Next Article in Journal
Mechanism of Cs Immobilization within a Sodalite Framework: The Role of Alkaline Cations and the Si/Al Ratio
Next Article in Special Issue
Assessing the Phenotype of a Homologous Recombination Deficiency Using High Resolution Array-Based Comparative Genome Hybridization in Ovarian Cancer
Previous Article in Journal
Viral Diversity in Samples of Freshwater Gastropods Benedictia baicalensis (Caenogastropoda: Benedictiidae) Revealed by Total RNA-Sequencing
Previous Article in Special Issue
Inhibition of CDK1 by RO-3306 Exhibits Anti-Tumorigenic Effects in Ovarian Cancer Cells and a Transgenic Mouse Model of Ovarian Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Early-Onset Ovarian Cancer <30 Years: What Do We Know about Its Genetic Predisposition?

1
Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital in Prague, 128 00 Prague, Czech Republic
2
Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University and General University Hospital in Prague, 128 00 Prague, Czech Republic
3
Institute of Pathological Physiology, First Faculty of Medicine, Charles University, 128 00 Prague, Czech Republic
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(23), 17020; https://doi.org/10.3390/ijms242317020
Submission received: 25 October 2023 / Revised: 27 November 2023 / Accepted: 29 November 2023 / Published: 30 November 2023
(This article belongs to the Special Issue Gynecological Cancer 2023)

Abstract

:
Ovarian cancer (OC) is one of the leading causes of cancer-related deaths in women. Most patients are diagnosed with advanced epithelial OC in their late 60s, and early-onset adult OC diagnosed ≤30 years is rare, accounting for less than 5% of all OC cases. The most significant risk factor for OC development are germline pathogenic/likely pathogenic variants (GPVs) in OC predisposition genes (including BRCA1, BRCA2, BRIP1, RAD51C, RAD51D, Lynch syndrome genes, or BRIP1), which contribute to the development of over 20% of all OC cases. GPVs in BRCA1/BRCA2 are the most prevalent. The presence of a GPV directs tailored cancer risk-reducing strategies for OC patients and their relatives. Identification of OC patients with GPVs can also have therapeutic consequences. Despite the general assumption that early cancer onset indicates higher involvement of hereditary cancer predisposition, the presence of GPVs in early-onset OC is rare (<10% of patients), and their heritability is uncertain. This review summarizes the current knowledge on the genetic predisposition to early-onset OC, with a special focus on epithelial OC, and suggests other alternative genetic factors (digenic, oligogenic, polygenic heritability, genetic mosaicism, imprinting, etc.) that may influence the development of early-onset OC in adult women lacking GPVs in known OC predisposition genes.

1. Introduction

Ovarian cancer (OC) accounts for 4.7% of cancer-related deaths in women worldwide [1]. Early detection of OC remains challenging due to the predominance of non-specific symptoms that occur mainly at advanced clinical stages [2]. The majority of patients are diagnosed with advanced disease with an unfavorable prognosis (5-year survival rate of approximately 50%), which is even worse in cases with metastatic disease (5-year survival rate of approximately 30%) [3]. Thus, it is important to identify women at increased OC risk early, especially when we know that the proportion of hereditary OC is high, reaching even over 20% [4,5,6]. Surprisingly, despite all assumptions, the abundance of germline pathogenic/likely pathogenic variant (GPV) carriers among early-onset OC patients (<30 years) falls below 10% [7,8,9], and the genetic predisposition to early-onset OC remains uncertain.
This review focuses on the characterization of early-onset adult OC patients (diagnosed between 18 and 30 years old) from the perspective of cancer predisposition, with a special focus on epithelial OC.

2. Characteristics of Early-Onset and Late-Onset OC

Ovarian cancer is a heterogeneous group, including tumors in the ovaries, fallopian tubes, and peritoneum. Approximately 90% of OC cases are of epithelial origin. The remaining 10% of OC are non-epithelial tumors consisting of germ cell, sex cord-stromal, and other rare non-epithelial ovarian tumors (including sarcoma or small cell carcinoma).
Epithelial OC can be stratified into five major histologic subtypes, including high-grade serous carcinoma (HGSC; accounting for up to 70% of all epithelial OC cases), endometrioid (~10%), clear cell (~10%), mucinous (~3%), and low-grade serous carcinoma (LGSC; <5%) [10]. The histopathologic classification of epithelial OC largely determines the clinical course of the disease. From this perspective, epithelial OC is divided into tumors with a good (type I) or poor (type II) prognosis. Type II OC is more prevalent as it comprises the most common HGSC and several less common OC subtypes, including carcinosarcoma and other mixed or undefined epithelial OC. Type II is characterized by frequent abnormalities in p53-related and homologous recombination (HR) DNA repair pathways, resulting in genome instability, and includes high-grade tumors with high proliferative potential and rapid progression, contributing to the late diagnosis at advanced stages [11,12]. In contrast, less common type I ovarian tumors include LGSC, clear cell carcinoma, endometrioid carcinoma, mucinous carcinoma, seromucinous carcinoma, and transitional cell carcinoma. Compared to the known genetic instability of type II tumors, type I tumors are relatively genetically stable. Type I OC are typically low-grade tumors with low proliferative activity and slow progression developing from benign lesions, particularly borderline tumors of the ovary, and, therefore, these tumors are more often diagnosed at an earlier stage [13,14].
Typically, OC develops in late adulthood, with a median age at diagnosis of 63 years [3]. Extremely early-onset ovarian tumors diagnosed between 18 and 30 years of age account for less than 5% of all OC cases (Figure 1) [3,15], and owing to their rarity, only a few studies have been conducted up-to-date. However, all the reports pointed out some striking differences between late- and early-onset adult OC, including genetic background, clinicopathologic features, or clinical outcomes compared with late-onset tumors (Table 1). Notably, while type II OC and particularly HGSC predominate in late-onset OC, approximately 50% of early-onset ovarian tumors are of germ-cell origin (representing a juvenile form of ovarian tumors diagnosed most frequently between the ages of 15 and 20 years), and only approximately 40% of the tumors belong to epithelial OC, particularly LGSC [16]. Correspondingly, late-onset OC is typically diagnosed at advanced stages, frequently with metastatic spread [3,17,18], whereas early-onset OC patients are typically diagnosed with localized disease. This would imply a better prognosis for early-onset OC patients [3,17,19], as also indicated by the significant survival advantage shown in a population-based study of OC patients [17]. However, the age at disease onset has not been confirmed as an independent stratifying factor concerning the prognosis for early-onset OC patients [19,20,21,22]. Particularly, in the long-term follow-up study, Gershenson et al. [22] observed a significantly worsened outcome, including both progression-free survival and overall survival in early-onset (≤35 years) compared with late-onset OC patients. Although LGSC generally have a more favorable prognosis in general [3,16,18], LGSC in early-onset OC patients have a worse prognosis and lower 5-year survival [22], providing another reason for separating this cohort from the majority of late-onset OC.

3. OC Risk Factors and Predisposition

The lifetime OC risk is about 1.1% in developed countries [3] and positively associated with lifetime ovulatory years (except for rare mucinous tumors) [27]. Moreover, the relative risk (RR) of OC development can increase approximately three times in cases of positive family epithelial OC history [28]. This is related to the high proportion of hereditary forms of OC, as >20% of OC patients are carriers of a GPV in an established cancer predisposition gene (Figure 1) [4,5,9]. Nevertheless, the frequency of GPV carriers differs substantially among patients with various histological subtypes. The highest proportion of GPVs are present in epithelial type II OC and, particularly in HGSC, the most prevalent late-onset OC, while the proportion of GPVs decreases in type I OC, including LGSC, and is the lowest in clear cell and mucinous type I carcinomas [29]. The predisposition to non-epithelial ovarian tumors is much less understood.

3.1. Established OC Predisposition Genes

The genetic predisposition to epithelial OC is well established, with major contributions from GPVs in genes coding for HR and mismatch repair (MMR) proteins (Table 2). The BRCA1 and BRCA2 HR genes are the most commonly germline-altered cancer predisposition genes responsible for the development of hereditary breast and ovarian cancer (HBOC) syndrome. GPVs in both genes could be found in nearly 20% of all OC patients [30]. The OC risk for BRCA1 and BRCA2 carriers rises significantly from the age of 35 and 45 years and reaches 58% and 29%, respectively [31,32]. An order of magnitude lower frequency but still significant risk is associated with carriers of GPVs in other HR genes, RAD51D, RAD51C, and BRIP1, having a life-time risk of epithelial OC of 20%, 15%, and 15%, respectively [33,34]. The median age of OC onset in BRCA1 GPV carriers is significantly lower (53 years) compared to BRCA2, RAD51D, RAD51C, and BRIP1 GPV carriers (59, 57, 62, and 65 years, respectively) or the general population; however, tumors before the age of 30 are rare, which is also reflected by the clinical management guidelines [31,33,34,35,36]. An increased risk of epithelial OC has also been documented for carriers of GPVs in MMR genes associated with Lynch syndrome (LS) (Table 2). The life-time risk varies in a range from a few percent (MSH6 and PMS2) up to 20% (MLH1) and 38% (MSH2/EPCAM) [37]. However, while MLH1, MSH2, and MSH6 are strongly associated with OC, the role of PMS2 in OC predisposition is limited. Lynch syndrome OC patients are typically younger, with a median age at diagnosis of 43 and 46 years for MSH2 and MLH1, respectively [38,39], and have also been described in early-onset OC patients [40]. In addition to the high-penetrance epithelial OC predisposition genes, GPVs in other genes involved in double strand break repair, including ATM and PALB2, have been associated with moderate risk to late-onset epithelial OC [41,42,43]. However, the clinical utility of the moderate-penetrance genes is low and largely depends on family OC history.
In addition, other rarely mutated genes are associated with non-epithelial ovarian tumors (Table 2). The GPVs in STK11, causing rare Peutz–Jeghers syndrome, are associated with a non-epithelial ovarian tumor risk exceeding 10% and the development of early-onset tumors in patients <30 years [44,45,46]. Only a few episodical reports document the development of early-onset epithelial OC in carriers of GPVs in STK11 (Table 2) [4,41]. GPVs in DICER1 have been linked to early-onset, non-epithelial sex cord-stromal ovarian neoplasms [47,48] accounting for nearly half of the stromal ovarian tumors [49]. However, DICER1 appears to be exclusively characteristic of non-epithelial, early-onset ovarian tumors. Furthermore, GPVs in SMARCA4 were associated with small cell carcinoma of the ovary hypercalcemic type (SCCOHT), a rare, aggressive OC similar to malignant rhabdoid tumors that primarily affects women under 40 years of age [50]. GPVs in SMARCA4 have been reported recently in two early-onset OC patients; nevertheless, selectively in SCCOHT [51].
Generally, early cancer onset indicates the involvement of hereditary cancer predispositions [52,53]. Thus, one might anticipate an enrichment of GPV carriers in high-penetrance genes in early-onset OC patients. However, this assumption does not apply for early-onset epithelial OC patients, who were found to rarely carry GPVs, with the frequency not exceeding 10% (Figure 1) [4,7,8,9,38]. Nevertheless, only a limited number of studies focusing on the genetic predispositions of early-onset OC have been performed so far (Table 3).
Table 2. Established OC predisposition genes.
Table 2. Established OC predisposition genes.
GeneHeterozygoteHomozygote/
Compound Heterozygote [54]
Associated OC
Histotype
Absolute Risk for OC [35]GPV Identified in Early-Onset OCOther Associated Cancer Types [35]
High penetrance
BRCA1Epithelial [41]39–58%Yes [4,9]BC, PaC, PrCFA-S
BRCA2Epithelial [41]13–29%Yes [4]BC, PaC, PrC, MMFA-D1
BRIP1Epithelial [41]5–15%Yes [4,9]BC, CrC, ECFA-J
DICER1Sex cord-stromal [47]NAYes [47,48]DICER1 sy-
MLH1Epithelial [37,41]4–20%NoLynch sy—CrC, EC, PaCCMMRD
MSH2Epithelial [37]8–38%Yes [40,52]Lynch sy—CrC, EC, PaCCMMRD
RAD51CEpithelial [41]10–15%Yes [4]BCFA-O
RAD51DEpithelial [41]10–20%NoBC-
SMARCA4SCCOHT [50]NAYes [50,51]Rhabdoid tumor predisposition sy-
STK11Non-epithelial [45]>10%Yes [46]Peutz–Jeghers sy, BC, PaC, CrC-
Moderate penetrance/Insufficient evidence
ATMEpithelial [41]2–3%Yes [4,9]PaCAT
MSH6Epithelial [37]1–13%NoLynch sy—CrC, EC, PaCCMMRD
PMS2Epithelial [37]1–3%Yes [40,52]Lynch sy—CrC, ECCMMRD
PALB2Epithelial [42]3–5%NoBC, PaCFA-N
AT, ataxia-telangiectasia; BC, breast cancer; CMMRD, constitutional mismatch repair deficiency syndrome; CrC, colorectal cancer; EC, endometrial cancer; FA, Fanconi anemia; GPV, germline pathogenic/likely pathogenic variant; MM, malignant melanoma; NA, not available; OC, ovarian cancer; PaC, pancreatic cancer; PrC, prostate cancer; SCCOHT, small cell carcinoma of the ovary hypercalcemic type; sy, syndrome.
This controversy was first acknowledged by Stratton et al. in 1999 [8], who tested only a few genetic loci (BRCA1, MLH1, MSH2, and a part of BRCA2) in the largest cohort of early-onset OC patients up to date. Since then, few studies have been conducted, but all of them confirmed a low frequency of GPVs among early-onset OC patients diagnosed <30 years [4,7,9,38,40,51,56,57]. Four recent larger studies, Carter et al. (2018) [9], Lhotova et al. (2020) [4], Flaum et al. (2023) [40], and Herold et al. (2023) [51], identified 2/147 (1.4%), 6/84 (7.1%), 4/77 (5.2%), and 3/83 (3.6%) GPV carriers in established high-penetrance OC predisposition genes (including 0.7%, 3.6%, 0%, and 0% mutations in BRCA1/BRCA2), respectively (Table 3). All four studies used the panel NGS approach, but with a different range of analyzed genes. While Carter [9] and Flaum [40] analyzed 15 and Herold [51] included 25 established cancer predisposition genes in their panel (Supplementary Table S1), Lhotova [4] used a much wider panel, targeting 219 established and candidate cancer predisposition genes. Despite the different design of the analysis, the similar results pointing to a very low frequency of GPV carriers in early-onset OC patients were strikingly similar. Interestingly, Flaum [40] associated GPVs in MSH2 with early-onset OC, as 5.2% (4/77) of the patients carried the same MSH2 GPV. This association was in coherence with the findings of another study focusing on OC in LS patients that included also three early-onset OC patients (Table 3) [52]; however, no other study further supported MSH2-association with early-onset OC. Due to the rarity of early-onset OC, further studies evaluating genetic predisposition to OC have included only a few OC cases with diagnoses at such a young age [52,55,58,59,60], leading to a limited understanding of the genetic factors underlying early-onset OC development. The overall lack of GPV carriers among early-onset OC patients is, however, evident and implies the need for the separation of this cohort from OC. Based on the frequency of GPVs in established OC predisposition genes, the cut-off age for distinguishing between early- and late-onset epithelial OC is around 30, as the frequency of BRCA1 GPVs (a major genetic contributor to OC) starts to increase from age 35 [4].

3.2. Candidate OC Predisposition Genes

Candidate OC predisposition genes can be proposed based on their predisposition to other cancer types or associated diseases, which, however, have not yet been associated with OC (Table 4). The high-penetrance cancer predisposition genes APC, BMPR1A, BAP1, FH, MEN1, PTEN, VHL, WT1, and TP53 (on top of their strong association with gastrointestinal tumors, melanoma, leiomyomatosis, multiple endocrine neoplasia, hamartomas, kidney tumors, breast cancer (BC), central nervous system tumors, and sarcomas, respectively) were also identified in OC patients and/or families [5,62,63,64,65,66,67,68,69,70], suggesting a potential wider cancer manifestation in these syndromes. Moreover, BARD1 and CHEK2 [5,69,70,71,72,73,74,75] were associated with a high to moderate risk of other cancer types, but their contribution to OC risk remains to be confirmed. Nevertheless, GPVs in both of these genes have been reported in several early-onset OC patients [4,9,55,58,59,60].
Furthermore, as the majority of established OC predisposition genes (Table 2) code for proteins involved in DNA repair and/or DNA damage response, the new candidate OC predisposition genes are often sought among genes involved in these pathways. GPVs in genes of the Fanconi anemia complex (including FANCA, FANCC, FANCL, FANCM, and SLX4), MRN complex genes (MRE11-RAD50-NBN), and other genes associated with DNA repair pathways have been reported in OC patients, suggesting their possible role in OC predisposition [5,41,51,55,68,75,76,77,78,79,80,81,82,83,84,85]. Moreover, new potential candidate genes emerged from complex sequencing studies of OC patients without being previously clearly established with any cancer, namely ABRAXAS1 (also known as FAM175A), CNKSR1, and PIK3C2G [56,86,87]. However, it cannot be ruled out that all these more or less isolated findings may be coincidental and do not significantly affect the OC risk. Summarizing, the role of all the above-mentioned, candidate genes in early-onset OC is unclear, calling for further investigation in this field.
Table 4. Candidate OC predisposition genes.
Table 4. Candidate OC predisposition genes.
GeneGPVs Identified in Early-Onset OCAssociated Disease (Inheritance Mode) [54]OC-Association Reported in
ABRAXAS1 (FAM175A)No-[56,86]
ATRNoCutaneous telangiectasia and cancer sy (AD)[78]
APCYes (early 30s) [63]Familial adenomatous polyposis (AD)[62,63]
BAP1NoMelanoma (AD)[68]
BARD1Yes [6,25,55,59,60]BC (AD)[71]
BLMNoBloom sy (AR)[5,76]
BMPR1AYes [64]Juvenile polyposis sy, primary ovarian insufficiency (AD)[64]
BRATYes (in their 30s) [85]Neurodevelopmental disorder (AR)[85]
CNKSR1No-[87]
CDKN2ANoMM, MM-PaC sy (AD)[5,72]
CHEK2Yes [6,25,55,59,60]BC (AD)[5,73]
ERCC3NoTrichothiodystrophy, xeroderma pigmentosum (AR)[88,89]
FANCANoFA (AR)[77,78]
FANCCNoFA (AR)[77]
FANCLNoFA (AR)[77]
FANCMYes [51]-[77,79]
FHNoLeiomyomatosis and renal cell cancer (AD), fumarase deficiency (AR)[68]
MEN1NoMultiple endocrine neoplasia (AD)[65]
MRE11NoAT-like disorder (AR)[81,84]
NBNYes [25]Nijmegen breakage sy (AR)[41]
NF1NoNeurofribromatosis (AD)[5]
PIK3C2GNo-[87]
POLD1NoCrC, EC (AD)[74]
POLENoCrC, EC (AD), IMAGE-I sy (AR)[73]
POLKNo-[75]
PTENYes [66]Cowden sy (AD)[66,67]
RAD50NoNijmegen breakage syndrome-like disorder (AR)[84]
RAD51BNo-[5,82]
RAD52No-[83]
RAD54BNo-[83]
RAD54LNo-[55]
RB1NoRetinoblastoma (AD)[5]
RTEL1NoDyskeratosis congenita (AD/AR), telomere-related pulmonary fibrosis, and/or bone marrow failure sy (AD)[68]
SLX4NoFA (AR)[75,81]
TP53Yes [51,69]Li–Fraumeni sy (AD)[5,69,70]
TSC2NoTuberous sclerosis (AD)[68]
VHLNovon Hippel–Lindau sy, pheochromocytoma (AD)[68]
WT1NoWilms tumor (AD)[68]
XRCC3No-[82]
AD, autosomal dominant; AR, autosomal recessive; AT, ataxia-telangiectasia; BC, breast cancer; CrC, colorectal cancer; FA, Fanconi anemia; IMAGE, intrauterine growth restriction, metaphyseal dysplasia, adrenal hypoplasia congenita, and genitourinary abnormalities; MM, malignant melanoma; PaC, pancreatic cancer; sy, syndrome.

4. Alternative Approaches to Germline Genetic Testing in Early-Onset OC

The lack of identified GPVs in established and candidate cancer predisposition genes in early-onset OC patients could indicate a different disease-causing genetic basis compared to late-onset OC. Applying wider-scope germline whole exome sequencing (WES) or even whole genome sequencing (WGS) may provide new insight into early-onset OC genetics. WES has been used for the genetic analysis of OC (including some early-onset cases) [5,55,84]; however, the results have not shed light on an apparent, clinically relevant, new predisposition gene. Promisingly, WGS could identify yet missed variants (including deep intronic, untranslated regions, and copy number variants) in established and candidate or completely newly associated genes involved in OC predisposition [90,91,92]. Considering complex WES/WGS data analysis and interpretation, complementation with RNA-NGS could help to better understand the cancer-predisposition molecular mechanisms [93]. Nonetheless, these methods are currently not commonly used in clinical practice, as the limitations are not only the substantial cost of the testing and the massive abundance of the NGS data, but most importantly, the unclear clinical significance of the identified variants [94,95].

4.1. Alternative Ways of Cancer Predisposition Inheritance

Apart from applying new testing and analytical methods, various genetic and non-genetic factors (Figure 2) need to be considered in a complex analysis of early-onset OC predisposition. The association of proposed genetic components, as well as other factors predisposing to OC mentioned below has not been established for early-onset OC yet, but there are some pieces of evidence suggesting it could cause or contribute to early-onset cancer or to OC development in general.
In addition to autosomal dominant Mendelian inheritance, recessive inheritance may rarely contribute to the early-onset of OC, as exemplified by isolated cases of biallelic GPV carriers in BRCA1 [96] or PMS2 [52]. These sporadic cases suggest that recessive inheritance represents an uncommon cause of early-onset OC. Similarly, the development of de novo GPVs in BRCA1 has been reported in rare cases of early-onset HBOC patients [97,98]. Also, a de novo GPV in SMARCA4 has been observed in a patient with early-onset OC and other types of childhood/early-onset rhabdoid tumors [99]. The low frequency of de novo mutations among OC patients may have been biased by the HBOC genetic testing criteria, which used to prioritize patients with strongly positive family histories [100]. As early-onset OC patients lack a strong family cancer history [8], the de novo or compound heterozygous OC-predisposition mutations may be under-reported, and their identification, e.g., using a trio WES (analysis of the patient + her parents) similar to other rare childhood/early-onset diseases [101] may help to uncover the underlying genetic causes.
In addition, constitutional mosaicism of GPVs (with lower variant allele frequency in peripheral blood) in established/candidate genes with Mendelian inheritance may be a specific issue. Constitutional mosaicism of GPVs in BRCA1 and BRCA2 [98,102,103] has been previously reported in HBOC patients, suggesting that even the low-level mosaic GPVs in peripheral blood may be significant for the phenotype.
Finally, epigenetic inactivation can contribute to hereditary OC, as shown by promoter methylation of BRCA1 [104], or MLH1 in LS patients recently [105]. However, the prevalence of this phenomenon is largely unknown due to the limited data.

4.2. Family History and X-Linked Inheritance

Despite the low proportion of strong cancer family history in early-onset OC patients compared to patients with OC diagnosed > 30 years, the familiar form of early-onset OC still raises some important questions. Stratton et al. [8] described a slightly elevated OC risk, but also significantly enriched non-Hodgkin lymphoma and myeloma among first-degree relatives of early-onset OC patients (in the majority of cases without known germline genetic predisposition). Similarly, Rantala et al. [106] associated an increased incidence of early-onset (<40 years) OC and testicular cancer in patients whose aunts suffered from early-onset BC. On the other hand, Imbert-Bouteille et al. [107] found no relation between the occurrence of early-onset OC in families with BRCA1/BRCA2 mutations and the age at diagnosis of BC or OC in their relatives. When focusing on the sex of the affected relatives, Stratton et al. [108] noticed that an affected woman’s sisters are at higher risk of disease than their mothers. In sync, Eng et al. described an X-linked association between prostate cancer in men and OC in their mothers and daughters [109]. These somewhat contradictory findings about the importance of family history in early-onset OC predisposition may point in the direction of an X-linked inheritance.
Apart from single nucleotide variants, X-linked OC predisposition can include complex genetic effects like X chromosome inactivation (XCI). Buller et al. [110] described a higher frequency of skewed XCI among OC patients, suggesting X-linked tumor suppressor genes or X-linked low penetrance susceptibility alleles affected by the inactivation pattern [111]. On the other hand, skewed XCI has been described at an increased frequency in BRCA1 mutation carriers compared with controls, and is associated with a statistically significant increase in age at diagnosis of breast and ovarian cancer in BRCA1/BRCA2 GPV carriers [112]. In addition to the effect on the age of onset, molecular signatures of XCI were associated with clinical outcomes in epithelial OC, as patients with dysregulated XCI had shorter progression and overall survival than those with regulated XCI [113], suggesting a complex involvement of the X chromosome in OC.

4.3. Polygenic Inheritance

A complex polygenic inheritance stemming from an additive effect of multiple genetic variants may also be involved in early-onset OC predisposition (Figure 2). Polygenic inheritance could also explain the observed lack of positive family history in close relatives of early-onset OC patients [8]. A polygenic model of inherited predisposition to cancer was proposed by Qing et al. [114], who identified a higher burden of germline variants in protein-coding cancer hallmark genes predicted to alter the structure, expression, or function in early-onset patients compared to late-onset ones. They hypothesized that the early-onset carriers of more germline low-risk variants needed to harbor fewer somatic mutations for malignant transformation. Their hypothesis was supported by a significant association with several cancer types, including the OC.
Currently, hundreds of cancer-risk single nucleotide polymorphisms (SNPs) have been identified by genome-wide association studies, improving their understanding but not fully uncovering their polygenic heritability. These SNPs are considered causal or linked to causal variants. Using a polygenic risk score (PRS), some studies have recently shown a cumulative impact of SNPs in patients with several cancer diagnoses, including OC [115]. Especially serous OC showed association with PRS [116,117], and, interestingly, association was also found between PRS and early diagnosis of BC [118], together suggesting PRS might be associated with early-onset OC. In addition to PRS, another OC risk score evaluating epistatic gene interactions via chromosomal-scale length variation was proposed [119].

4.4. Di/Oligenic Inheritance

Since both monogenic and polygenic inheritance in OC have been demonstrated, the involvement of digenic to oligogenic inheritance might presumably also be a part of the OC genetic predisposition (Figure 2). In general, several di/oligogenic mechanisms have been suggested.
Firstly, a higher incidence and earlier onset of cancer have been proposed in carriers of two or more GPVs in high-penetrance cancer predisposition genes called multilocus inherited neoplasia allele syndrome (MINAS). However, BRCA1/BRCA2-MINAS patients did not develop OC significantly earlier than single GPV carriers [120], further disputing the BRCA1/BRCA2 involvement in early-onset OC predisposition.
Secondly, a combination of a monogenic GPVs and a phenotype-modifying (e.g., age of onset) variant were reported. Carriers of GPVs in established OC-predisposition genes BRCA1/BRCA2, together with the newly associated gene PPARGC1A, were diagnosed at significantly earlier ages, suggesting PPARGC1A is a modifier of OC onset in BRCA1/BRCA2 carriers [121]. Moreover, ethnically-specific modifiers were proposed to influence the phenotype in BRCA1 GPV carriers [122], suggesting missing clues even in BRCA1/BRCA2 otherwise well-established OC predisposition. On the other hand, focusing on BRCA1/BRCA2 negative OC patients, Eng et al. [109] proposed one X-linked SNP in the MAGEC3 gene to advance the age of OC onset by almost seven years. The OC risk or age at onset might also be modified by genetic variants in regulatory elements such as miRNA. Dysregulation of BRCA1/BRCA2 functions in OC was reported in the absence of BRCA1/BRCA2 GPVs stemming from miRNA dysregulation [123], and, especially, miR-146a polymorphism was associated with an earlier age of onset in BRCA1/BRCA2-negative HBOC patients [124].
Thirdly, a truly digenic disorder might be considered. Despite several clearly reported oligogenic disease associations, such as familial hemophagocytic lymphohistiocytosis, primary immunodeficiency, or familial hypercholesterolemia [125,126,127,128], the majority of proposed di/oligogenic allele combinations found in affected patients remain of uncertain significance [129]. Currently, we can only hypothesize about combinations of genetic variants that are separately nonpathogenic (e.g., missense) but together pathogenic when present in mutually interacting domains of proteins involved in OC predisposition, asking for a battery of functional tests and clinical investigation. Despite the currently unavailable clear association with early-onset OC, some evidence about the di/oligogenic inheritance involvement in cancer and, particularly, OC predisposition was proposed. Mouse model-based results suggest that oligogenic inheritance is also a part of cancer predisposition [130], supported by the first few reported clinical cases of digenic colorectal and gastric cancer in the OLIDA (OLIgogenic diseases DAtabase) [129].

4.5. Immune-Related Modifiers of OC

To further expand the complexity of OC predisposition, the immune system is also involved in cancer development (Figure 2). It has already been established that local chronic inflammation and autoimmune disease, including genetic immune dysregulations, predispose to certain types of cancer, such as autoimmune hepatitis-induced cirrhosis to hepatocellular carcinoma [131], Helicobacter pylori infection to gastric cancer [132], or inflammatory bowel disease to colorectal cancer [133]. Therefore, one could speculate that a systemic or ovarian-localized inflammatory condition may also, with time, lead to OC, possibly on an immunogenetic inherited basis. Currently, the knowledge of inherited immunity aspects associated with OC is limited, but a few examples have been described in the literature. Namely, systemic lupus erythematosus was reported to be associated with a higher incidence of cancer, including ovarian cancer [134]. Moreover, a significantly increased incidence of specific HLA-class II haplotypes (namely, DRB1*0301-DQA1*0501-DQB1*0201 and DRB1*1001-DQA1*0101-DQB1*0501) has been observed in OC patients, suggesting their role in OC pathogenesis [135].

4.6. Non-Genetic Factors

Similarly, genotype-environment interactions are a great unknown in the field of OC predisposition. The fact that the interaction of exposure to environmental carcinogens and constitutional genetics modulates cancer risk was first suggested in autosomal recessive Bloom and Werner syndromes caused by biallelic mutations in BLM and WRN, respectively [136]. Since then, several genotype-environment interactions have been recognized, including the combined presence of GPVs in BAP1 and exposure to asbestos fibers, which together increase the risk of disease more than either component alone [137]. Moreover, the genotype-environment interaction may possibly modulate phenotypic heterogeneity, as shown in Birt–Hogg–Dubé syndrome caused by GPVs in FLCN or suggested for DICER1 [138,139], which were also both reported in families with OC cases [47,48].
Moreover, lifestyle can also modify pre-existing OC predisposition. Namely, alcohol intake elevates the risk for OC [140], as well as exposure to smoking in childhood. Interestingly, smoking exposure was more likely associated with LGSC and non-serous OC [141], which are more frequent OC types among early-onset OC patients [16,17]. On the other hand, smoking has also been identified as a protective factor in endometroid and clear-cell OC [142]; however, smokers among early-onset OC patients have a short smoking history, limiting the effect of this risk factor.
Finally, we can also raise the question of whether survival differs in extremely young OC patients compared to histology-matched late-onset patients, as shown in early-onset LGSC OC patients with a less favorable prognosis and decreased 5-year survival [22]. Isolated studies point to some predictive markers associated with better survival in ovarian cancer patients, such as PRDM1 variant rs2185379, which is suggested to be positively associated with the long-term recurrence-free survival of advanced OC [143]. On the other hand, the EXO1 variant rs851797 was negatively associated with progression-free and overall survival in OC patients [144]. However, these isolated association studies typically deal with HGSC with late-onset and focus on patients from a specific population and should be interpreted with caution.

5. Conclusions

Despite being rare, early-onset OC represents a significant healthcare and socioeconomic problem, and its development is currently poorly understood. The risk of the disease can be modulated not only by genetics but also by environmental/lifestyle factors and/or their interactions. Supported by the rarity of the disease, it cannot be ruled out that early-onset OC (<30 years) is an unpredictable stochastic event caused just due to chance by random variables of different values.
Knowledge of the genetic factors could help identify women at risk of early-onset OC and might also be of predictive and prognostic value. Nevertheless, this is currently impossible, as we do not fully understand the factors associated with OC risk at such a young age. Since early-onset OC shows a significantly distinct germline mutation status compared to late-onset OC and the number of studies analyzing the GPVs in early-onset OC is limited, it is necessary to investigate early-onset OC patients in detail and separately from late-onset OC. Finding new approaches to germline genetic testing of early-onset OC patients is crucial for the identification of the expected yet unknown genetic causes, if present. These new insights could not only help to better understand the specifics of early-onset OC and provide new potential diagnostic and preventive targets for patient care and counseling, but they could also help to tailor the treatment modalities, thus improving the quality of life and survival of the early-onset OC patients.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/1422-0067/24/23/17020/s1.

Author Contributions

K.H.: writing—original draft. M.J.: funding acquisition, writing—review and editing. P.K.: funding acquisition, writing—review and editing. Z.K.: funding acquisition, writing—review and editing. J.S.: Funding acquisition, writing—original draft. All authors have read and agreed to the published version of the manuscript.

Funding

This work has been supported by the Ministry of Health of the Czech Republic: NU20-03-00016, NU20-09-00355, NU23-03-00150, RVO-VFN 00064165; Charles University: COOPERATIO, SVV260516; and the Ministry of Education Youth and Sports of the Czech Republic: Programme EXCELES, ID Project No. LX22NPO5102—Funded by the European Union—Next Generation EU.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Conflicts of Interest

No potential conflicts of interest were reported. We declare that the results summarized in this manuscript have not been published previously and have not been submitted for consideration to any other journal.

Correction Statement

This article has been republished with a minor correction to the Funding statement. This change does not affect the scientific content of the article.

Abbreviations

ADAutosomal Dominant
ARAutosomal Recessive
ATAtaxia-Telangiectasia
BCBreast Cancer
CMMRDConstitutional mismatch repair deficiency syndrome
CrCColorectal Cancer
DgDiagnosis
ECEndometrial Cancer
FAFanconi Anemia
GPVGermline Pathogenic/Likely Pathogenic Variant
HBOCHereditary Breast and Ovarian Cancer
HGSCHigh-Grade Serous Carcinoma
HRHomologous Recombination
IMAGEIntrauterine growth restriction, Metaphyseal dysplasia, Adrenal hypoplasia congenita, and Genitourinary abnormalities
LGSCLow-Grade Serous Carcinoma
LSLynch Syndrome
MINASMultilocus Inherited Neoplasia Allele Syndrome
MMMalignant Melanoma
MMRMismatch Repair
NANot Available
NGSNext-Generation Sequencing
No.Number
OCOvarian cancer
OLIDAOligogenic Diseases Database
PaCPancreatic Cancer
PrCProstate Cancer
PRSPolygenic Risk Score
SCCOHTSmall Cell Carcinoma of the Ovary Hypercalcemic Type
SNPSingle Nucleotide Polymorphism
SySyndrome
WESWhole Exome Sequencing
WGSWhole Genome Sequencing
XCIX Chromosome Inactivation
YoYears Old

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F.; Bsc, M.F.B.; Me, J.F.; Soerjomataram, M.I.; et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Kim, M.-K.; Kim, K.; Kim, S.M.; Kim, J.W.; Park, N.-H.; Song, Y.-S.; Kang, S.-B. A hospital-based case-control study of identifying ovarian cancer using symptom index. J. Gynecol. Oncol. 2009, 20, 238–242. [Google Scholar] [CrossRef] [PubMed]
  3. National Cancer Institute: Surveillance, Epidemiology, and End Results Program. Available online: www.seer.cancer.gov/ (accessed on 1 September 2023).
  4. Ray-Coquard, I.; Morice, P.; Lorusso, D.; Prat, J.; Oaknin, A.; Pautier, P.; Colombo, N. Non-epithelial ovarian cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2018, 29 (Suppl. S4), iv1–iv18. [Google Scholar] [CrossRef] [PubMed]
  5. Lhotova, K.; Stolarova, L.; Zemankova, P.; Vocka, M.; Janatova, M.; Borecka, M.; Cerna, M.; Jelinkova, S.; Kral, J.; Volkova, Z.; et al. Multigene Panel Germline Testing of 1333 Czech Patients with Ovarian Cancer. Cancers 2020, 12, 956. [Google Scholar] [CrossRef] [PubMed]
  6. Kanchi, K.L.; Johnson, K.J.; Lu, C.; McLellan, M.D.; Leiserson, M.D.M.; Wendl, M.C.; Zhang, Q.; Koboldt, D.C.; Xie, M.; Kandoth, C.; et al. Integrated analysis of germline and somatic variants in ovarian cancer. Nat. Commun. 2014, 5, 3156. [Google Scholar] [CrossRef] [PubMed]
  7. Risch, H.A.; McLaughlin, J.R.; Cole, D.E.; Rosen, B.; Bradley, L.; Kwan, E.; Jack, E.; Vesprini, D.J.; Kuperstein, G.; Abrahamson, J.L.; et al. Prevalence and penetrance of germline BRCA1 and BRCA2 mutations in a population series of 649 women with ovarian cancer. Am. J. Hum. Genet. 2001, 68, 700–710. [Google Scholar] [CrossRef] [PubMed]
  8. Stratton, J.F.; Thompson, D.; Bobrow, L.; Dalal, N.; Gore, M.; Bishop, D.; Scott, I.; Evans, G.; Daly, P.; Easton, D.F.; et al. The genetic epidemiology of early-onset epithelial ovarian cancer: A population-based study. Am. J. Hum. Genet. 1999, 65, 1725–1732. [Google Scholar] [CrossRef]
  9. Carter, N.J.; Marshall, M.L.; Susswein, L.R.; Zorn, K.K.; Hiraki, S.; Arvai, K.J.; Torene, R.I.; McGill, A.K.; Yackowski, L.; Murphy, P.D.; et al. Germline pathogenic variants identified in women with ovarian tumors. Gynecol. Oncol. 2018, 151, 481–488. [Google Scholar] [CrossRef]
  10. Prat, J. New insights into ovarian cancer pathology. Ann. Oncol. 2012, 23 (Suppl. S10), x111–x117. [Google Scholar] [CrossRef]
  11. Cancer Genome Atlas Research Network. Integrated genomic analyses of ovarian carcinoma. Nature 2011, 474, 609–615. [Google Scholar] [CrossRef]
  12. Morden, C.R.; Farrell, A.C.; Sliwowski, M.; Lichtensztejn, Z.; Altman, A.D.; Nachtigal, M.W.; McManus, K.J. Chromosome instability is prevalent and dynamic in high-grade serous ovarian cancer patient samples. Gynecol. Oncol. 2021, 161, 769–778. [Google Scholar] [CrossRef] [PubMed]
  13. Matz, M.; Coleman, M.P.; Sant, M.; Chirlaque, M.D.; Visser, O.; Gore, M.; Allemani, C.; Bouzbid, S.; Hamdi-Chérif, M.; Zaidi, Z.; et al. The histology of ovarian cancer: Worldwide distribution and implications for international survival comparisons (CONCORD-2). Gynecol. Oncol. 2017, 144, 405–413. [Google Scholar] [CrossRef] [PubMed]
  14. Kurman, R.J.; Shih, I.M. The Dualistic Model of Ovarian Carcinogenesis: Revisited, Revised, and Expanded. Am. J. Pathol. 2016, 186, 733–747. [Google Scholar] [CrossRef] [PubMed]
  15. GLOBOCAN 2020. Available online: https://gco.iarc.fr/ (accessed on 1 September 2023).
  16. Lockley, M.; Stoneham, S.J.; Olson, T.A. Ovarian cancer in adolescents and young adults. Pediatr. Blood Cancer 2019, 66, e27512. [Google Scholar] [CrossRef]
  17. Chan, J.K.; Urban, R.; Cheung, M.K.; Osann, K.; Husain, A.; Teng, N.N.; Kapp, D.S.; Berek, J.S.; Leiserowitz, G.S. Ovarian cancer in younger vs older women: A population-based analysis. Br. J. Cancer. 2006, 95, 1314–1320. [Google Scholar] [CrossRef] [PubMed]
  18. Grimley, P.M.; Matsuno, R.K.; Rosenberg, P.S.; Henson, D.E.; Schwartz, A.M.; Anderson, W.F. Qualitative Age Interactions between Low-grade and High-grade Serous Ovarian Carcinomas. Cancer Epidemiol. Biomark. Prev. 2009, 18, 2256–2261. [Google Scholar] [CrossRef] [PubMed]
  19. Massi, D.; Susini, T.; Savino, L.; Boddi, V.; Amunni, G.; Colafranceschi, M. Epithelial ovarian tumors in the reproductive age group: Age is not an independent prognostic factor. Cancer Interdiscip. Int. J. Am. Cancer Soc. 1996, 77, 1131–1136. [Google Scholar] [CrossRef]
  20. Yoshikawa, K.; Fukuda, T.; Uemura, R.; Matsubara, H.; Wada, T.; Kawanishi, M.; Tasaka, R.; Kasai, M.; Hashiguchi, Y.; Ichimura, T.; et al. Age-related differences in prognosis and prognostic factors among patients with epithelial ovarian cancer. Mol. Clin. Oncol. 2018, 9, 329–334. [Google Scholar] [CrossRef]
  21. Schildkraut, J.M.; Halabi, S.; Bastos, E.; Marchbanks, P.A.; McDonald, J.A.; Berchuck, A. Prognostic factors in early-onset epithelial ovarian cancer: A population-based study. Obstet. Gynecol. 2000, 95, 119–127. [Google Scholar] [CrossRef]
  22. Gershenson, D.M.; Bodurka, D.C.; Lu, K.H.; Nathan, L.C.; Milojevic, L.; Wong, K.K.; Malpica, A.; Sun, C.C. Impact of Age and Primary Disease Site on Outcome in Women With Low-Grade Serous Carcinoma of the Ovary or Peritoneum: Results of a Large Single-Institution Registry of a Rare Tumor. J. Clin. Oncol. 2015, 33, 2675–2682. [Google Scholar] [CrossRef]
  23. Lalrinpuii, E.; Bhageerathy, P.S.; Sebastian, A.; Jeyaseelan, L.; Thomas, V.; Thomas, A.; Chandy, R.; Peedicayil, A. Ovarian Cancer in Young Women. Indian J. Surg. Oncol. 2017, 8, 540–547. [Google Scholar] [CrossRef]
  24. Zhang, J.; Ugnat, A.-M.; Clarke, K.; Mao, Y. Ovarian cancer histology-specific incidence trends in Canada 1969–1993: Age-period-cohort analyses. Br. J. Cancer 1999, 81, 152–158. [Google Scholar] [CrossRef] [PubMed]
  25. Schneider, D.T.; Terenziani, M.; Cecchetto, G.; Olson, T.A.; Schneider, D.T.; Terenziani, M.; Olson, T.A.; Schneider, D.T.; Cecchetto, G.; Olson, T.A.; et al. Gonadal and Extragonadal Germ Cell Tumors, Sex Cord Stromal and Rare Gonadal Tumors. In Rare Tumors in Children and Adolescents; Springer International Publishing: Cham, Switzerland, 2012; pp. 327–402. [Google Scholar]
  26. Huang, Y.; Ming, X.; Li, B.; Li, Z. Histological Characteristics and Early-Stage Diagnosis Are Associated With Better Survival in Young Patients With Epithelial Ovarian Cancer: A Retrospective Analysis Based on Surveillance Epidemiology and End Results Database. Front. Oncol. 2020, 10, 595789. [Google Scholar] [CrossRef]
  27. Fu, Z.; Brooks, M.M.; Irvin, S.; Jordan, S.; Aben, K.K.H.; Anton-Culver, H.; Bandera, E.V.; Beckmann, M.W.; Berchuck, A.; Brooks-Wilson, A.; et al. Lifetime ovulatory years and risk of epithelial ovarian cancer: A multinational pooled analysis. JNCI J. Natl. Cancer Inst. 2023, 115, 539–551. [Google Scholar] [CrossRef] [PubMed]
  28. Pavanello, M.; Chan, I.H.; Ariff, A.; Pharoah, P.D.; Gayther, S.A.; Ramus, S.J. Rare Germline Genetic Variants and the Risks of Epithelial Ovarian Cancer. Cancers 2020, 12, 3046. [Google Scholar] [CrossRef] [PubMed]
  29. Witjes, V.M.; van Bommel, M.H.; Ligtenberg, M.J.; Vos, J.R.; Mourits, M.J.; Ausems, M.G.; de Hullu, J.A.; Bosse, T.; Hoogerbrugge, N. Probability of detecting germline BRCA1/2 pathogenic variants in histological subtypes of ovarian carcinoma. A meta-analysis. Gynecol. Oncol. 2022, 164, 221–230. [Google Scholar] [CrossRef]
  30. Toss, A.; Tomasello, C.; Razzaboni, E.; Contu, G.; Grandi, G.; Cagnacci, A.; Schilder, R.J.; Cortesi, L. Hereditary ovarian cancer: Not only BRCA 1 and 2 genes. BioMed Res. Int. 2015, 2015, 341723. [Google Scholar] [CrossRef]
  31. Chen, J.; Bae, E.; Zhang, L.; Hughes, K.; Parmigiani, G.; Braun, D.; Rebbeck, T.R. Penetrance of Breast and Ovarian Cancer in Women Who Carry a BRCA1/2 Mutation and Do Not Use Risk-Reducing Salpingo-Oophorectomy: An Updated Meta-Analysis. JNCI Cancer Spectr. 2020, 4, pkaa029. [Google Scholar] [CrossRef]
  32. Kuchenbaecker, K.B.; Hopper, J.L.; Barnes, D.R.; Phillips, K.A.; Mooij, T.M.; Roos-Blom, M.J.; Jervis, S.; Van Leeuwen, F.E.; Milne, R.L.; Andrieu, N.; et al. Risks of Breast, Ovarian, and Contralateral Breast Cancer for BRCA1 and BRCA2 Mutation Carriers. JAMA 2017, 317, 2402–2416. [Google Scholar] [CrossRef] [PubMed]
  33. Weber-Lassalle, N.; Hauke, J.; Ramser, J.; Richters, L.; Groß, E.; Blümcke, B.; Gehrig, A.; Kahlert, A.-K.; Müller, C.R.; Hackmann, K.; et al. BRIP1 loss-of-function mutations confer high risk for familial ovarian cancer, but not familial breast cancer. Breast Cancer Res. 2018, 20, 7. [Google Scholar] [CrossRef]
  34. Lilyquist, J.; LaDuca, H.; Polley, E.; Davis, B.T.; Shimelis, H.; Hu, C.; Hart, S.N.; Dolinsky, J.S.; Couch, F.J.; Goldgar, D.E. Frequency of mutations in a large series of clinically ascertained ovarian cancer cases tested on multi-gene panels compared to reference controls. Gynecol. Oncol. 2017, 147, 375–380. [Google Scholar] [CrossRef]
  35. National Comprehensive Cancer Network. Genetic/Familial High-Risk Assessment: Breast, Ovarian, and Pancreatic (Version 1.2023). 2022. Available online: https://www.nccn.org/professionals/physician_gls/pdf/genetics_bop.pdf (accessed on 1 September 2023).
  36. Cummings, S.; Roman, S.S.; Saam, J.; Bernhisel, R.; Brown, K.; Lancaster, J.M.; Usha, L. Age of ovarian cancer diagnosis among BRIP1, RAD51C, and RAD51D mutation carriers identified through multi-gene panel testing. J. Ovarian Res. 2021, 14, 61. [Google Scholar] [CrossRef] [PubMed]
  37. Dominguez-Valentin, M.; Sampson, J.R.; Seppälä, T.T.; ten Broeke, S.W.; Plazzer, J.-P.; Nakken, S.; Engel, C.; Aretz, S.; Jenkins, M.A.; Sunde, L.; et al. Cancer risks by gene, age, and gender in 6350 carriers of pathogenic mismatch repair variants: Findings from the Prospective Lynch Syndrome Database. Genet. Med. 2020, 22, 15–25. [Google Scholar] [CrossRef] [PubMed]
  38. Bernards, S.S.; Norquist, B.M.; Harrell, M.I.; Agnew, K.J.; Lee, M.K.; Walsh, T.; Swisher, E.M. Genetic characterization of early onset ovarian carcinoma. Gynecol. Oncol. 2016, 140, 221–225. [Google Scholar] [CrossRef] [PubMed]
  39. Curtius, K.; Gupta, S.; Boland, C.R. Review article: Lynch Syndrome-a mechanistic and clinical management update. Aliment. Pharmacol. Ther. 2022, 55, 960–977. [Google Scholar] [CrossRef]
  40. Flaum, N.; Crosbie, E.J.; Woodward, E.R.; Lalloo, F.; Morgan, R.; Ryan, N.; Evans, D.G. MSH2 is the very young onset ovarian cancer predisposition gene, not BRCA1. J. Med. Genet. 2023, 60, 576–577. [Google Scholar] [CrossRef] [PubMed]
  41. Kurian, A.W.; Hughes, E.; Handorf, E.A.; Gutin, A.; Allen, B.; Hartman, A.-R.; Hall, M.J. Breast and Ovarian Cancer Penetrance Estimates Derived From Germline Multiple-Gene Sequencing Results in Women. JCO Precis. Oncol. 2017, 1, 1–12. [Google Scholar] [CrossRef]
  42. Yang, X.; Leslie, G.; Doroszuk, A.; Schneider, S.; Allen, J.; Decker, B.; Dunning, A.M.; Redman, J.; Scarth, J.; Plaskocinska, I.; et al. Cancer Risks Associated With Germline PALB2 Pathogenic Variants: An International Study of 524 Families. J. Clin. Oncol. 2020, 38, 674–685. [Google Scholar] [CrossRef]
  43. Narayan, P.; Ahsan, M.D.; Webster, E.M.; Perez, L.; Levi, S.R.; Harvey, B.; Wolfe, I.; Beaumont, S.; Brewer, J.T.; Siegel, D.; et al. Partner and localizer of BRCA2 (PALB2) pathogenic variants and ovarian cancer: A systematic review and meta-analysis. Gynecol. Oncol. 2023, 177, 72–85. [Google Scholar] [CrossRef]
  44. Hearle, N.; Schumacher, V.; Menko, F.H.; Olschwang, S.; Boardman, L.A.; Gille, J.J.; Keller, J.J.; Westerman, A.M.; Scott, R.J.; Lim, W.; et al. Frequency and spectrum of cancers in the Peutz-Jeghers syndrome. Clin. Cancer Res. 2006, 12, 3209–3215. [Google Scholar] [CrossRef]
  45. Klimkowski, S.; Ibrahim, M.; Ibarra Rovira, J.J.; Elshikh, M.; Javadi, S.; Klekers, A.R.; Abusaif, A.A.; Moawad, A.W.; Ali, K.; Elsayes, K.M. Peutz-Jeghers Syndrome and the Role of Imaging: Pathophysiology, Diagnosis, and Associated Cancers. Cancers 2021, 13, 5121. [Google Scholar] [CrossRef] [PubMed]
  46. Giardiello, F.M.; Brensinger, J.D.; Tersmette, A.C.; Goodman, S.N.; Petersen, G.M.; Booker, S.V.; Cruz–Correa, M.; Offerhaus, J.A. Very high risk of cancer in familial Peutz-Jeghers syndrome. Gastroenterology 2000, 119, 1447–1453. [Google Scholar] [CrossRef] [PubMed]
  47. De Paolis, E.; Paragliola, R.M.; Concolino, P. Spectrum of DICER1 Germline Pathogenic Variants in Ovarian Sertoli-Leydig Cell Tumor. J. Clin. Med. 2021, 10, 1845. [Google Scholar] [CrossRef]
  48. Frio, T.R.; Bahubeshi, A.; Kanellopoulou, C.; Hamel, N.; Niedziela, M.; Sabbaghian, N.; Pouchet, C.; Gilbert, L.; O’Brien, P.K.; Serfas, K.; et al. DICER1 mutations in familial multinodular goiter with and without ovarian Sertoli-Leydig cell tumors. JAMA 2011, 305, 68–77. [Google Scholar] [CrossRef] [PubMed]
  49. Schultz, K.A.P.; Harris, A.; Doros, L.A.; Young, R.H.; Dehner, L.P.; Frazier, A.L.; Hill, D.A.; Messinger, Y.H. Clinical and genetic aspects of ovarian stromal tumors: A report from the International Ovarian and Testicular Stromal Tumor Registry. J. Clin. Oncol. 2014, 32 (Suppl. S15), 5520. [Google Scholar] [CrossRef]
  50. Witkowski, L.; Carrot-Zhang, J.; Albrecht, S.; Fahiminiya, S.; Hamel, N.; Tomiak, E.; Grynspan, D.; Saloustros, E.; Nadaf, J.; Rivera, B.; et al. Germline and somatic SMARCA4 mutations characterize small cell carcinoma of the ovary, hypercalcemic type. Nat. Genet. 2014, 46, 438–443. [Google Scholar] [CrossRef] [PubMed]
  51. Herold, N.; Schmolling, J.; Ernst, C.; Ataseven, B.; Blümcke, B.; Schömig-Markiefka, B.; Heikaus, S.; Göhring, U.; Engel, C.; Lampe, B.; et al. Pathogenic germline variants in SMARCA4 and further cancer predisposition genes in early onset ovarian cancer. Cancer Med. 2023, 12, 15256–15260. [Google Scholar] [CrossRef]
  52. Ryan, N.; Evans, D.; Green, K.; Crosbie, E. Pathological features and clinical behavior of Lynch syndrome-associated ovarian cancer. Gynecol. Oncol. 2017, 144, 491–495. [Google Scholar] [CrossRef]
  53. Huang, K.L.; Mashl, R.J.; Wu, Y.; Ritter, D.I.; Wang, J.; Oh, C.; Paczkowska, M.; Reynolds, S.; Wyczalkowski, M.A.; Oak, N.; et al. Pathogenic Germline Variants in 10,389 Adult Cancers. Cell 2018, 173, 355–370.e14. [Google Scholar] [CrossRef]
  54. McKusick-Nathans Institute of Genetic Medicine Johns Hopkins University; Baltimore, M. Online Mendelian Inheritance in Man, OMIM®. 2022. Available online: https://omim.org/ (accessed on 1 September 2023).
  55. Felicio, P.S.; Grasel, R.S.; Campacci, N.; de Paula, A.E.; Galvão, H.C.; Torrezan, G.T.; Sabato, C.S.; Fernandes, G.C.; Souza, C.P.; Michelli, R.D.; et al. Whole-exome sequencing of non-BRCA1/BRCA2 mutation carrier cases at high-risk for hereditary breast/ovarian cancer. Hum. Mutat. 2021, 42, 290–299. [Google Scholar] [CrossRef]
  56. da Costa e Silva Carvalho, S.; Cury, N.M.; Brotto, D.B.; De Araujo, L.F.; Rosa, R.C.A.; Texeira, L.A.; Plaça, J.R.; Marques, A.A.; Peronni, K.C.; Ruy, P.D.C.; et al. Germline variants in DNA repair genes associated with hereditary breast and ovarian cancer syndrome: Analysis of a 21 gene panel in the Brazilian population. BMC Med. Genom. 2020, 13, 21. [Google Scholar] [CrossRef] [PubMed]
  57. Boyd, J.; Sonoda, Y.; Federici, M.G.; Bogomolniy, F.; Rhei, E.; Maresco, D.L.; Saigo, P.E.; Almadrones, L.A.; Barakat, R.R.; Brown, C.L.; et al. Clinicopathologic features of BRCA-linked and sporadic ovarian cancer. JAMA 2000, 283, 2260–2265. [Google Scholar] [CrossRef] [PubMed]
  58. Cibula, D.; Laco, J.; Dundr, P.; Hájková, N.; Tichá, I.; Hojný, J.; Němejcová, K.; Bártů, M.; Michálková, R.; Zikán, M.; et al. Synchronous endometrioid endometrial and ovarian carcinomas are biologically related: A clinico-pathological and molecular (next generation sequencing) study of 22 cases. Oncol. Lett. 2019, 17, 2207–2214. [Google Scholar]
  59. Jarhelle, E.; Stensland, H.M.F.R.; Hansen, G.M.; Skarsfjord, S.; Jonsrud, C.; Ingebrigtsen, M.; Strømsvik, N.; Van Ghelue, M. Identifying sequence variants contributing to hereditary breast and ovarian cancer in BRCA1 and BRCA2 negative breast and ovarian cancer patients. Sci. Rep. 2019, 9, 19986. [Google Scholar] [CrossRef] [PubMed]
  60. Koczkowska, M.; Krawczynska, N.; Stukan, M.; Kuzniacka, A.; Brozek, I.; Sniadecki, M.; Debniak, J.; Wydra, D.; Biernat, W.; Kozlowski, P.; et al. Spectrum and Prevalence of Pathogenic Variants in Ovarian Cancer Susceptibility Genes in a Group of 333 Patients. Cancers 2018, 10, 442. [Google Scholar] [CrossRef] [PubMed]
  61. Pal, T.; Permuth-Wey, J.; Betts, J.A.; Krischer, J.P.; Fiorica, J.; Arango, H.; LaPolla, J.; Hoffman, M.; Martino, M.A.; Wakeley, K.; et al. BRCA1 and BRCA2 mutations account for a large proportion of ovarian carcinoma cases. Cancer 2005, 104, 2807–2816. [Google Scholar] [CrossRef] [PubMed]
  62. Anand, L.; Padmavathi, V.; Dhivya, V.; Mahalaxmi, I.; Balachandar, V. De novo germ-line mutation of APC gene in periampullary carcinoma with familial adenomatous polyps—A novel familial case report in South India. Karbala Int. J. Mod. Sci. 2016, 2, 266–270. [Google Scholar] [CrossRef]
  63. Vibert, R.; Le Gall, J.; Buecher, B.; Mouret-Fourme, E.; Bataillon, G.; Becette, V.; Trabelsi-Grati, O.; Moncoutier, V.; Dehainault, C.; Carriere, J.; et al. APC germline pathogenic variants and epithelial ovarian cancer: Causal or coincidental findings? J. Med. Genet. 2023, 60, 460–463. [Google Scholar] [CrossRef]
  64. Babovic, N.; Simmons, P.S.; Moir, C.; Thorland, E.C.; Scheithauer, B.; Gliem, T.J.; Babovic-Vuksanovic, D. Mucinous cystadenoma of ovary in a patient with juvenile polyposis due to 10q23 microdeletion: Expansion of phenotype. Am. J. Med. Genet. A 2010, 152A, 2623–2627. [Google Scholar] [CrossRef]
  65. Lou, L.; Zhou, L.; Wang, W.; Li, H.; Li, Y. Atypical ovarian carcinoid tumor with widespread skeletal metastases: A case report of multiple endocrine neoplasia type 1 in a young woman. BMC Cancer 2019, 19, 1107. [Google Scholar] [CrossRef]
  66. Yauy, K.; Imbert-Bouteille, M.; Bubien, V.; Lindet-Bourgeois, C.; Rathat, G.; Perrochia, H.; MacGrogan, G.; Longy, M.; Bessis, D.; Tinat, J.; et al. Ovarian Clear Cell Carcinoma in Cowden Syndrome. J. Natl. Compr. Cancer Netw. 2019, 17, 7–11. [Google Scholar] [CrossRef]
  67. Cho, M.-Y.; Kim, H.S.; Eng, C.; Kim, D.S.; Kang, S.J.; Eom, M.; Yi, S.Y.; Bronner, M.P. First report of ovarian dysgerminoma in Cowden syndrome with germline PTEN mutation and PTEN-related 10q loss of tumor heterozygosity. Am. J. Surg. Pathol. 2008, 32, 1258–1264. [Google Scholar] [CrossRef] [PubMed]
  68. Sia, T.Y.; Maio, A.; Kemel, Y.M.; Arora, K.S.; Gordhandas, S.B.; Kahn, R.M.; Salo-Mullen, E.E.; Sheehan, M.A.; Tejada, P.R.; Bandlamudi, C.; et al. Germline Pathogenic Variants and Genetic Counseling by Ancestry in Patients With Epithelial Ovarian Cancer. JCO Precis. Oncol. 2023, 7, e2300137. [Google Scholar] [CrossRef] [PubMed]
  69. Janavičius, R.; Andrėkutė, K.; Mickys, U.; Rudaitis, V.; Brasiūnienė, B.; Griškevičius, L. Apparently “BRCA-related” breast and ovarian cancer patient with germline TP53 mutation. Breast J. 2011, 17, 409–415. [Google Scholar] [CrossRef] [PubMed]
  70. Blanco, A.; Grana, B.; Fachal, L.; Santamarina, M.; Cameselle-Teijeiro, J.; Ruíz-Ponte, C.; Carracedo, A.; Vega, A. Beyond BRCA1 and BRCA2 wild-type breast and/or ovarian cancer families: Germline mutations in TP53 and PTEN. Clin. Genet. 2010, 77, 193–196. [Google Scholar] [CrossRef] [PubMed]
  71. Norquist, B.M.; Harrell, M.I.; Brady, M.F.; Walsh, T.; Lee, M.K.; Gulsuner, S.; Bernards, S.S.; Casadei, S.; Yi, Q.; Burger, R.A.; et al. Inherited Mutations in Women With Ovarian Carcinoma. JAMA Oncol. 2016, 2, 482–490. [Google Scholar] [CrossRef] [PubMed]
  72. Dace, P.; Olita, H.; Ludmila, E.; Ingrida, D. Tumour suppressor gene CDKN2A/p16 germline mutations in melanoma patients with additional cancer and cancer in their family history. Acta Univ. Latv. 2003, 662, 25–32. [Google Scholar]
  73. Eoh, K.J.; Kim, J.E.; Park, H.S.; Lee, S.T.; Park, J.S.; Han, J.W.; Lee, J.Y.; Kim, S.; Kim, S.W.; Kim, J.H.; et al. Detection of Germline Mutations in Patients with Epithelial Ovarian Cancer Using Multi-gene Panels: Beyond BRCA1/2. Cancer Res. Treat. 2018, 50, 917–925. [Google Scholar] [CrossRef]
  74. Mur, P.; García-Mulero, S.; del Valle, J.; Magraner-Pardo, L.; Vidal, A.; Pineda, M.; Cinnirella, G.; Martín-Ramos, E.; Pons, T.; López-Doriga, A.; et al. Role of POLE and POLD1 in familial cancer. Genet. Med. 2020, 22, 2089–2100. [Google Scholar] [CrossRef]
  75. Song, H.; Dicks, E.M.; Tyrer, J.; Intermaggio, M.; Chenevix-Trench, G.; Bowtell, D.D.; Traficante, N.; AOCS Group; Brenton, J.; Goranova, T.; et al. Population-based targeted sequencing of 54 candidate genes identifies PALB2 as a susceptibility gene for high-grade serous ovarian cancer. J. Med Genet. 2020, 58, 305–313. [Google Scholar] [CrossRef]
  76. Schubert, S.; van Luttikhuizen, J.L.; Auber, B.; Schmidt, G.; Hofmann, W.; Penkert, J.; Davenport, C.F.; Hille-Betz, U.; Wendeburg, L.; Bublitz, J.; et al. The identification of pathogenic variants in BRCA1/2 negative, high risk, hereditary breast and/or ovarian cancer patients: High frequency of FANCM pathogenic variants. Int. J. Cancer. 2019, 144, 2683–2694. [Google Scholar] [CrossRef]
  77. del Valle, J.; Rofes, P.; Moreno-Cabrera, J.M.; López-Dóriga, A.; Belhadj, S.; Vargas-Parra, G.; Teulé, À.; Cuesta, R.; Muñoz, X.; Campos, O.; et al. Exploring the Role of Mutations in Fanconi Anemia Genes in Hereditary Cancer Patients. Cancers 2020, 12, 829. [Google Scholar] [CrossRef] [PubMed]
  78. Bertelsen, B.; Tuxen, I.V.; Yde, C.W.; Gabrielaite, M.; Torp, M.H.; Kinalis, S.; Oestrup, O.; Rohrberg, K.; Spangaard, I.; Santoni-Rugiu, E.; et al. High frequency of pathogenic germline variants within homologous recombination repair in patients with advanced cancer. NPJ Genom. Med. 2019, 4, 13. [Google Scholar] [CrossRef] [PubMed]
  79. Cavaillé, M.; Uhrhammer, N.; Privat, M.; Ponelle-Chachuat, F.; Gay-Bellile, M.; Lepage, M.; Molnar, I.; Viala, S.; Bidet, Y.; Bignon, Y. Analysis of 11 candidate genes in 849 adult patients with suspected hereditary cancer predisposition. Genes Chromosomes Cancer 2021, 60, 73–78. [Google Scholar] [CrossRef] [PubMed]
  80. Fostira, F.; Kostantopoulou, I.; Apostolou, P.; Papamentzelopoulou, M.S.; Papadimitriou, C.; Faliakou, E.; Christodoulou, C.; Boukovinas, I.; Razis, E.; Tryfonopoulos, D.; et al. One in three highly selected Greek patients with breast cancer carries a loss-of-function variant in a cancer susceptibility gene. J. Med Genet. 2019, 57, 53–61. [Google Scholar] [CrossRef] [PubMed]
  81. Norquist, B.; Harrell, M.; Walsh, T.; Lee, M.; King, M.; Davidson, S.; Mannel, R.; DiSilvestro, P.; Swisher, E.; Birrer, M. Germline mutations in DNA repair genes in women with ovarian, peritoneal, or fallopian tube cancer treated on GOG protocols 218 and 262. Gynecol. Oncol. 2014, 133, 6. [Google Scholar] [CrossRef]
  82. Golmard, L.; Castéra, L.; Krieger, S.; Moncoutier, V.; Abidallah, K.; Tenreiro, H.; Laugé, A.; Tarabeux, J.; Millot, G.A.; Nicolas, A.; et al. Contribution of germline deleterious variants in the RAD51 paralogs to breast and ovarian cancers. Eur. J. Hum. Genet. 2017, 25, 1345–1353. [Google Scholar] [CrossRef] [PubMed]
  83. Zhao, Q.; Yang, J.; Li, L.; Cao, D.; Yu, M.; Shen, K. Germline and somatic mutations in homologous recombination genes among Chinese ovarian cancer patients detected using next-generation sequencing. J. Gynecol. Oncol. 2017, 28, e39. [Google Scholar] [CrossRef]
  84. Subramanian, D.N.; Zethoven, M.; McInerny, S.; Morgan, J.A.; Rowley, S.M.; Lee, J.E.A.; Li, N.; Gorringe, K.L.; James, P.A.; Campbell, I.G. Exome sequencing of familial high-grade serous ovarian carcinoma reveals heterogeneity for rare candidate susceptibility genes. Nat. Commun. 2020, 11, 1640. [Google Scholar] [CrossRef]
  85. Srivastava, S.; Olson, H.E.; Cohen, J.S.; Gubbels, C.S.; Lincoln, S.; Davis, B.T.; Shahmirzadi, L.; Gupta, S.; Picker, J.; Yu, T.W.; et al. BRAT1 mutations present with a spectrum of clinical severity. Am. J. Med. Genet. A 2016, 170, 2265–2273. [Google Scholar] [CrossRef]
  86. Pennington, K.P.; Walsh, T.; Harrell, M.I.; Lee, M.K.; Pennil, C.C.; Rendi, M.H.; Thornton, A.; Norquist, B.M.; Casadei, S.; Nord, A.S.; et al. Germline and somatic mutations in homologous recombination genes predict platinum response and survival in ovarian, fallopian tube, and peritoneal carcinomas. Clin. Cancer Res. 2014, 20, 764–775. [Google Scholar] [CrossRef]
  87. Lu, C.; Xie, M.; Wendl, M.C.; Wang, J.; McLellan, M.D.; Leiserson, M.D.M.; Huang, K.-L.; Wyczalkowski, M.A.; Jayasinghe, R.; Banerjee, T.; et al. Patterns and functional implications of rare germline variants across 12 cancer types. Nat. Commun. 2015, 6, 10086. [Google Scholar] [CrossRef]
  88. Stradella, A.; Del Valle, J.; Rofes, P.; Vargas-Parra, G.; Salinas, M.; González, S.; Montes, E.; López-Doriga, A.; Gómez, C.; de Cid, R.; et al. ERCC3, a new ovarian cancer susceptibility gene? Eur. J. Cancer 2020, 141, 1–8. [Google Scholar] [CrossRef] [PubMed]
  89. Soukupova, J.; Zemankova, P.; Nehasil, P.; Kleibl, Z.; Kleibl, Z.; Soukupová, J.; Janatová, M.; Zemánková, P.; Černá, M.; Jelínková, S.; et al. Re: ERCC3, a new ovarian cancer susceptibility gene? Eur. J. Cancer 2021, 150, 278–280. [Google Scholar] [CrossRef]
  90. Meienberg, J.; Bruggmann, R.; Oexle, K.; Matyas, G. Clinical sequencing: Is WGS the better WES? Hum. Genet. 2016, 135, 359–362. [Google Scholar] [CrossRef] [PubMed]
  91. Cavalieri, S.; Pozzi, E.; Gatti, R.A.; Brusco, A. Deep-intronic ATM mutation detected by genomic resequencing and corrected in vitro by antisense morpholino oligonucleotide (AMO). Eur. J. Hum. Genet. 2013, 21, 774–778. [Google Scholar] [CrossRef] [PubMed]
  92. Evans, D.R.; van Veen, E.M.; Byers, H.J.; Wallace, A.J.; Ellingford, J.M.; Beaman, G.; Santoyo-Lopez, J.; Aitman, T.J.; Eccles, D.M.; Lalloo, F.I.; et al. A Dominantly Inherited 5’ UTR Variant Causing Methylation-Associated Silencing of BRCA1 as a Cause of Breast and Ovarian Cancer. Am. J. Hum. Genet. 2018, 103, 213–220. [Google Scholar] [CrossRef] [PubMed]
  93. Rusch, M.; Nakitandwe, J.; Shurtleff, S.; Newman, S.; Zhang, Z.; Edmonson, M.N.; Parker, M.; Jiao, Y.; Ma, X.; Liu, Y.; et al. Clinical cancer genomic profiling by three-platform sequencing of whole genome, whole exome and transcriptome. Nat. Commun. 2018, 9, 3962. [Google Scholar] [CrossRef] [PubMed]
  94. Funingana, I.; Trotman, J.; Ambrose, J.; Roberts, T.; Watkins, J.; Ridley, M.; Gilson, B.; Freeman, S.; Jimenez-Linan, M.; Sosinsky, A.; et al. 7P Integration of whole genome sequencing (WGS) into NHS pathways for high-grade ovarian cancer (HGOC): A single-centre prospective experience. ESMO Open 2023, 8, 100861. [Google Scholar] [CrossRef]
  95. Guan, Z.; Begg, C.B.; Shen, R. Predicting Cancer Risk from Germline Whole-exome Sequencing Data Using a Novel Context-based Variant Aggregation Approach. Cancer Res. Commun. 2023, 3, 483–488. [Google Scholar] [CrossRef]
  96. Domchek, S.M.; Tang, J.; Stopfer, J.; Lilli, D.R.; Hamel, N.; Tischkowitz, M.; Monteiro, A.N.A.; Messick, T.E.; Powers, J.; Yonker, A.; et al. Biallelic deleterious BRCA1 mutations in a woman with early-onset ovarian cancer. Cancer Discov. 2013, 3, 399–405. [Google Scholar] [CrossRef] [PubMed]
  97. Scherz, A.; Stoll, S.; Rothlisberger, B.; Rabaglio, M. A New de novo BRCA1 Mutation in a Young Breast Cancer Patient: A Case Report. Appl. Clin. Genet. 2023, 16, 83–87. [Google Scholar] [CrossRef] [PubMed]
  98. Tenedini, E.; Piana, S.; Toss, A.; Marino, M.; Barbieri, E.; Artuso, L.; Venturelli, M.; Gasparini, E.; Mandato, V.D.; Marchi, I.; et al. Constitutional Mosaicism: A Critical Issue in the Definition of BRCA-Inherited Cancer Risk. JCO Precis. Oncol. 2022, 6, e2200138. [Google Scholar] [CrossRef] [PubMed]
  99. Witkowski, L.; Lalonde, E.; Zhang, J.; Albrecht, S.; Hamel, N.; Cavallone, L.; May, S.T.; Nicholson, J.C.; Coleman, N.; Murray, M.J.; et al. Familial rhabdoid tumour ‘avant la lettre’—From pathology review to exome sequencing and back again. J. Pathol. 2013, 231, 35–43. [Google Scholar] [CrossRef] [PubMed]
  100. Golmard, L.; Delnatte, C.; Laugé, A.; Moncoutier, V.; Lefol, C.; Abidallah, K.; Tenreiro, H.; Copigny, F.; Giraudeau, M.; Guy, C.; et al. Breast and ovarian cancer predisposition due to de novo BRCA1 and BRCA2 mutations. Oncogene 2016, 35, 1324–1327. [Google Scholar] [CrossRef] [PubMed]
  101. von Hardenberg, S.; Wallaschek, H.; Du, C.; Schmidt, G.; Auber, B. A holistic approach to maximise diagnostic output in trio exome sequencing. Front. Pediatr. 2023, 11, 1183891. [Google Scholar] [CrossRef] [PubMed]
  102. Speight, B.; Colvin, E.; Epurescu, E.D.; Drummond, J.; Verhoef, S.; Pereira, M.; Evans, D.G.; Tischkowitz, M. Low-level constitutional mosaicism of BRCA1 in two women with young onset ovarian cancer. Hered. Cancer Clin. Pract. 2022, 20, 32. [Google Scholar] [CrossRef]
  103. Alhopuro, P.; Vainionpää, R.; Anttonen, A.-K.; Aittomäki, K.; Nevanlinna, H.; Pöyhönen, M. Constitutional mosaicism for a BRCA2 mutation as a cause of early-onset breast cancer. Fam. Cancer 2020, 19, 307–310. [Google Scholar] [CrossRef]
  104. Schwartz, M.; Ibadioune, S.; Chansavang, A.; Vacher, S.; Caputo, S.M.; Delhomelle, H.; Wong, J.; Abidallah, K.; Moncoutier, V.; Becette, V.; et al. Mosaic BRCA1 promoter methylation contribution in hereditary breast/ovarian cancer pedigrees. J. Med. Genet. 2023. [Google Scholar] [CrossRef]
  105. Pinto, D.; Pinto, C.; Guerra, J.; Pinheiro, M.; Santos, R.; Vedeld, H.M.; Yohannes, Z.; Peixoto, A.; Santos, C.; Pinto, P.; et al. Contribution of MLH1 constitutional methylation for Lynch syndrome diagnosis in patients with tumor MLH1 downregulation. Cancer Med. 2018, 7, 433–444. [Google Scholar] [CrossRef]
  106. Rantala, J.N.J.; Heikkinen, S.M.M.; Hirvonen, E.M.; Tanskanen, T.; Malila, N.K.; Pitkäniemi, J.M. Familial aggregation of early-onset cancers in early-onset breast cancer families. Int. J. Cancer 2023, 153, 331–340. [Google Scholar] [CrossRef] [PubMed]
  107. Imbert-Bouteille, M.; Corsini, C.; Picot, M.-C.; Mizrahy, L.; Akouete, S.; Huguet, H.; Thomas, F.; Geneviève, D.; Taourel, P.; Ychou, M.; et al. No Association of Early-Onset Breast or Ovarian Cancer with Early-Onset Cancer in Relatives in BRCA1 or BRCA2 Mutation Families. Genes 2021, 12, 1100. [Google Scholar] [CrossRef] [PubMed]
  108. Stratton, J.F.; Pharoah, P.; Smith, S.K.; Easton, D.; Ponder, B.A.J. A systematic review and meta-analysis of family history and risk of ovarian cancer. BJOG Int. J. Obstet. Gynaecol. 1998, 105, 493–499. [Google Scholar] [CrossRef] [PubMed]
  109. Eng, K.H.; Szender, J.B.; Etter, J.L.; Kaur, J.; Poblete, S.; Huang, R.Y.; Zhu, Q.; Grzesik, K.A.; Battaglia, S.; Cannioto, R.; et al. Paternal lineage early onset hereditary ovarian cancers: A Familial Ovarian Cancer Registry study. PLoS Genet. 2018, 14, e1007194. [Google Scholar] [CrossRef] [PubMed]
  110. Buller, R.E.; Sood, A.K.; Lallas, T.; Buekers, T.; Skilling, J.S. Association between nonrandom X-chromosome inactivation and BRCA1 mutation in germline DNA of patients with ovarian cancer. J. Natl. Cancer Inst. 1999, 91, 339–346. [Google Scholar] [CrossRef] [PubMed]
  111. Liu, R.; Kain, M.; Wang, L. Inactivation of X-linked tumor suppressor genes in human cancer. Future Oncol. 2012, 8, 463–481. [Google Scholar] [CrossRef] [PubMed]
  112. Lose, F.; Duffy, D.L.; Kay, G.F.; Kedda, M.A.; Spurdle, A.B. Skewed X chromosome inactivation and breast and ovarian cancer status: Evidence for X-linked modifiers of BRCA1. J. Natl. Cancer Inst. 2008, 100, 1519–1529. [Google Scholar] [CrossRef]
  113. Winham, S.J.; Larson, N.B.; Armasu, S.M.; Fogarty, Z.C.; Larson, M.C.; McCauley, B.M.; Wang, C.; Lawrenson, K.; Gayther, S.; Cunningham, J.M.; et al. Molecular signatures of X chromosome inactivation and associations with clinical outcomes in epithelial ovarian cancer. Hum. Mol. Genet. 2019, 28, 1331–1342. [Google Scholar] [CrossRef]
  114. Qing, T.; Mohsen, H.; Marczyk, M.; Ye, Y.; O’meara, T.; Zhao, H.; Townsend, J.P.; Gerstein, M.; Hatzis, C.; Kluger, Y.; et al. Germline variant burden in cancer genes correlates with age at diagnosis and somatic mutation burden. Nat. Commun. 2020, 11, 2438. [Google Scholar] [CrossRef]
  115. Jia, G.; Lu, Y.; Wen, W.; Long, J.; Liu, Y.; Tao, R.; Li, B.; Denny, J.C.; Shu, X.-O.; Zheng, W. Evaluating the Utility of Polygenic Risk Scores in Identifying High-Risk Individuals for Eight Common Cancers. JNCI Cancer Spectr. 2020, 4, pkaa021. [Google Scholar] [CrossRef]
  116. Yang, X.; Leslie, G.; Gentry-Maharaj, A.; Ryan, A.; Intermaggio, M.; Lee, A.; Kalsi, J.K.; Tyrer, J.; Gaba, F.; Manchanda, R.; et al. Evaluation of polygenic risk scores for ovarian cancer risk prediction in a prospective cohort study. J. Med Genet. 2018, 55, 546–554. [Google Scholar] [CrossRef] [PubMed]
  117. Dareng, E.O.; Tyrer, J.P.; Barnes, D.R.; Jones, M.R.; Yang, X.; Aben, K.K.H.; Adank, M.A.; Agata, S.; Andrulis, I.L.; Anton-Culver, H.; et al. Polygenic risk modeling for prediction of epithelial ovarian cancer risk. Eur. J. Hum. Genet. 2022, 30, 349–362. [Google Scholar] [CrossRef] [PubMed]
  118. Borde, J.; Laitman, Y.; Blümcke, B.; Niederacher, D.; Weber-Lassalle, K.; Sutter, C.; Rump, A.; Arnold, N.; Wang-Gohrke, S.; Horváth, J.; et al. Polygenic risk scores indicate extreme ages at onset of breast cancer in female BRCA1/2 pathogenic variant carriers. BMC Cancer 2022, 22, 706. [Google Scholar] [CrossRef] [PubMed]
  119. Fatapour, Y.; Brody, J.P. Genetic Risk Scores and Missing Heritability in Ovarian Cancer. Genes 2023, 14, 762. [Google Scholar] [CrossRef] [PubMed]
  120. Rebbeck, T.R.; Friebel, T.M.; Mitra, N.; Wan, F.; Chen, S.; Andrulis, I.L.; Apostolou, P.; Arnold, N.; Arun, B.K.; Barrowdale, D.; et al. Inheritance of deleterious mutations at both BRCA1 and BRCA2 in an international sample of 32,295 women. Breast Cancer Res. 2016, 18, 112. [Google Scholar] [CrossRef] [PubMed]
  121. Zhu, Q.; Wang, J.; Yu, H.; Hu, Q.; Bateman, N.W.; Long, M.; Rosario, S.; Schultz, E.; Dalgard, C.L.; Wilkerson, M.D.; et al. Whole-Genome Sequencing Identifies PPARGC1A as a Putative Modifier of Cancer Risk in BRCA1/2 Mutation Carriers. Cancers 2022, 14, 2350. [Google Scholar] [CrossRef] [PubMed]
  122. Laitman, Y.; Michaelson-Cohen, R.; Chen-Shtoyerman, R.; Goldberg, Y.; Reish, O.; Bernstein-Molho, R.; Levy-Lahad, E.; Ben Baruch, N.E.; Kedar, I.; Evans, D.G.; et al. Age at diagnosis of cancer in 185delAG BRCA1 mutation carriers of diverse ethnicities: Tentative evidence for modifier factors. Fam. Cancer 2021, 20, 189–194. [Google Scholar] [CrossRef]
  123. Lee, C.H.; Subramanian, S.; Beck, A.H.; Espinosa, I.; Senz, J.; Zhu, S.X.; Huntsman, D.; van de Rijn, M.; Gilks, C.B. MicroRNA profiling of BRCA1/2 mutation-carrying and non-mutation-carrying high-grade serous carcinomas of ovary. PLoS ONE 2009, 4, e7314. [Google Scholar] [CrossRef]
  124. Pastrello, C.; Polesel, J.; Della Puppa, L.; Viel, A.; Maestro, R. Association between hsa-mir-146a genotype and tumor age-of-onset in BRCA1/BRCA2-negative familial breast and ovarian cancer patients. Carcinogenesis 2010, 31, 2124–2126. [Google Scholar] [CrossRef]
  125. Zhang, K.; Chandrakasan, S.; Chapman, H.; Valencia, C.A.; Husami, A.; Kissell, D.; Johnson, J.A.; Filipovich, A.H. Synergistic defects of different molecules in the cytotoxic pathway lead to clinical familial hemophagocytic lymphohistiocytosis. Blood 2014, 124, 1331–1334. [Google Scholar] [CrossRef]
  126. Moreno-Ruiz, N.; Ambrose, J.C.; Arumugam, P.; Baple, E.L.; Bleda, M.; Boardman-Pretty, F.; Boissiere, J.M.; Boustred, C.R.; Brittain, H.; Caulfield, M.J.; et al. Assessing the digenic model in rare disorders using population sequencing data. Eur. J. Hum. Genet. 2022, 30, 1439–1443. [Google Scholar] [CrossRef]
  127. Kamar, A.; Khalil, A.; Nemer, G. The Digenic Causality in Familial Hypercholesterolemia: Revising the Genotype-Phenotype Correlations of the Disease. Front. Genet. 2020, 11, 572045. [Google Scholar] [CrossRef] [PubMed]
  128. Ameratunga, R.; Woon, S.-T.; Bryant, V.L.; Steele, R.; Slade, C.; Leung, E.Y.; Lehnert, K. Clinical Implications of Digenic Inheritance and Epistasis in Primary Immunodeficiency Disorders. Front. Immunol. 2017, 8, 1965. [Google Scholar] [CrossRef]
  129. Nachtegael, C.; Gravel, B.; Dillen, A.; Smits, G.; Nowé, A.; Papadimitriou, S.; Lenaerts, T. Scaling up oligogenic diseases research with OLIDA: The Oligogenic Diseases Database. Database 2022, 2022, baac023. [Google Scholar] [CrossRef] [PubMed]
  130. Fijneman, R.J.; de Vries, S.S.; Jansen, R.C.; Demant, P. Complex interactions of new quantitative trait loci, Sluc1, Sluc2, Sluc3, and Sluc4, that influence the susceptibility to lung cancer in the mouse. Nat. Genet. 1996, 14, 465–467. [Google Scholar] [CrossRef] [PubMed]
  131. Tansel, A.; Katz, L.H.; El-Serag, H.B.; Thrift, A.P.; Parepally, M.; Shakhatreh, M.H.; Kanwal, F. Incidence and Determinants of Hepatocellular Carcinoma in Autoimmune Hepatitis: A Systematic Review and Meta-analysis. Clin. Gastroenterol. Hepatol. 2017, 15, 1207–1217.e4. [Google Scholar] [CrossRef]
  132. Helicobacter and Cancer Collaborative Group. Gastric cancer and Helicobacter pylori: A combined analysis of 12 case control studies nested within prospective cohorts. Gut 2001, 49, 347. [Google Scholar] [CrossRef]
  133. Gausman, V.; Dornblaser, D.; Anand, S.; Hayes, R.B.; O’Connell, K.; Du, M.; Liang, P.S. Risk Factors Associated With Early-Onset Colorectal Cancer. Clin. Gastroenterol. Hepatol. 2020, 18, 2752–2759.e2. [Google Scholar] [CrossRef]
  134. Bae, E.; Lim, S.; Han, K.-D.; Jung, J.-H.; Choi, H.; Kim, C.; Ma, S.; Kim, S. Systemic lupus erythematosus is a risk factor for cancer: A nationwide population-based study in Korea. Lupus 2019, 28, 317–323. [Google Scholar] [CrossRef]
  135. Kübler, K.; Arndt, P.F.; Wardelmann, E.; Krebs, D.; Kuhn, W.; van der Ven, K. HLA-class II haplotype associations with ovarian cancer. Int. J. Cancer. 2006, 119, 2980–2985. [Google Scholar] [CrossRef]
  136. Carbone, M.; Arron, S.T.; Beutler, B.; Bononi, A.; Cavenee, W.; Cleaver, J.E.; Croce, C.M.; D’andrea, A.; Foulkes, W.D.; Gaudino, G.; et al. Tumour predisposition and cancer syndromes as models to study gene-environment interactions. Nat. Rev. Cancer 2020, 20, 533–549. [Google Scholar] [CrossRef] [PubMed]
  137. Novelli, F.; Bononi, A.; Wang, Q.; Bai, F.; Patergnani, S.; Kricek, F.; Haglund, E.; Suarez, J.S.; Tanji, M.; Xu, R.; et al. BAP1 forms a trimer with HMGB1 and HDAC1 that modulates gene x environment interaction with asbestos. Proc. Natl. Acad. Sci. USA 2021, 118, e2111946118. [Google Scholar] [CrossRef] [PubMed]
  138. Kurzynska-Kokorniak, A.; Koralewska, N.; Pokornowska, M.; Urbanowicz, A.; Tworak, A.; Mickiewicz, A.; Figlerowicz, M. The many faces of Dicer: The complexity of the mechanisms regulating Dicer gene expression and enzyme activities. Nucleic Acids Res. 2015, 43, 4365–4380. [Google Scholar] [CrossRef] [PubMed]
  139. Nickerson, M.L.; Warren, M.B.; Toro, J.R.; Matrosova, V.; Glenn, G.; Turner, M.L.; Duray, P.; Merino, M.; Choyke, P.; Pavlovich, C.P.; et al. Mutations in a novel gene lead to kidney tumors, lung wall defects, and benign tumors of the hair follicle in patients with the Birt-Hogg-Dubé syndrome. Cancer Cell 2002, 2, 157–164. [Google Scholar] [CrossRef] [PubMed]
  140. L’espérance, K.; Grundy, A.; Abrahamowicz, M.; Arseneau, J.; Gilbert, L.; Gotlieb, W.H.; Provencher, D.; Koushik, A. Alcohol intake and the risk of epithelial ovarian cancer. Cancer Causes Control 2023, 34, 533–541. [Google Scholar] [CrossRef] [PubMed]
  141. Wang, T.; Townsend, M.K.; Vinci, C.; Jake-Schoffman, D.E.; Tworoger, S.S. Early life exposure to tobacco smoke and ovarian cancer risk in adulthood. Int. J. Epidemiol. 2021, 50, 965–974. [Google Scholar] [CrossRef] [PubMed]
  142. Collaborative Group on Epidemiological Studies of Ovarian Cancer. Ovarian cancer and smoking: Individual participant meta-analysis including 28,114 women with ovarian cancer from 51 epidemiological studies. Lancet Oncol. 2012, 13, 946–956. [Google Scholar] [CrossRef] [PubMed]
  143. Mitamura, T.; Zhai, T.; Hatanaka, K.C.; Hatanaka, Y.; Amano, T.; Wang, L.; Tanaka, S.; Watari, H. Germline PRDM1 Variant rs2185379 in Long-Term Recurrence-Free Survivors of Advanced Ovarian Cancer. Pharmacogenomics Pers. Med. 2022, 15, 977–984. [Google Scholar] [CrossRef] [PubMed]
  144. Shi, T.; Jiang, R.; Wang, P.; Xu, Y.; Yin, S.; Cheng, X.; Zang, R. Significant association of the EXO1 rs851797 polymorphism with clinical outcome of ovarian cancer. Onco Targets Ther. 2017, 10, 4841–4851. [Google Scholar] [CrossRef]
Figure 1. Frequency of germline pathogenic/likely pathogenic variants in established cancer predisposition genes in early-onset vs. late-onset OC patients.
Figure 1. Frequency of germline pathogenic/likely pathogenic variants in established cancer predisposition genes in early-onset vs. late-onset OC patients.
Ijms 24 17020 g001
Figure 2. Proposed components of early-onset OC predisposition (AD, autosomal dominant; AR, autosomal recessive; GPV, germline pathogenic/likely pathogenic variant; OC, ovarian cancer).
Figure 2. Proposed components of early-onset OC predisposition (AD, autosomal dominant; AR, autosomal recessive; GPV, germline pathogenic/likely pathogenic variant; OC, ovarian cancer).
Ijms 24 17020 g002
Table 1. Characteristics of early-onset and late-onset OC.
Table 1. Characteristics of early-onset and late-onset OC.
PatientsEarly-Onset OCLate-Onset OC
Incidence in females1.6/100,000 [3,15]22.0/100,000 [3,15]
5-year relative survival rate58–87% [17,19,20,23]
-lower in LGSC [22]
Approx. 50% [3,17]
Clinicopathology
Histology~40% epithelial—LGSC prevails [16,17]
~50% germ-cell [16,24]
~10% sex cord-stromal [24,25]
~90% epithelial—HGSC prevails (70%) [13,26]
~6% sex cord-stromal [6]
~3% germ-cell [6]
Dominant tumor stageLocalized disease [3]Distant disease [3]
Genetic predispositions
GPVLow <10% [7,8,9]High >20% [4,5,9]
GPV, germline pathogenic/likely pathogenic variant; HGSC, high-grade serous carcinoma; LGSC, low-grade serous carcinoma; OC, ovarian cancer.
Table 3. Studies involving early-onset OC patients.
Table 3. Studies involving early-onset OC patients.
StudyPopulationStudy DetailsNo. of Tested Genes *No. of All OC PatientsNo. of Early-Onset OC PatientsRange of Early-Onset Patients’ Age at Dg.Early-Onset OC Patients
No. of High-Penetrance GPV CarriersGPVs in Established High-Penetrance OC Predisposition GenesGPVs in Candidate OC Predisposition Genes
Stratton (1999) [8]UKEarly-onset epithelial OC4 **16916913–300 ***00
Carter (2018) [9]USOC1544391476–302 (1.4%)BRCA1; 1×BRIP1ATM; 1×BARD1; 5×CHEK2
Lhotova (2020) [4]CZOC21913338415–306 (7.1%)BRCA1; 1×BRCA2; 2×RAD51C; 1×STK11ATM; 1×BARD1; 4×CHEK2; 1×NBN;
Herold (2023) [51]GEROC252068313–303 (3.6%)BRIP1; 2×SMARCA4 FANCM; 1×MUTYH het; 1×PMS2; 1×TP53
Flaum (2023) [40]UKEarly-onset OC15777715–304 (5.2%)MSH2 PMS2
Bernards (2015) [38]USEarly-onset OC1847527–30000
Felicio (2020) [55]BRABRCA neg., TP53 neg. HBOC patientsWES11320–2100CHEK2
Da Costa (2020) [56]BRAHBOC216222–30000
Boyd (2000) [57]JewOC2189125000
Hajkova (2019) [58]CZSynchronous EC and OC732212900BARD1
Jarhelle (2019) [59]NORHBOC BRCA1/BRCA2 neg. 942012700CHEK2
Risch (2001) [7]USOC2649NA (96 <40 yo)20-30000
Koczkowska (2018) [60]PLOC25333NANA00CHEK2
Pal (2005) [61]USepithelial OC2209NA (11 <40 yo)18-30000
Ryan (2017) [52]UKLS assoc. preselected OC positive for LS-GPV453NA24-302MSH2PMS2 biallelic
Hajkova (2019) [58]CZSynchronous EC and OC732212900BARD1
Dg, diagnosis; GPV, germline pathogenic variants; HBOC, hereditary breast and ovarian cancer; LS, Lynch syndrome; OC, ovarian cancer; NA, not available; No.; number; WES, whole exome sequencing; yo, years old; * The list of tested genes in each study is available in Supplementary Table S1; ** in the case of BRCA2, only the OC cluster region was analyzed; *** identified variants; MLH1: c.1853A > C/p.Lys618Thr and MLH1: c.1000G > C/p.Asp304His are not currently considered GPVs.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Horackova, K.; Janatova, M.; Kleiblova, P.; Kleibl, Z.; Soukupova, J. Early-Onset Ovarian Cancer <30 Years: What Do We Know about Its Genetic Predisposition? Int. J. Mol. Sci. 2023, 24, 17020. https://doi.org/10.3390/ijms242317020

AMA Style

Horackova K, Janatova M, Kleiblova P, Kleibl Z, Soukupova J. Early-Onset Ovarian Cancer <30 Years: What Do We Know about Its Genetic Predisposition? International Journal of Molecular Sciences. 2023; 24(23):17020. https://doi.org/10.3390/ijms242317020

Chicago/Turabian Style

Horackova, Klara, Marketa Janatova, Petra Kleiblova, Zdenek Kleibl, and Jana Soukupova. 2023. "Early-Onset Ovarian Cancer <30 Years: What Do We Know about Its Genetic Predisposition?" International Journal of Molecular Sciences 24, no. 23: 17020. https://doi.org/10.3390/ijms242317020

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop