Next Article in Journal
Multifactorial Diseases of the Heart, Kidneys, Lungs, and Liver and Incident Cancer: Epidemiology and Shared Mechanisms
Next Article in Special Issue
Uptake and Effectiveness of Risk-Reducing Surgeries in Unaffected Female BRCA1 and BRCA2 Carriers: A Single Institution Experience in the Czech Republic
Previous Article in Journal
Diagnostic and Therapeutic Algorithm for Appendiceal Tumors and Pseudomyxoma Peritonei: A Consensus of the Peritoneal Malignancies Oncoteam of the Italian Society of Surgical Oncology (SICO)
Previous Article in Special Issue
Long-Term Non-Cancer Risks in People with BRCA Mutations following Risk-Reducing Bilateral Salpingo-Oophorectomy and the Role of Hormone Replacement Therapy: A Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Is Reflex Germline BRCA1/2 Testing Necessary in Women Diagnosed with Non-Mucinous High-Grade Epithelial Ovarian Cancer Aged 80 Years or Older?

1
Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester M20 4BX, UK
2
Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK
3
Manchester Centre for Genomic Medicine, North West Genomic Laboratory Hub, Saint Mary’s Hospital, Oxford Road, Manchester M13 9WL, UK
4
Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK
5
Department of Clinical Genetics, Saint Mary’s Hospital, Oxford Road, Manchester M13 9WL, UK
6
Department of Gynaecological Oncology, Saint Mary’s Hospital, Oxford Road, Manchester M13 9WL, UK
*
Authors to whom correspondence should be addressed.
Cancers 2023, 15(3), 730; https://doi.org/10.3390/cancers15030730
Submission received: 4 December 2022 / Revised: 21 January 2023 / Accepted: 23 January 2023 / Published: 25 January 2023

Abstract

:

Simple Summary

Approximately 15% of patients diagnosed with high-grade non-mucinous epithelial ovarian cancer (EOC) have a germline BRCA1/2 mutation, although all patients are often able to access germline testing. Importantly, the risk of familial ovarian cancer reduces with advancing age at diagnosis. The aim of our study was to determine the prevalence of germline and somatic BRCA1/2 mutations in women diagnosed with non-mucinous high-grade EOC aged ≥80. We found that somatic BRCA1/2 mutations occurred nine times more frequently than germline BRCA1/2 mutations in women aged ≥80. The only germline BRCA1/2 mutation reported in a patient aged ≥80 was detected in both germline and tumour DNA. These data suggest that germline BRCA1/2 testing in women diagnosed with high-grade non-mucinous EOC aged ≥80 can be reserved for those with a detectable tumour BRCA1/2 mutation.

Abstract

Women diagnosed with non-mucinous high-grade epithelial ovarian cancer (EOC) in England are often reflex-tested for germline and tumour BRCA1/2 variants. The value of germline BRCA1/2 testing in women diagnosed aged ≥80 is questionable. We performed an observational study of all women diagnosed with non-mucinous high-grade EOC who underwent germline and tumour BRCA1/2 testing by the North West of England Genomic Laboratory Hub. A subgroup of women also underwent germline testing using a panel of homologous recombination repair (HRR) genes and/or tumour testing for homologous recombination deficiency (HRD) using Myriad’s myChoice® companion diagnostic. Seven-hundred-two patients successfully underwent both germline and tumour BRCA1/2 testing. Of these, 48 were diagnosed with non-mucinous high-grade EOC aged ≥80. In this age group, somatic BRCA1/2 pathogenic/likely pathogenic variants (PV/LPVs) were detected nine times more often than germline BRCA1/2 PV/LPVs. The only germline PV reported in a patient aged ≥80 was detected in germline and tumour DNA (BRCA2 c.4478_4481del). No patient aged ≥80 had a germline PV/LPVs in a non-BRCA1/2 HRR gene. Thirty-eight percent of patients aged ≥80 had a tumour positive for HRD. Our data suggest that tumour BRCA1/2 and HRD testing is adequate for patients diagnosed with non-mucinous high-grade EOC aged ≥80, with germline BRCA1/2 testing reserved for women with a tumour BRCA1/2 PV/LPVs.

1. Introduction

Inclusion of poly (ADP-ribose) polymerase inhibitors (PARPi) as standard therapy for non-mucinous high-grade epithelial ovarian cancer (EOC) led to mainstream, reflex germline and tumour BRCA1/2 testing [1]. Prior to the era of PARPi, index cases of ovarian cancer were selected for germline BRCA1/2 testing based on their risk of hereditary cancer, with age at diagnosis and family history used to determine risk [2,3,4]. The present lack of selection criteria for germline BRCA1/2 testing means many women at low risk of being a germline heterozygote, especially elderly women, undergo germline testing unnecessarily [5,6,7,8,9,10,11].
The range of genetic tests available for women diagnosed with ovarian cancer is expanding. In England, national guidelines specify that all women with newly diagnosed high-grade ovarian cancer undergo tumour testing for BRCA1/2 pathogenic/likely pathogenic variants and homologous recombination deficiency (HRD). National guidelines also specify that all women diagnosed with non-mucinous high-grade EOC undergo germline testing for genes associated with familial ovarian cancer regardless of age. These cancer predisposition genes include BRCA1, BRCA2, BRIP1, PALB2, RAD51C, RAD51D, MLH1, MSH2 and MSH6 [12]. This strategy of germline testing all unselected cases of ovarian cancer provides an opportunity to determine prevalence rates across age groups, thereby optimizing future testing pathways [13].
In this study, we report the prevalence of pathogenic/likely pathogenic variants (hereon referred to as ‘pathogenic variants’) in ovarian cancer susceptibility genes in a large group of unselected women diagnosed with non-mucinous high-grade EOC. In addition, we report the prevalence of somatic BRCA1/2 pathogenic variants and tumours positive for HRD. Our aim was to define the optimal germline and tumour testing strategy for women diagnosed with non-mucinous high-grade EOC aged ≥80.

2. Materials and Methods

Unselected women were included who had been diagnosed with non-mucinous high-grade carcinoma of the ovary, fallopian tube or primary peritoneum who successfully underwent both germline and tumour BRCA1/2 testing by the Genomic Laboratory Hub in the North West of England between 1 May 2016 and 15 November 2022. Eligible histological subtypes included high-grade serous, high-grade endometrioid (moderately differentiated (grade 2) or poorly differentiated (grade 3)), clear cell and poorly differentiated adenocarcinoma not otherwise specified [14]. Cases of ovarian carcinosarcoma were excluded. All FIGO (International Federation of Gynecology and Obstetrics) stages of ovarian cancer were eligible for inclusion [15]. All index cases that failed germline or tumour BRCA1/2 testing were excluded.
The next-generation sequencing (NGS) and multiplex ligation-dependent probe amplification (MLPA) assays used to test for germline BRCA1/2 pathogenic variants have been described previously [16]. From 1 April 2022, germline DNA from index cases also routinely underwent multi-gene panel testing for pathogenic variants in BRCA1, BRCA2, BRIP1, PALB2, RAD51C, RAD51D, MLH1, MSH2 and MSH6 [12]. The genes associated with homologous recombination repair (HRR) included BRCA1, BRCA2, BRIP1, PALB2, RAD51C and RAD51D. The NGS enrichment method used a custom-designed Agilent SureSelectTM panel, including the coding region of transcripts and splice sites +/- 15 base pairs and known intronic pathogenic variants. Following NGS enrichment, samples were sequenced using the Illumina NextSeq 550 System. The overall coverage had to be >99% at 100X to pass the sample. Single nucleotide variants and small deletions, duplications, insertions, and insertion/deletions (<40 base pairs) were called using an in-house custom bioinformatic analysis pipeline validated to detect heterozygous and mosaic variants to a variant allele frequency (VAF) of ≥4%. The reference sequences used included NM_007294.3 (BRCA1), NM_000059.3 (BRCA2), NM_032043.2 (BRIP1), NM_024675.3 (PALB2), NM_058216.1 (RAD51C), NM_002878.3 (RAD51D), NM_001142571.1 (RAD51D), NM_000249.3 (MLH1), NM_000251.1 (MSH2) and NM_000179.2 (MSH6). Sequence variant nomenclature followed Human Genome Variation Society (HGVS) guidelines (http://varnomen.hgvs.org accessed on 16 November 2022). Sequence variants were classified according to the Association for Clinical Genomic Science (ACGS) Best Practice Guidelines [17].
The NGS assay used to test for tumour BRCA1/2 pathogenic variants has been described previously [18]. From 11 April 2021, tumour DNA from index cases was also routinely tested using Myriad’s myChoice® companion diagnostic (CDx) [19]. Myriad Genetics, Inc. (Salt Lake City, UT, USA) performed the myChoice® CDx. The eligibility criteria for myChoice® CDx testing included newly diagnosed FIGO stage III/IV high-grade epithelial ovarian, fallopian tube or primary peritoneal cancer. The myChoice® CDx reported BRCA1/2 pathogenic variants and a genomic instability score (GIS) in tumour DNA. The GIS was a composite score of three bioinformatic algorithms that detected putative biomarkers of HRD, including loss of heterozygosity (LOH), telomeric allelic imbalance (TAI) and large-scale state transitions (LST) [20,21,22]. A GIS of ≥42 was reported as GIS-positive, while a GIS of <42 was reported as GIS-negative. Any tumour with a BRCA1/2 pathogenic variant or a GIS of ≥42 was reported as HRD-positive. Any tumour with BRCA1/2 wild type and a GIS of <42 was reported as HRD-negative.
Categorical data were reported as number and percentage. Continuous data were reported as mean and range. For categorical data, the chi-squared test was used to determine if significant differences occurred between groups. For continuous data, Student’s t-test was used to determine if significant differences occurred between the means of two groups.

3. Results

Seven-hundred-two women diagnosed with non-mucinous high-grade EOC underwent germline and tumour BRCA1/2 testing (Table 1). Seventy-five (11%) and fifty-four (8%) women were found to have a germline or somatic BRCA1/2 pathogenic variant, respectively (Table 1). Most BRCA1/2 pathogenic variants were detected in women diagnosed with high-grade serous carcinoma (110/129; 85%), although this was the commonest histological subtype tested (623/702; 89%) (Table 1).
The concordance between germline BRCA1/2 pathogenic variants detected in germline and tumour DNA was 94% (44/47) and 100% (28/28) using our in-house tumour BRCA1/2 assay and Myriad’s myChoice® CDx, respectively. The three germline BRCA1/2 variants missed using our in-house tumour BRCA1/2 assay included BRCA1 Exon 13 duplication, BRCA2 Exon 1–2 deletion and BRCA2 Exon 14–16 deletion. Three germline BRCA1/2 large genomic rearrangements were detected in germline and tumour DNA using Myriad’s myChoice® CDx, including BRCA1 Exon 9-12 deletion (GIS 56), BRCA1 Exon 17 deletion (GIS 70) and BRCA2 Exon 14-16 deletion (GIS 36).
By categorizing germline and somatic BRCA1/2 pathogenic variants according to age, it was clear that no patient diagnosed aged ≥80 had a germline BRCA1 pathogenic variant (Table 2). The only germline BRCA2 pathogenic variant reported in a patient diagnosed aged ≥80 was detected in germline and tumour DNA (BRCA2 c.4478_4481del; GIS 83). Patients diagnosed aged ≥80 were nine times more likely to have a somatic versus germline BRCA1/2 pathogenic variant (Table 2). The likelihood of having a germline versus somatic BRCA1/2 pathogenic variant reduced with each decade of age at diagnosis ≥60 (Figure 1). These data suggest that routine germline BRCA1/2 testing is unnecessary in women diagnosed with non-mucinous high-grade EOC aged ≥80.
We next categorized GIS status according to age at diagnosis to determine whether genomic instability testing was necessary in women diagnosed with non-mucinous high-grade EOC aged ≥80. Of the 702 patients included in this study, 346 (49%) had been tested using Myriad’s myChoice® CDx (Table 3). Over 90% of the patients tested had been diagnosed with high-grade serous or high-grade endometrioid carcinoma (320/346; 92%) (Table 3). The mean age of patients with a GIS-positive versus GIS-negative tumour differed significantly (62.7 versus 66.0 years; p = 0.004) (Figure 2). For those patients aged ≥80, the likelihood of having a GIS-positive versus GIS-negative tumour was similar (Table 4). There was little variation in the likelihood of having a GIS-positive versus GIS-negative tumour across age groups (Figure 3). These data support the use of tumour BRCA1/2 and genomic instability score testing in women diagnosed with non-mucinous high-grade EOC aged ≥80.
Finally, we investigated the prevalence of germline pathogenic variants in non-BRCA1/2 HRR-associated genes in women diagnosed with non-mucinous high-grade EOC aged ≥80 who also underwent tumour testing using Myriad’s myChoice® CDx. Of the 702 patients included in this study, 174 had undergone germline multi-gene panel testing since 1 April 2022. In the 119 patients with germline and somatic BRCA1/2 wild type, four germline BRIP1 pathogenic variants were detected (Table 5). No other germline HRR genes were detected in this cohort. All four germline BRIP1 pathogenic variants were detected in women aged <70 at diagnosis with at least one first- or second-degree relative diagnosed with breast or ovarian cancer (Table 6). Two of the four BRIP1 pathogenic variants were detected in women with a GIS-positive tumour (Table 6). These data provide limited support for the use of germline multi-gene panel testing in women diagnosed aged ≥80, regardless of GIS status.

4. Discussion

Germline BRCA1/2 pathogenic variants are predictive and prognostic biomarkers in ovarian cancer. BRCA1/2-mutant ovarian tumours are highly sensitive to DNA damaging agents, such as platinum chemotherapy and PARPi [23,24,25,26,27]. Women diagnosed with germline BRCA1/2-mutant EOC have improved survival outcomes compared to sporadic cases [28]. More broadly, identifying germline BRCA1/2 pathogenic variants leads to cascade testing and risk-reducing strategies in related, unaffected germline heterozygotes [29,30]. Thus, detection of germline BRCA1/2 pathogenic variants is important for index cases and their family. However, germline BRCA1/2 pathogenic variants occur in only around 15% of EOC, meaning many patients, especially elderly women, undergo unnecessary testing [31].
Opinions differ regarding whether or not multi-gene germline and somatic BRCA1/2 testing is cost-effective for accessing PARPi therapy [32,33,34]. To reduce overall costs, multi-disciplinary teams could omit reflex germline testing in women at low risk of germline BRCA1/2 heterozygotes. Data from our study show that women diagnosed with non-mucinous high-grade EOC aged ≥80 can be considered ‘very low risk’ and, therefore, do not require reflex germline BRCA1/2 testing. Instead, germline testing could be reserved for women who have a BRCA1/2 pathogenic variant detected first in tumour DNA, or when tumour testing fails. Based on our data, germline BRCA1/2 testing in women aged ≥80 would be reduced by around 80% (10/48 patients would have required testing in our cohort). In the United Kingdom, over 1000 women are diagnosed each year with non-mucinous high-grade EOC aged ≥80, meaning an estimated 800 cases could avoid germline testing [35]. The multi-gene germline panel NGS test used in the North West Genomic Laboratory Hub costs GBP 550 per test. If testing costs are similar across the United Kingdom, we estimate cost savings of GBP 440,000 each year by omitting reflex germline testing in women aged ≥80.
The limitation of relying upon upfront tumour BRCA1/2 testing to identify all possible pathogenic variants is that most local tumour NGS assays are not yet validated to detect whole gene/exon deletions or duplications [11,36,37], although Myriad’s myChoice® CDx does reliably detect these variants [19]. Large genomic rearrangements account for <10% of all germline BRCA1/2 pathogenic variants [38]. In our study, all 8% of germline large genomic rearrangements occurred in women aged <70. The only patient diagnosed with a germline BRCA1/2 pathogenic variant aged ≥80 had a small deletion (four base pairs in size) in BRCA2 that was detected first in tumour DNA and then subsequently in germline DNA. These data support a strategy of performing only upfront tumour BRCA1/2 testing in women aged ≥80.
Identifying women with newly diagnosed advanced high-grade EOC and a GIS-positive tumour expands access to first-line olaparib plus bevacizumab maintenance therapy [39]. In addition, women with HRD-positive tests have better survival outcomes following PARPi maintenance monotherapy compared to those with HRD-negative tests [40,41,42,43,44]. Our study shows that genomic instability/mutational ‘scarring’ is not an age-defined biomarker. Indeed, in our cohort, there was little variation in the likelihood of an index case having a GIS-positive versus GIS-negative tumour across age groups. These data support use of HRD tumour testing in unselected cases of non-mucinous high-grade EOC regardless of age.
To assess the correlation between GIS-positive tumours and germline pathogenic variants in HRR genes, we analysed a subgroup of patients that were tested using Myriad’s myChoice® CDx and a standard panel of ovarian cancer susceptibility genes [12]. Unsurprisingly, germline BRCA1/2 pathogenic variants were the most frequently detected HRR mutations in women with GIS-positive tumours. The low prevalence rate of germline pathogenic variants in other HRR genes was predictable due to the lack of selection criteria used for HRR testing [45,46,47]. The absence of germline pathogenic variants in non-BRCA1/2 HRR genes in women aged ≥80, combined with the fact that only 50% of these variants were found in women with a GIS-positive tumour, suggest germline multi-gene panel testing has limited value in women aged ≥80 regardless of GIS status. Indeed, in all patients tested, the addition of four HRR genes brought about a very small uplift in the potential causes of HRD in BRCA1/2 wild type/GIS-positive tumours, with only 2/35 additional pathogenic variants identified. We recognize that the panel of genes used in our study includes only commonly tested ovarian cancer susceptibility genes, therefore not accounting for alternative genetic and epigenetic drivers of HRD [48,49,50,51].
It is unclear why two of the four patients with a germline BRIP1 pathogenic variant had a GIS-negative tumour. By contrast, only 3% (1/31) of patients with a germline BRCA1/2 pathogenic variant had a GIS-negative tumour (p = 0.002). One explanation could be the absence of biallelic inactivation of the BRIP1 gene, meaning such tumours were not BRIP1-protein-deficient and, therefore, HRR proficient [52]. This finding raises the question as to whether HRD-test-negative tumours containing a non-BRCA1/2 HRR pathogenic variant will respond to PARPi if they are in fact HRR proficient [53,54,55,56,57,58]. This will require close monitoring for poorer responses to PARPi in women with lower-penetrance HRR genes, in which ovarian cancers may have occurred sporadically. Interrogating tumour DNA for gene-specific loss of heterozygosity in germline carriers of non-BRCA1/2 HRR pathogenic variants may help to clarify whether borderline GIS-negative tumours have biallelic inactivation and are more likely to respond to PARPi [52].
There are two limitations with this study. Firstly, genetic testing practices varied during the study period. Thus, information on germline multi-gene panel and GIS testing was only available for a subgroup of patients. Secondly, the number of patients tested for non-BRCA1/2 HRR genes was relatively small when considering the population frequency of these moderate-to-low-penetrance genes. These two limitations make it difficult to draw any conclusions regarding age distribution and certainty, in order to make changes to policy recommendations.

5. Conclusions

We provide evidence demonstrating that the age-based threshold for reflex, mainstream germline BRCA1/2 testing in unselected women diagnosed with non-mucinous high-grade EOC could be set at <80 years old. In this age group, it may be more appropriate to focus resources on reflex BRCA1/2 and HRD tumour testing, with confirmatory germline BRCA1/2 testing reserved for those patients with a tumour-pathogenic variant.

Author Contributions

Conceptualization, R.D.M. and D.G.R.E.; Methodology, R.D.M., G.J.B. and D.G.R.E.; Formal analysis, R.D.M. and D.G.R.E.; Data curation, R.D.M. and D.G.R.E.; Writing—original draft preparation, R.D.M., G.J.B. and D.G.R.E.; Writing—review and editing, R.D.M., G.J.B., N.F., M.B., P.S., A.R.C., J.H., C.L.M., Z.S., E.R.W., F.L., E.J.C., R.J.E., H.S., G.C.J. and D.G.R.E. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki. The germline BRCA1/2 database is approved by North Manchester Research Ethics Committee (08/H1006/77). The Genetic Variants in Gynaecological Cancer database is approved by the Christie NHS Foundation Trust (59).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

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

Acknowledgments

E.R.W., E.J.C. and D.G.R.E. are supported by the Manchester National Institute for Health Research (NIHR) Manchester Biomedical Research Centre (IS-BRC-1215–20007). E.J.C. is supported by an NIHR Advanced Fellowship (NIHR300650). The services provided by the North West Genomic Laboratory Hub Manchester site and the Liverpool site are accredited by the United Kingdom Accreditation Service (UKAS) to ISO 15189 standards.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Mateo, J.; Lord, C.J.; Serra, V.; Tutt, A.; Balmana, J.; Castroviejo-Bermejo, M.; Cruz, C.; Oaknin, A.; Kaye, S.B.; de Bono, J.S. A decade of clinical development of PARP inhibitors in perspective. Ann. Oncol. 2019, 30, 1437–1447. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Parmigiani, G.; Berry, D.; Aguilar, O. Determining carrier probabilities for breast cancer-susceptibility genes BRCA1 and BRCA2. Am. J. Hum. Genet. 1998, 62, 145–158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Antoniou, A.C.; Pharoah, P.P.; Smith, P.; Easton, D.F. The BOADICEA model of genetic susceptibility to breast and ovarian cancer. Br. J. Cancer 2004, 91, 1580–1590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Evans, D.G.; Eccles, D.M.; Rahman, N.; Young, K.; Bulman, M.; Amir, E.; Shenton, A.; Howell, A.; Lalloo, F. A new scoring system for the chances of identifying a BRCA1/2 mutation outperforms existing models including BRCAPRO. J. Med. Genet. 2004, 41, 474–480. [Google Scholar] [CrossRef] [PubMed]
  5. George, A.; Riddell, D.; Seal, S.; Talukdar, S.; Mahamdallie, S.; Ruark, E.; Cloke, V.; Slade, I.; Kemp, Z.; Gore, M.; et al. Implementing rapid, robust, cost-effective, patient-centred, routine genetic testing in ovarian cancer patients. Sci. Rep. 2016, 6, 29506. [Google Scholar] [CrossRef] [Green Version]
  6. Plaskocinska, I.; Shipman, H.; Drummond, J.; Thompson, E.; Buchanan, V.; Newcombe, B.; Hodgkin, C.; Barter, E.; Ridley, P.; Ng, R.; et al. New paradigms for BRCA1/BRCA2 testing in women with ovarian cancer: Results of the Genetic Testing in Epithelial Ovarian Cancer (GTEOC) study. J. Med. Genet. 2016, 53, 655–661. [Google Scholar] [CrossRef] [Green Version]
  7. Rust, K.; Spiliopoulou, P.; Tang, C.Y.; Bell, C.; Stirling, D.; Phang, T.; Davidson, R.; Mackean, M.; Nussey, F.; Glasspool, R.; et al. Routine germline BRCA1 and BRCA2 testing in patients with ovarian carcinoma: Analysis of the Scottish real-life experience. BJOG Int. J. Obstet. Gynaecol. 2018, 125, 1451–1458. [Google Scholar] [CrossRef] [Green Version]
  8. Rahman, B.; Lanceley, A.; Kristeleit, R.S.; Ledermann, J.A.; Lockley, M.; McCormack, M.; Mould, T.; Side, L. Mainstreamed genetic testing for women with ovarian cancer: First-year experience. J. Med. Genet. 2019, 56, 195–198. [Google Scholar] [CrossRef]
  9. Rumford, M.; Lythgoe, M.; McNeish, I.; Gabra, H.; Tookman, L.; Rahman, N.; George, A.; Krell, J. Oncologist-led BRCA ‘mainstreaming’ in the ovarian cancer clinic: A study of 255 patients and its impact on their management. Sci. Rep. 2020, 10, 3390. [Google Scholar] [CrossRef] [Green Version]
  10. Flaum, N.; Morgan, R.D.; Burghel, G.J.; Bulman, M.; Clamp, A.R.; Hasan, J.; Mitchell, C.L.; Badea, D.; Moon, S.; Hogg, M.; et al. Mainstreaming germline BRCA1/2 testing in non-mucinous epithelial ovarian cancer in the North West of England. Eur. J. Hum. Genet. 2020, 28, 1541–1547. [Google Scholar] [CrossRef]
  11. Frugtniet, B.; Morgan, S.; Murray, A.; Palmer-Smith, S.; White, R.; Jones, R.; Hanna, L.; Fuller, C.; Hudson, E.; Mullard, A.; et al. The detection of germline and somatic BRCA1/2 genetic variants through parallel testing of patients with high-grade serous ovarian cancer: A national retrospective audit. BJOG Int. J. Obstet. Gynaecol. 2022, 129, 433–442. [Google Scholar] [CrossRef] [PubMed]
  12. Taylor, A.; Brady, A.F.; Frayling, I.M.; Hanson, H.; Tischkowitz, M.; Turnbull, C.; Side, L.; UK Cancer Genetics Group (UK-CGG). Consensus for genes to be included on cancer panel tests offered by UK genetics services: Guidelines of the UK Cancer Genetics Group. J. Med. Genet. 2018, 55, 372–377. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Mandelker, D.; Donoghue, M.; Talukdar, S.; Bandlamudi, C.; Srinivasan, P.; Vivek, M.; Jezdic, S.; Hanson, H.; Snape, K.; Kulkarni, A.; et al. Germline-focussed analysis of tumour-only sequencing: Recommendations from the ESMO Precision Medicine Working Group. Ann. Oncol. 2019, 30, 1221–1231. [Google Scholar] [CrossRef] [Green Version]
  14. WHO. Female Genital Tumours. WHO Classification of Tumours, 5th ed.International Agency for Research on Cancer: Lyon, France, 2020; Volume 4. [Google Scholar]
  15. Prat, J.; FIGO Committee on Gynecologic Oncology. Staging classification for cancer of the ovary, fallopian tube, and peritoneum. Int. J. Gynaecol. Obstet. 2014, 124, 1–5. [Google Scholar] [CrossRef] [PubMed]
  16. Morgan, R.D.; Burghel, G.J.; Flaum, N.; Bulman, M.; Clamp, A.R.; Hasan, J.; Mitchell, C.L.; Schlecht, H.; Woodward, E.R.; Lalloo, F.I.; et al. Prevalence of germline pathogenic BRCA1/2 variants in sequential epithelial ovarian cancer cases. J. Med. Genet. 2019, 56, 301–307. [Google Scholar] [CrossRef] [Green Version]
  17. Garrett, A.; Durkie, M.; Callaway, A.; Burghel, G.J.; Robinson, R.; Drummond, J.; Torr, B.; Cubuk, C.; Berry, I.R.; Wallace, A.J.; et al. Combining evidence for and against pathogenicity for variants in cancer susceptibility genes: CanVIG-UK consensus recommendations. J. Med. Genet. 2021, 58, 297–304. [Google Scholar] [CrossRef]
  18. Ellison, G.; Huang, S.; Carr, H.; Wallace, A.; Ahdesmaki, M.; Bhaskar, S.; Mills, J. A reliable method for the detection of BRCA1 and BRCA2 mutations in fixed tumour tissue utilizing multiplex PCR-based targeted next generation sequencing. BMC Clin. Pathol. 2015, 15, 5. [Google Scholar] [CrossRef] [Green Version]
  19. Telli, M.L.; Timms, K.M.; Reid, J.; Hennessy, B.; Mills, G.B.; Jensen, K.C.; Szallasi, Z.; Barry, W.T.; Winer, E.P.; Tung, N.M.; et al. Homologous Recombination Deficiency (HRD) Score Predicts Response to Platinum-Containing Neoadjuvant Chemotherapy in Patients with Triple-Negative Breast Cancer. Clin. Cancer Res. 2016, 22, 3764–3773. [Google Scholar] [CrossRef] [Green Version]
  20. Abkevich, V.; Timms, K.M.; Hennessy, B.T.; Potter, J.; Carey, M.S.; Meyer, L.A.; Smith-McCune, K.; Broaddus, R.; Lu, K.H.; Chen, J.; et al. Patterns of genomic loss of heterozygosity predict homologous recombination repair defects in epithelial ovarian cancer. Br. J. Cancer 2012, 107, 1776–1782. [Google Scholar] [CrossRef] [Green Version]
  21. Birkbak, N.J.; Wang, Z.C.; Kim, J.Y.; Eklund, A.C.; Li, Q.; Tian, R.; Bowman-Colin, C.; Li, Y.; Greene-Colozzi, A.; Iglehart, J.D.; et al. Telomeric allelic imbalance indicates defective DNA repair and sensitivity to DNA-damaging agents. Cancer Discov. 2012, 2, 366–375. [Google Scholar] [CrossRef]
  22. Popova, T.; Manie, E.; Rieunier, G.; Caux-Moncoutier, V.; Tirapo, C.; Dubois, T.; Delattre, O.; Sigal-Zafrani, B.; Bollet, M.; Longy, M.; et al. Ploidy and large-scale genomic instability consistently identify basal-like breast carcinomas with BRCA1/2 inactivation. Cancer Res. 2012, 72, 5454–5462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Tan, D.S.; Rothermundt, C.; Thomas, K.; Bancroft, E.; Eeles, R.; Shanley, S.; Ardern-Jones, A.; Norman, A.; Kaye, S.B.; Gore, M.G. “BRCAness” syndrome in ovarian cancer: A case-control study describing the clinical features and outcome of patients with epithelial ovarian cancer associated with BRCA1 and BRCA2 mutations. J. Clin. Oncol. 2008, 26, 5530–5536. [Google Scholar] [CrossRef] [PubMed]
  24. Gelmon, K.A.; Tischkowitz, M.; Mackay, H.; Swenerton, K.; Robidoux, A.; Tonkin, K.; Hirte, A.; Huntsman, D.; Clemons, M.; Gilks, B.; et al. Olaparib in patients with recurrent high-grade serous or poorly differentiated ovarian carcinoma or triple-negative breast cancer: A phase 2, multicentre, open-label, non-randomised study. Lancet Oncol. 2011, 12, 852–861. [Google Scholar] [CrossRef] [PubMed]
  25. Alsop, K.; Fereday, S.; Meldrum, C.; deFazio, A.; Emmanuel, C.; George, J.; Dobrovic, A.; Birrer, M.J.; Webb, P.M.; Stewart, C.; et al. BRCA mutation frequency and patterns of treatment response in BRCA mutation-positive women with ovarian cancer: A report from the Australian Ovarian Cancer Study Group. J. Clin. Oncol. 2012, 30, 2654–2663. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Ledermann, J.; Harter, P.; Gourley, C.; Friedlander, M.; Vergote, I.; Rustin, G.; Scott, C.L.; Meier, W.; Shapira-Frommer, R.; Safra, T.; et al. Olaparib maintenance therapy in patients with platinum-sensitive relapsed serous ovarian cancer: A preplanned retrospective analysis of outcomes by BRCA status in a randomised phase 2 trial. Lancet Oncol. 2014, 15, 852–861. [Google Scholar] [CrossRef]
  27. Norquist, B.M.; Brady, M.F.; Harrell, M.I.; Walsh, T.; Lee, M.K.; Gulsuner, S.; Bernards, S.S.; Casadei, S.; Burger, R.A.; Tewari, K.S.; et al. Mutations in Homologous Recombination Genes and Outcomes in Ovarian Carcinoma Patients in GOG 218: An NRG Oncology/Gynecologic Oncology Group Study. Clin. Cancer Res. 2018, 24, 777–783. [Google Scholar] [CrossRef] [Green Version]
  28. Zhong, Q.; Peng, H.L.; Zhao, X.; Zhang, L.; Hwang, W.T. Effects of BRCA1- and BRCA2-related mutations on ovarian and breast cancer survival: A meta-analysis. Clin. Cancer Res. 2015, 21, 211–220. [Google Scholar] [CrossRef] [Green Version]
  29. Daly, M.B.; Pal, T.; Berry, M.P.; Buys, S.S.; Dickson, P.; Domchek, S.M.; Elkhanany, A.; Friedman, S.; Goggins, M.; Hutton, M.L.; et al. Genetic/Familial High-Risk Assessment: Breast, Ovarian, and Pancreatic, Version 2.2021, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Cancer Netw. 2021, 19, 77–102. [Google Scholar] [CrossRef]
  30. Sessa, C.; Balmana, J.; Bober, S.L.; Cardoso, M.J.; Colombo, N.; Curigliano, G.; Domchek, S.; Evans, D.G.; Fischerova, D.; Harbeck, N.; et al. Risk reduction and screening of cancer in hereditary breast-ovarian cancer syndromes: ESMO Clinical Practice Guideline. Ann. Oncol. 2022; Online ahead of print. [Google Scholar] [CrossRef]
  31. Witjes, V.M.; van Bommel, M.H.D.; Ligtenberg, M.J.L.; Vos, J.R.; Mourits, M.J.E.; Ausems, M.; 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]
  32. Sun, L.; Sobocan, M.; Rodriguez, I.V.; Wei, X.; Kalra, A.; Oxley, S.; Sideris, M.; Morgan, R.D.; Chandrasekaran, D.; Rust, K.; et al. Cost-effectiveness of unselected multigene germline and somatic genetic testing for epithelial ovarian cancer. Int. J. Gynecol. Cancer 2022, 32, A321–A322. [Google Scholar]
  33. Cancer Research, UK. Available online: www.cancerresearch.org.uk (accessed on 3 December 2022).
  34. Kwon, J.S.; Tinker, A.V.; Santos, J.; Compton, K.; Sun, S.; Schrader, K.A.; Karsan, A. Germline testing and somatic tumour testing for BRCA1/2 pathogenic variants in ovarian cancer: What is the optimal sequence testing. JCO Precis Oncol. 2022, 6. [Google Scholar]
  35. Jang, J.; Kim, Y.; Kim, J.-H.; Cho, S.-M.; Lee, K.A. Cost-effectiveness analysis of germline and somatic BRCA testing in patients with advanced ovarian cancer. Ann. Lab. Med. 2023, 43, 73–81. [Google Scholar] [CrossRef] [PubMed]
  36. Hauke, J.; Hahnen, E.; Schneider, S.; Reuss, A.; Richters, L.; Kommoss, S.; Heimbach, A.; Marme, F.; Schmidt, S.; Prieske, K.; et al. Deleterious somatic variants in 473 consecutive individuals with ovarian cancer: Results of the observational AGO-TR1 study (NCT02222883). J. Med. Genet. 2019, 56, 574–580. [Google Scholar] [CrossRef]
  37. Morgan, R.D.; Burghel, G.J.; Flaum, N.; Bulman, M.; Smith, P.; Clamp, A.R.; Hasan, J.; Mitchell, C.L.; Salih, Z.; Woodward, E.R.; et al. BRCA1/2 in non-mucinous epithelial ovarian cancer: Tumour with or without germline testing? Br. J. Cancer 2022, 127, 163–167. [Google Scholar] [CrossRef] [PubMed]
  38. Kwong, A.; Chen, J.; Shin, V.Y.; Ho, J.C.; Law, F.B.; Au, C.H.; Chan, T.-L.; Ma, E.S.K.; Ford, J.M. The importance of analysis of long-range rearrangement of BRCA1 and BRCA2 in genetic diagnosis of familial breast cancer. Cancer Genet. 2015, 208, 448–454. [Google Scholar] [CrossRef]
  39. Ray-Coquard, I.; Pautier, P.; Pignata, S.; Perol, D.; Gonzalez-Martin, A.; Berger, R.; Fujiwara, K.; Vergote, I.; Colombo, N.; Maenpaa, J.; et al. Olaparib plus Bevacizumab as First-Line Maintenance in Ovarian Cancer. N. Engl. J. Med. 2019, 381, 2416–2428. [Google Scholar] [CrossRef]
  40. Mirza, M.R.; Monk, B.J.; Herrstedt, J.; Oza, A.M.; Mahner, S.; Redondo, A.; Fabbro, M.; Ledermann, J.A.; Lorusso, D.; Vergote, I.; et al. Niraparib Maintenance Therapy in Platinum-Sensitive, Recurrent Ovarian Cancer. N. Engl. J. Med. 2016, 375, 2154–2164. [Google Scholar] [CrossRef]
  41. Coleman, R.L.; Oza, A.M.; Lorusso, D.; Aghajanian, C.; Oaknin, A.; Dean, A.; Colombo, N.; Weberpals, J.I.; Clamp, A.; Scambia, G.; et al. Rucaparib maintenance treatment for recurrent ovarian carcinoma after response to platinum therapy (ARIEL3): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2017, 390, 1949–1961. [Google Scholar] [CrossRef] [Green Version]
  42. Gonzalez-Martin, A.; Pothuri, B.; Vergote, I.; DePont Christensen, R.; Graybill, W.; Mirza, M.R.; McCormick, C.; Lorusso, D.; Hoskins, P.; Freyer, G.; et al. Niraparib in Patients with Newly Diagnosed Advanced Ovarian Cancer. N. Engl. J. Med. 2019, 381, 2391–2402. [Google Scholar] [CrossRef] [Green Version]
  43. Coleman, R.L.; Fleming, G.F.; Brady, M.F.; Swisher, E.M.; Steffensen, K.D.; Friedlander, M.; Okamoto, A.; Moore, K.N.; Ben-Baruch, N.E.; Werner, T.L.; et al. Veliparib with First-Line Chemotherapy and as Maintenance Therapy in Ovarian Cancer. N. Engl. J. Med. 2019, 381, 2403–2415. [Google Scholar] [CrossRef]
  44. Monk, B.J.; Parkinson, C.; Lim, M.C.; O’Malley, D.M.; Oaknin, A.; Wilson, M.K.; Coleman, R.L.; Lorusso, D.; Bessette, P.; Ghamande, S.; et al. A Randomized, Phase III Trial to Evaluate Rucaparib Monotherapy as Maintenance Treatment in Patients With Newly Diagnosed Ovarian Cancer (ATHENA-MONO/GOG-3020/ENGOT-ov45). J. Clin. Oncol. 2022, 40, 3952–3964. [Google Scholar] [CrossRef] [PubMed]
  45. Ramus, S.J.; Song, H.; Dicks, E.; Tyrer, J.P.; Rosenthal, A.N.; Intermaggio, M.P.; Fraser, L.; Gentry-Maharaj, A.; Hayward, J.; Philpott, S.; et al. Germline Mutations in the BRIP1, BARD1, PALB2, and NBN Genes in Women With Ovarian Cancer. J. Natl. Cancer Inst. 2015, 107, djv214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Song, H.; Dicks, E.; Ramus, S.J.; Tyrer, J.P.; Intermaggio, M.P.; Hayward, J.; Edlund, C.K.; Conti, D.; Harrington, P.; Fraser, L.; et al. Contribution of Germline Mutations in the RAD51B, RAD51C, and RAD51D Genes to Ovarian Cancer in the Population. J. Clin. Oncol. 2015, 33, 2901–2907. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Kurian, A.W.; Ward, K.C.; Howlader, N.; Deapen, D.; Hamilton, A.S.; Mariotto, A.; Miller, D.; Penberthy, L.S.; Katz, S.J. Genetic Testing and Results in a Population-Based Cohort of Breast Cancer Patients and Ovarian Cancer Patients. J. Clin. Oncol. 2019, 37, 1305–1315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Cancer Genome Atlas Research Network. Integrated genomic analyses of ovarian carcinoma. Nature 2011, 474, 609–615. [Google Scholar] [CrossRef] [Green Version]
  49. 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]
  50. Bernards, S.S.; Pennington, K.P.; Harrell, M.I.; Agnew, K.J.; Garcia, R.L.; Norquist, B.M.; Swisher, E.M. Clinical characteristics and outcomes of patients with BRCA1 or RAD51C methylated versus mutated ovarian carcinoma. Gynecol. Oncol. 2018, 148, 281–285. [Google Scholar] [CrossRef]
  51. 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] [Green Version]
  52. Rafnar, T.; Gudbjartsson, D.F.; Sulem, P.; Jonasdottir, A.; Sigurdsson, A.; Jonasdottir, A.; Besenbacher, S.; Lundin, P.; Stacey, S.N.; Gudmundsson, J.; et al. Mutations in BRIP1 confer high risk of ovarian cancer. Nat. Genet. 2011, 43, 1104–1107. [Google Scholar] [CrossRef]
  53. Hodgson, D.R.; Dougherty, B.A.; Lai, Z.; Fielding, A.; Grinsted, L.; Spencer, S.; O’Connor, M.J.; Ho, T.W.; Robertson, J.D.; Launchbury, J.S.; et al. Candidate biomarkers of PARP inhibitor sensitivity in ovarian cancer beyond the BRCA genes. Br. J. Cancer 2018, 119, 1401–1409. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Mirza, M.R.; Lindahl, G.; Mahner, S.; Redondo, A.; Fabbro, M.; Rimel, B.J.; Herrstedt, J.; Oza, A.M.; Canzier, U.; Berek, J.S.; et al. Ad-hoc analysis of the phase III ENGOT-OV16/NOVA study: Niraparib efficacy in germline BRCA wild-type recurrent ovarian cancer with homologous recombination repair defects. Cancer Res. Commun. 2022, 2, 1462–1470. [Google Scholar] [CrossRef]
  55. O’Malley, D.M.; Oza, A.M.; Lorusso, D.; Aghajanian, C.; Oaknin, A.; Dean, A.; Colombo, N.; Weberpals, J.I.; Clamp, A.R.; Scambia, G.; et al. Clinical and molecular characteristics of ARIEL3 patients who derived exceptional benefit from rucaparib maintenance treatment for high-grade ovarian carcinoma. Gynecol. Oncol. 2022; Online ahead of print. [Google Scholar] [CrossRef] [PubMed]
  56. Swisher, E.M.; Kwan, T.T.; Oza, A.M.; Tinker, A.V.; Ray-Coquard, I.; Oaknin, A.; Coleman, R.L.; Aghajanian, C.; Konecny, G.E.; O’Malley, D.M.; et al. Molecular and clinical determinants of response and resistance to rucaparib for recurrent ovarian cancer treatment in ARIEL2 (Parts 1 and 2). Nat. Commun. 2021, 12, 2487. [Google Scholar] [CrossRef] [PubMed]
  57. Swisher, E.M.; Kristeleit, R.S.; Oza, A.M.; Tinker, A.V.; Ray-Coquard, I.; Oaknin, A.; Coleman, R.L.; Burris, H.A.; Aghajanian, C.; O’Malley, D.M.; et al. Characterization of patients with long-term responses to rucaparib treatment in recurrent ovarian cancer. Gynecol. Oncol. 2021, 163, 490–497. [Google Scholar] [CrossRef]
  58. Pujade-Lauraine, E.; Brown, J.; Barnicle, A.; Rowe, P.; Lao-Sirieix, P.; Criscione, S.; du Bois, A.; Lorusso, D.; Romero, I.; Petru, E.; et al. Homologous recombination repair mutation gene panels (excluding BRCA) are not predictive of maintenance olaparib plus bevacizumab efficacy in the first-line PAOLA-1/ENGOT-ov25 trial. Gynecol. Oncol. 2021, 162, S26–S27. [Google Scholar] [CrossRef]
Figure 1. Percentage ratio of germline versus somatic BRCA1/2 pathogenic variants according to age group. Key: BRCAm, BRCA1/2 pathogenic variant.
Figure 1. Percentage ratio of germline versus somatic BRCA1/2 pathogenic variants according to age group. Key: BRCAm, BRCA1/2 pathogenic variant.
Cancers 15 00730 g001
Figure 2. Percentage ratio of GIS-positive versus GIS-negative tumours according to age group.
Figure 2. Percentage ratio of GIS-positive versus GIS-negative tumours according to age group.
Cancers 15 00730 g002
Figure 3. Distribution of age in GIS-positive and GIS-negative tumours. Key: each dot represents the age of an individual patient; grey bar represents mean average.
Figure 3. Distribution of age in GIS-positive and GIS-negative tumours. Key: each dot represents the age of an individual patient; grey bar represents mean average.
Cancers 15 00730 g003
Table 1. Demographic data for patients that underwent BRCA1/2 testing. Data are presented as mean (range) or number (percentage; the denominator being the ‘Total number of patients tested’). Key: BRCAm, BRCA1/2 pathogenic variant; BRCAwt, BRCA1/2 wild type; NOS, not otherwise specified.
Table 1. Demographic data for patients that underwent BRCA1/2 testing. Data are presented as mean (range) or number (percentage; the denominator being the ‘Total number of patients tested’). Key: BRCAm, BRCA1/2 pathogenic variant; BRCAwt, BRCA1/2 wild type; NOS, not otherwise specified.
DemographicNumber of Patients TestedBRCAmBRCAwt
GermlineSomatic
Age at diagnosis—years64 (20–92)58 (35–80)67 (40–92)65 (20–89)
Histology
High-grade serous623 (89)6149513
High-grade endometrioid34 (5)2230
Clear cell32 (5)4325
Adenocarcinoma, NOS11 (2)803
Mixed2 (<1)002
Total70275 (11)54 (8)573
Table 2. Germline and somatic BRCA1/2 pathogenic variants categorized by age group. Data are presented as number (percentage; the denominator is the ‘Number of patients tested’ in each age group). Key: BRCAm, BRCA1/2 pathogenic variant.
Table 2. Germline and somatic BRCA1/2 pathogenic variants categorized by age group. Data are presented as number (percentage; the denominator is the ‘Number of patients tested’ in each age group). Key: BRCAm, BRCA1/2 pathogenic variant.
Age at Diagnosis—YearsNumber of Patients TestedGermline MutationsSomatic Mutations% Total Germline to Somatic BRCAm Ratio
BRCA1BRCA2TotalBRCA1BRCA2Total
<507512214 (19)415 (7)19:7
50–59168181634 (20)7310 (6)10:3
60–6920751419 (9)9615 (7)9:7
70–79204167 (3)7815 (7)3:7
≥8048011 (2)369 (19)1:9
Total702363975 (11)302454 (8)11:8
Table 3. Demographic data for patients that were tested using Myriad’s myChoice® CDx. Data are presented as mean (range) or number (percentage; the denominator being the ‘Total’). Key: BRCAm, BRCA1/2 pathogenic variant; BRCAwt, BRCA1/2 wild type; GIS-, GIS-negative; GIS+, GIS-positive; NOS, not otherwise specified. The four BRCAm/GIS- tumors included germline BRCA2 Exon 14-16 deletion (GIS 36), somatic BRCA1 Exon 13-24 deletion (GIS 34), somatic BRCA2 c.9097dup (GIS 5) and somatic BRCA2 c.3760G>T (GIS 24).
Table 3. Demographic data for patients that were tested using Myriad’s myChoice® CDx. Data are presented as mean (range) or number (percentage; the denominator being the ‘Total’). Key: BRCAm, BRCA1/2 pathogenic variant; BRCAwt, BRCA1/2 wild type; GIS-, GIS-negative; GIS+, GIS-positive; NOS, not otherwise specified. The four BRCAm/GIS- tumors included germline BRCA2 Exon 14-16 deletion (GIS 36), somatic BRCA1 Exon 13-24 deletion (GIS 34), somatic BRCA2 c.9097dup (GIS 5) and somatic BRCA2 c.3760G>T (GIS 24).
DemographicNumber of
Patients Tested
HRD-Negative TumoursHRD-Positive Tumours
BRCAm/GIS+BRCAm/GIS-BRCAwt/GIS+Total
Age at Diagnosis—Years65 (30–89)66 (30–88)63 (41–88)64 (53–75)63 (34–89)63 (34–89)
Histology
High-grade serous292 (84)18342265109
High-grade endometrioid28 (8)1802810
Clear cell22 (6)135049
Adenocarcinoma, NOS2 (<1)01012
Mixed2 (<1)20000
Total34621648478130
Table 4. Genomic instability score status categorized by age group. Data are presented as number (percentage; the denominator is the ‘Number of patients tested’ in each age group). Key: BRCAm, BRCA1/2 pathogenic variant; BRCAwt, BRCA1/2 wild type.
Table 4. Genomic instability score status categorized by age group. Data are presented as number (percentage; the denominator is the ‘Number of patients tested’ in each age group). Key: BRCAm, BRCA1/2 pathogenic variant; BRCAwt, BRCA1/2 wild type.
Age at Diagnosis—YearsNumber of
Patients Tested
GIS-PositiveGIS-Negative% Total GIS-Positive to GIS-Negative Ratio
BRCAmBRCAwtTotalBRCAmBRCAwtTotal
<50232911 (48)01212 (52)12:13
50–5984202040 (48)24244 (52)12:13
60–6998152439 (40)05959 (60)2:3
70–7912882331 (24)29597 (76)6:19
≥8013325 (38)088 (62)19:31
Total3464878126 (36)4216220 (64)9:16
Table 5. Homologous recombination repair genes categorized by genomic instability score status. Data are presented as number (percentage; the denominator being the ‘Total BRCAwt’). Key: a BRCA2 Exon 14-16 deletion (GIS 36); b BRCA1 Exon 13-24 deletion (GIS 34); c BRCA2 c.9097dup (GIS 5) and BRCA2 c.3760G>T (GIS 24); BRCAm, BRCA1/2 pathogenic variant; BRCAwt, BRCA1/2 wild type; HRRm, homologous recombination repair pathogenic variant.
Table 5. Homologous recombination repair genes categorized by genomic instability score status. Data are presented as number (percentage; the denominator being the ‘Total BRCAwt’). Key: a BRCA2 Exon 14-16 deletion (GIS 36); b BRCA1 Exon 13-24 deletion (GIS 34); c BRCA2 c.9097dup (GIS 5) and BRCA2 c.3760G>T (GIS 24); BRCAm, BRCA1/2 pathogenic variant; BRCAwt, BRCA1/2 wild type; HRRm, homologous recombination repair pathogenic variant.
GIS StatusNumber of
Patients Tested
GermlineSomaticBRCAwtGermline Non-BRCA1/2 HRRm
BRCA1BRCA2BRCA1BRCA2BRIP1PALB2RAD51CRAD51D
GIS-positive861416129352000
GIS-negative8801 a1 b2 c842000
Total174141713111194 (3)000
Table 6. Demographic data for patients diagnosed with a germline BRIP1 pathogenic variant. Key: BRCAm, BRCA1/2 pathogenic variant; FDR, first-degree relative; GIS, genomic instability score; HGSOC, high-grade serous ovarian cancer; SDR, second-degree relative; WT, wild type.
Table 6. Demographic data for patients diagnosed with a germline BRIP1 pathogenic variant. Key: BRCAm, BRCA1/2 pathogenic variant; FDR, first-degree relative; GIS, genomic instability score; HGSOC, high-grade serous ovarian cancer; SDR, second-degree relative; WT, wild type.
BRIP1 VariantAge at Diagnosis—YearsHistologyFIGO
Stage
Family HistoryBRCAmGISHRD status
Nucleotide
Level
Protein
Level
GermlineTumour
c.1888dup(p.Thr630fs)68HGSOCIIIC1 × FDR Breast CancerWTWT40Negative
c.2108delinsTCC(p.Lys703fs)68HGSOCIIIC1 × FDR Ovarian CancerWTWT49Positive
c.2392C>T(p.Arg798Ter)59HGSOCIVB1 × SDR Breast CancerWTWT57Positive
c.2492+2dupp.(?)60HGSOCIVA3 × FDR Breast CancerWTWT41Negative
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

Morgan, R.D.; Burghel, G.J.; Flaum, N.; Bulman, M.; Smith, P.; Clamp, A.R.; Hasan, J.; Mitchell, C.L.; Salih, Z.; Woodward, E.R.; et al. Is Reflex Germline BRCA1/2 Testing Necessary in Women Diagnosed with Non-Mucinous High-Grade Epithelial Ovarian Cancer Aged 80 Years or Older? Cancers 2023, 15, 730. https://doi.org/10.3390/cancers15030730

AMA Style

Morgan RD, Burghel GJ, Flaum N, Bulman M, Smith P, Clamp AR, Hasan J, Mitchell CL, Salih Z, Woodward ER, et al. Is Reflex Germline BRCA1/2 Testing Necessary in Women Diagnosed with Non-Mucinous High-Grade Epithelial Ovarian Cancer Aged 80 Years or Older? Cancers. 2023; 15(3):730. https://doi.org/10.3390/cancers15030730

Chicago/Turabian Style

Morgan, Robert D., George J. Burghel, Nicola Flaum, Michael Bulman, Philip Smith, Andrew R. Clamp, Jurjees Hasan, Claire L. Mitchell, Zena Salih, Emma R. Woodward, and et al. 2023. "Is Reflex Germline BRCA1/2 Testing Necessary in Women Diagnosed with Non-Mucinous High-Grade Epithelial Ovarian Cancer Aged 80 Years or Older?" Cancers 15, no. 3: 730. https://doi.org/10.3390/cancers15030730

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