Cell Tumor in a 16-Year-Old Single Woman: A Case Report and Literature Review. Case Reports in Obstetrics and Gynecology 2013; 2013: 1-6
  105. 105. de Kock L, Terzic T, McCluggage WG, et al. DICER1 Mutations Are Consistently Present in Moderately and Poorly Differentiated Sertoli-Leydig Cell Tumors. Am J Surg Pathol 2017; 41: 1178-1187
  106. 106. Bonilla L, Oza AM. Targeting TGFβ Pathway in Adult Granulosa Cell Tumor: Opening Pandora’s Box? Clinical Cancer Research 2019; 25: 5432-5434
  107. 107. Yang AD, Curtin J, Muggia F. Ovarian adult-type granulosa cell tumor: focusing on endocrine-based therapies. International Journal of Endocrine Oncology 2018; 5: IJE08
  108. 108. Bell RJA, Rube HT, Xavier-Magalhaes A, et al. Understanding TERT Promoter Mutations: A Common Path to Immortality. Molecular Cancer Research 2016; 14: 315-323
  109. 109. Brown J, Friedlander M, Backes FJ, et al. Gynecologic Cancer Intergroup (GCIG) consensus review for ovarian germ cell tumors. Int J Gynecol Cancer 2014; 24: S48–S54
  110. 110. Smith HO, Berwick M, Verschraegen CF, et al. Incidence and survival rates for female malignant germ cell tumors. Obstet Gynecol 2006; 107: 1075-1085
  111. 111. Iavazzo C, Vorgias G, Iavazzo PE, et al. Is fertility sparing surgery a treatment option for premenopausal patients with dysgerminoma? Bratisl Lek Listy 2016; 117: 738-740
  112. 112. Van Nieuwenhuysen E, Busschaert P, Neven P, et al. The genetic landscape of 87 ovarian germ cell tumors. Gynecol Oncol 2018; 151: 61-68
  113. 113. Shaaban AM, Rezvani M, Elsayes KM, et al. Ovarian malignant germ cell tumors: cellular classification and clinical and imaging features. Radiographics 2014; 34: 777-801
  114. 114. Lee Y-L, Lai C-R, Yen M-S. Recurrent ovarian mixed germ cell tumor with unusual malignant transformation: a case report. J Ovarian Res 2019; 12: 1-6
  115. 115. Kota S, Pani J, Meher L, et al. Dysgerminoma in a female with turner syndrome and Y chromosome material: A case-based review of literature. Indian Journal of Endocrinology and Metabolism 2012; 16: 436
  116. 116. Maoz A, Matsuo K, Ciccone MA, et al. Molecular Pathways and Targeted Therapies for Malignant Ovarian Germ Cell Tumors and Sex Cord–Stromal Tumors: A Contemporary Review. Cancers 2020; 12: 1398
  117. 117. Cardoso HJ, Figueira MI, Socorro S. The stem cell factor (SCF)/c-KIT signalling in testis and prostate cancer. J Cell Commun Signal 2017; 11: 297-307
  118. 118. Guo Y, Pan W, Liu S, et al. ERK/MAPK signalling pathway and tumorigenesis (Review). Experimental and Therapeutic Medicine. Epub ahead of print 2020. DOI: 10.3892/etm.2020.8454
  119. 119. Maoz A, Ciccone MA, Matsuzaki S, et al. Emerging serine-threonine kinase inhibitors for treating ovarian cancer. Expert Opin Emerg Drugs 2019; 24: 239-253
  120. 120. Oneda E, Zorzi F, Gorio A, et al. Differential Diagnosis of Small Cell Carcinoma of the Ovary or Ovarian Metastases of Small Cell Carcinoma of the Lung: A Case Report and Review of the Literature. Case Rep Oncol 2020; 13: 822-828
  121. 121. Auguste A, Blanc-Durand F, Deloger M, et al. Small Cell Carcinoma of the Ovary, Hypercalcemic Type (SCCOHT) beyond Mutations: A Comprehensive Genomic Analysis. Cells; 9. Epub ahead of print 19 June 2020. DOI: 10.3390/cells9061496
  122. 122. Bailey S, Murray MJ, Witkowski L, et al. Biallelic somatic SMARCA4 mutations in small cell carcinoma of the ovary, hypercalcemic type (SCCOHT). Pediatr Blood Cancer 2015; 62: 728-730
  123. 123. Lu B, Shi H. An In-Depth Look at Small Cell Carcinoma of the Ovary, Hypercalcemic Type (SCCOHT): Clinical Implications from Recent Molecular Findings. J Cancer 2019; 10: 223-237
  124. 124. Wang Y, Chen SY, Karnezis AN, et al. The histone methyltransferase EZH2 is a therapeutic target in small cell carcinoma of the ovary, hypercalcaemic type. J Pathol 2017; 242: 371-383
  125. 125. Chan-Penebre E, Armstrong K, Drew A, et al. Selective Killing of SMARCA2- and SMARCA4-deficient Small Cell Carcinoma of the Ovary, Hypercalcemic Type Cells by Inhibition of EZH2: and Preclinical Models. Mol Cancer Ther 2017; 16: 850-860
  126. 126. Gamwell LF, Gambaro K, Merziotis M, et al. Small cell ovarian carcinoma: genomic stability and responsiveness to therapeutics. Orphanet J Rare Dis 2013; 8: 33
  127. 127. Tischkowitz M, Huang S, Banerjee S, et al. Small-Cell Carcinoma of the Ovary, Hypercalcemic Type–Genetics, New Treatment Targets, and Current Management Guidelines. Clinical Cancer Research 2020; 26: 3908-3917
  128. 128. Banerjee S, Gonzalez-Martin A, Harter P, et al. First-line PARP inhibitors in ovarian cancer: summary of an ESMO Open - Cancer Horizons round-table discussion. ESMO Open 2020; 5: e001110
  129. 129. Ray-Coquard I, Pautier P, Pignata S, et al. Olaparib plus Bevacizumab as First-Line Maintenance in Ovarian Cancer. N Engl J Med 2019; 381: 2416-2428
  130. 130. Tan FH, Putoczki TL, Stylli SS, et al. Ponatinib: a novel multi-tyrosine kinase inhibitor against human malignancies. Onco Targets Ther 2019; 12: 635-645
  131. 131. Lang JD, Hendricks WPD, Orlando KA, et al. Ponatinib Shows Potent Antitumor Activity in Small Cell Carcinoma of the Ovary Hypercalcemic Type (SCCOHT) through Multikinase Inhibition. Clin Cancer Res 2018; 24: 1932-1943
  132. 132. Xue Y, Meehan B, Macdonald E, et al. CDK4/6 inhibitors target SMARCA4-determined cyclin D1 deficiency in hypercalcemic small cell carcinoma of the ovary. Nat Commun 2019; 10: 558
  133. 133. Lee EK, Esselen KM, Kolin DL, et al. Combined CDK4/6 and PD-1 Inhibition in Refractory SMARCA4-Deficient Small-Cell Carcinoma of the Ovary, Hypercalcemic Type. JCO Precis Oncol 2020; 4: 736-742
  134. 134. Jelinic P, Ricca J, Van Oudenhove E, et al. Immune-Active Microenvironment in Small Cell Carcinoma of the Ovary, Hypercalcemic Type: Rationale for Immune Checkpoint Blockade. J Natl Cancer Inst 2018; 110: 787-790
  135. 135. Mottamal M, Zheng S, Huang TL, et al. Histone deacetylase inhibitors in clinical studies as templates for new anticancer agents. Molecules 2015; 20: 3898-3941
  136. 136. Gandellini P, Giovannetti E, Nicassio F. MicroRNAs in Cancer Management: Big Challenges for Small Molecules. BioMed Research International 2015; 2015: 1-2
  137. 137. Alshamrani AA. Roles of microRNAs in Ovarian Cancer Tumorigenesis: Two Decades Later, What Have We Learned? Front Oncol 2020; 10: 1084
  138. 138. Chen S-N, Chang R, Lin L-T, et al. MicroRNA in Ovarian Cancer: Biology, Pathogenesis, and Therapeutic Opportunities. Int J Environ Res Public Health; 16. Epub ahead of print 29 April 2019. DOI: 10.3390/ijerph16091510
  139. 139. Kraggerud SM, Hoei-Hansen CE, Alagaratnam S, et al. Molecular characteristics of malignant ovarian germ cell tumors and comparison with testicular counterparts: implications for pathogenesis. Endocr Rev 2013; 34: 339-376
  140. 140. Chang RK, Li X, Mu N, et al. MicroRNA expression profiles in non-epithelial ovarian tumors. Int J Oncol 2018; 52: 55-66
  141. 141. Ferlay J, Ervik M, Lam F, Colombet M, Mery L, Piñeros M, Znaor A, Soerjomataram I, Bray F (2018). Global Cancer Observatory: Cancer Tomorrow. Lyon, France: International Agency for Research on Cancer. Available from: https://gco.iarc.fr/tomorrow, accessed [01 Dec 2020]

Written By

Febina Ravindran and Bibha Choudhary

Submitted: 06 December 2020 Reviewed: 11 January 2021 Published: 05 February 2021