Next Article in Journal
The Singular Epidemiology of Plasmacytoma and Multiple Myeloma in French Guiana
Previous Article in Journal
Oral Problems in Oncology Patients Undergoing Chemotherapy for Solid Tumors: A Prospective Observational Study
Previous Article in Special Issue
Satisfaction with Long-Term Aesthetic and 10 Years Oncologic Outcome following Risk-Reducing Mastectomy and Implant-Based Breast Reconstruction with or without Nipple Preservation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Real-World Concordance between Germline and Tumour BRCA1/2 Status in Epithelial Ovarian Cancer

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, School of Medical Sciences, University of Manchester, Manchester M13 9PL, UK
3
North West Genomic Laboratory Hub, Manchester University NHS Foundation Trust, Oxford Road, Manchester M13 9WL, UK
4
Division of Evolution, Infection and Genomics, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester M13 9PL, UK
5
Department of Gynaecological Pathology, Manchester University NHS Foundation Trust, Oxford Road, Manchester M13 9WL, UK
6
Department of Pathology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester M20 4BX, UK
7
Department of Clinical Genetics, Manchester University NHS Foundation Trust, Oxford Road, Manchester M13 9WL, UK
*
Authors to whom correspondence should be addressed.
Cancers 2024, 16(1), 177; https://doi.org/10.3390/cancers16010177
Submission received: 24 November 2023 / Revised: 22 December 2023 / Accepted: 26 December 2023 / Published: 29 December 2023
(This article belongs to the Special Issue Advances in Inherited Breast and Ovarian Cancer and Its Imaging)

Abstract

:

Simple Summary

Approximately 10–15% of patients with epithelial ovarian cancer have an inherited (germline) BRCA1 or BRCA2 mutation. Following a diagnosis of epithelial ovarian cancer, patients are routinely tested for germline and/or somatic (tumour) BRCA1/2 mutations. Our study shows that if germline BRCA1/2 testing is only performed for patients with a positive tumour BRCA1/2 test result, and tumour testing is performed using Myriad’s myChoice® companion diagnostic, a proportion of germline BRCA1/2 large rearrangements could be missed. If paired germline-tumour DNA testing is not possible for all patients, our data shows that it would be appropriate to test all patients with epithelial ovarian cancer aged < 79 years old for germline BRCA1/2 mutations, regardless of the tumour BRCA1/2 result, whilst only needing to test patients aged ≥ 80 years old for a germline BRCA1/2 mutation if they have a positive tumour BRCA1/2 result.

Abstract

Patients diagnosed with epithelial ovarian cancer may undergo reflex tumour BRCA1/2 testing followed by germline BRCA1/2 testing in patients with a positive tumour test result. This testing model relies on tumour BRCA1/2 tests being able to detect all types of pathogenic variant. We analysed germline and tumour BRCA1/2 test results from patients treated for epithelial ovarian cancer at our specialist oncological referral centre. Tumour BRCA1/2 testing was performed using the next-generation sequencing (NGS)-based myChoice® companion diagnostic (CDx; Myriad Genetics, Inc.). Germline BRCA1/2 testing was performed in the North West Genomic Laboratory Hub using NGS and multiplex ligation-dependent probe amplification. Between 11 April 2021 and 11 October 2023, 382 patients were successfully tested for tumour BRCA1 and BRCA2 variants. Of these, 367 (96.1%) patients were tested for germline BRCA1/2 variants. In those patients who underwent tumour and germline testing, 15.3% (56/367) had a BRCA1/2 pathogenic variant (36 germline and 20 somatic). All germline BRCA1/2 pathogenic small sequencing variants were detected in tumour DNA. By contrast, 3 out of 8 germline BRCA1/2 pathogenic large rearrangements were not reported in tumour DNA. The overall concordance of germline BRCA1/2 pathogenic variants detected in germline and tumour DNA was clinically acceptable at 91.7% (33/36). The myChoice® CDx was able to detect most germline BRCA1/2 pathogenic variants in tumour DNA, although a proportion of pathogenic large rearrangements were not reported. If Myriad’s myChoice® CDx is used for tumour BRCA1/2 testing, our data supports a testing strategy of germline and tumour BRCA1/2 testing in all patients diagnosed with epithelial ovarian cancer aged < 79 years old, with germline BRCA1/2 testing only necessary for patients aged ≥ 80 years old with a tumour BRCA1/2 pathogenic variant.

1. Introduction

Ovarian cancer is the most lethal gynaecological cancer, with more than 200,000 deaths attributed to the disease each year worldwide [1]. The high mortality rate associated with ovarian cancer occurs largely because, at the time of diagnosis, ovarian cancer cells have metastasized to the peritoneum, meaning curing it is highly unlikely [2]. Indeed, the five-year overall survival for advanced-stage epithelial ovarian cancer is poor at around 30% to 35% [3]. Significant improvements in our understanding of the biology of epithelial ovarian cancer has led to new targeted therapies and subsequent improvement in survival outcomes. This is best exemplified by the synthetic lethal use of poly (ADP-ribose) polymerase-1/2 inhibitors (PARPi) to treat high-grade serous carcinomas that harbour a BRCA1 or BRCA2 pathogenic/likely pathogenic variant [4]. Registration trials for maintenance PARPi therapies in newly diagnosed or relapsed platinum-sensitive BRCA1/2-mutant high-grade serous or endometrioid carcinoma have reported some of the most impressive hazard ratios for survival outcomes seen in oncology [5,6,7,8,9,10,11].
To identify patients eligible for first line PARPi maintenance therapy, reflex tumour testing for BRCA1/2 pathogenic/likely pathogenic variants has been implemented for all patients diagnosed with advanced-stage high-grade epithelial ovarian cancer [12]. Subsequent germline BRCA1/2 testing is then requested for patients with a positive tumour BRCA1/2 test result, thereby determining whether the pathogenic/likely pathogenic variant is constitutional (i.e., germline) or somatically acquired (i.e., tumour-only) [13]. Those patients with a germline BRCA1/2 pathogenic/likely pathogenic variant are referred to clinical genetics to access familial cancer services. This model of selectively testing only certain index cases (i.e., those with a positive tumour test result) for germline BRCA1/2 pathogenic variants relies on tumour tests being able to detect all types of BRCA1/2 pathogenic/likely pathogenic variant, including point mutations, small insertions and deletions, and large rearrangements. If a germline BRCA1/2 pathogenic/likely pathogenic variant is missed, index cases may miss out on maintenance PARPi therapies, as well as breast cancer risk-reduction measures [14]. Moreover, family members will not gain access to cascade testing and breast/ovarian cancer risk-reduction strategies in related, unaffected germline BRCA1/2 heterozygotes.
There is limited real-world data reporting the number of germline BRCA1/2 pathogenic/likely pathogenic variants missed by testing tumour DNA using commercially available clinically validated genetic assays [15,16,17,18,19]. The aim of this study was to report the concordance of BRCA1/2 pathogenic/likely pathogenic variants detected in germline and tumour DNA in a large, real-world cohort of patients treated for epithelial ovarian cancer at our specialist oncological referral centre. Patients underwent local germline BRCA1/2 testing, whilst tumour testing was performed using the myChoice® companion diagnostic (CDx; Myriad Genetics, Inc., Salt Lake City, UT, USA).

2. Materials and Methods

Eligibility criteria included all patients diagnosed with epithelial ovarian, fallopian tube or primary peritoneal (Müllerian type) cancer that were treated at The Christie Hospital (Manchester, UK), and tested for tumour BRCA1/2 pathogenic/likely pathogenic variants using the myChoice® CDx (Myriad Genetics, Inc., Salt Lake City, UT, USA). Index cases of ovarian carcinosarcoma were eligible for inclusion. All patients diagnosed with non-epithelial ovarian cancer were excluded. All FIGO (International Federation of Gynaecology and Obstetrics) stages of disease were eligible for inclusion. Only patients aged ≥18 years at diagnosis were included.
MyChoice® CDx testing was coordinated by the North West Genomic Laboratory Hub (Manchester, UK) and was available from 11 April 2021 onwards. Tumour BRCA1/2 testing was planned for all newly diagnosed patients in line with the national genomic test directory for cancer, specified by NHS England [20]. The methodology used in Myriad’s myChoice® CDx has been reported [21]. The myChoice® CDx is a next-generation-sequencing (NGS) in vitro diagnostic assay that reports the homologous recombination deficiency (HRD) status of formalin-fixed paraffin-embedded (FFPE) tumour tissue based on the tumour BRCA1/2 status and the genomic instability score (GIS) [22]. In this study, we focussed on the tumour BRCA1/2 status only. The myChoice® CDx detects single nucleotide variants, insertions and deletions, and large rearrangements in the protein coding regions and intron/exon boundaries of BRCA1 and BRCA2 [22].
Archival FFPE tumour blocks were sent for HRD testing. The percentage tumour cell count was determined locally by a Histopathologist with a specialty interest in gynaecological pathology. Formalin-fixed paraffin-embedded tissue was assessed for tumour content by assessing the total cellularity and tumour cell content, expressed as a percentage of all viable nucleated cells on a haematoxylin and eosin stained 5 μm thick section.
Germline BRCA1/2 testing was planned for all eligible patients as per the national genomic test directory for rare and inherited disease, specified by NHS England [23]. Germline testing took place locally in the North West Genomic Laboratory Hub (Manchester, UK) using DNA extracted from peripheral lymphocytes. The chemagic Prime™ instrument (Revvity, Inc., Waltham, MA, USA) was used to extract DNA. Enrichment was performed using SureSelect custom designed probes (Agilent Technologies, Inc., Santa Clara, CA, USA) that targeted the coding regions of transcripts (NM_007294.3 [BRCA1] and NM_000059.3 [BRCA2]) including the immediate splice sites +/− 15 base pairs and known intronic pathogenic/likely pathogenic variants in BRCA1 and BRCA2. Next-generation sequencing was performed using the NextSeq 550 System (Illumina, Inc., San Diego, CA, USA). Target coverage was ≥90% of BRCA1 and BRCA2 at a depth of at least 100×. Only pathogenic [class 5] and likely pathogenic [class 4] variants were reported (hereon described together as ‘pathogenic variants’) [24,25]. A local, in-house bioinformatic pipeline reported single nucleotide variants, and small insertions, deletions, and duplications < 40 base pairs in length. Only germline pathogenic variants with a variant allele frequency of ≥5% were reported.
Testing for germline large rearrangements in BRCA1 and BRCA2 was performed using multiplex ligation-dependent probe amplification (MLPA®) [26]. The MLPA® probe kits P002-D1 (BRCA1) and P045-D1 (BRCA2; MRC Holland, Amsterdam, Netherlands) were used to analyse germline DNA. Amplified ligation products were subject to fragment analysis using an ABI 3130xl Genetic Analyser and sized using GeneMapper Software version 6.0 (Thermo Fisher Scientific, Inc., Waltham, MA, USA). Copy number status calling was performed using data exported from GeneMapper using custom-developed spreadsheets that report relative dosage quotient for each probe compared with five reference control samples. All MLPA® tests were performed in duplicate for confirmation of results.
Categorical data were reported as number (percentage). Continuous data were reported as median (range or interquartile range [IQR]). The Mann–Whitney U test was used to measure differences in the median of two groups with statistical significance defined as a p value < 0.05.

3. Results

3.1. Study Group

Between 11 April 2021 and 11 October 2023, 442 tumour samples from 412 patients were tested using Myriad’s myChoice® CDx. The tumour cell content ranged from 5% to 80% (tumour cell content not provided in 61/442 [13.8%] samples). The tumour BRCA1/2 assay failure rate was 10.2% (45/442 tumours). Those samples that failed tumour testing had a tumour cell content of 5% to 65% (tumour cell content not provided in 10/45 samples). Two or more tumour samples were tested from 29 patients due to prior complete assay failure (tumour BRCA1/2 and GIS testing; 23/29 patients) or because the patient was mistakenly tested twice (6/29 patients). Tumour BRCA1/2 testing was successful in 382 patients. Of these, 15 patients (3.9%) did not undergo germline BRCA1/2 testing. The study group comprised 367 patients who successfully underwent both tumour and germline BRCA1/2 testing (Table 1). Of these, most patients had been diagnosed with advanced-stage, high-grade, non-mucinous epithelial ovarian cancer.

3.2. BRCA1/2 Pathogenic Variants

There were 58 BRCA1/2 pathogenic variants detected in 56 patients (prevalence 15.3%; 56/367) (Table 2). Thirty-six patients had a germline BRCA1/2 pathogenic variant (18 BRCA1, 18 BRCA2; prevalence 9.8%), and 20 patients had a somatic BRCA1/2 pathogenic variant (11 BRCA1, 9 BRCA2; prevalence 5.4%). Two patients had two different BRCA1 or BRCA2 pathogenic variants (all somatic). Neither of the samples that harboured two somatic BRCA1/2 pathogenic variants had a reversion mutation (BRCA2:c.3337G>T and BRCA2:c.3342_3348del; BRCA1:c.457_486delinsC and BRCA1:c.524_526delinsTG). There were 51/58 unique BRCA1/2 pathogenic variants (32 germline, 19 somatic). Of the seven BRCA1/2 pathogenic variants detected twice, five were BRCA2 (four germline, one somatic). The somatic pathogenic variant detected twice was BRCA2:c.4688G>A.
The median age at diagnosis in patients with a germline or somatic BRCA1/2 pathogenic variant was 58 years old (IQR: 51–69 years old) and 69 years old (IQR: 57–77 years old), respectively (p = 0.018). BRCA1/2 pathogenic variants were more frequently detected in patients diagnosed with high-grade serous carcinoma (54 versus 2 [non-high-grade serous carcinoma]) and those with advanced-stage disease (8 [FIGO stage I/II] versus 48 [FIGO stage III/IV]).
Most BRCA1/2 pathogenic variants were small sequencing variants (46/58 [79.3%]; 28 germline, 18 somatic) and measured between 1 and 52 base pairs in length. Most small sequencing variants were less than five base pairs long (44/46 [95.7%]). Twelve BRCA1/2 pathogenic variants were large rearrangements (12/58 [20.7%]; eight germline, four somatic).

3.3. Real-World Concordance between Germline and Tumour BRCA1/2 Pathogenic Variants

All patients with a BRCA1/2 pathogenic variant detected in tumour DNA underwent germline BRCA1/2 testing. Of the 36 germline BRCA1/2 pathogenic variants, 33 were detected in tumour DNA. The concordance of germline BRCA1/2 pathogenic variants in germline and tumour DNA was 91.7% (33/36) (Table 3). All small sequencing BRCA1/2 pathogenic variants were detected in germline and tumour DNA. The three germline BRCA1/2 pathogenic variants not detected in tumour DNA were all large rearrangements and included two BRCA1 Exon 13 duplications (n = 2) and a BRCA2 Exon 1–24 deletion (n = 1; Table 4). All germline BRCA1/2 large rearrangements were detected in tumour samples from patients diagnosed with advanced-stage, high-grade serous carcinoma, aged < 79 years (age range: 43 to 77 years old).

4. Discussion

The implementation of mainstream BRCA1/2 testing for patients diagnosed with epithelial ovarian cancer occurred due to the development of PARPi as standard therapy. Tumour and germline BRCA1/2 testing can be performed in parallel (i.e., together) or sequentially (i.e., germline-first then tumour, or tumour-first then germline) [27,28]. In the parallel model of testing, germline (often blood) and tumour DNA are tested for all patients, regardless of the result of each test. In the sequential model of testing, only patients with germline BRCA1/2 wild-type undergo subsequent tumour BRCA1/2 testing to detect additional somatic BRCA1/2 pathogenic variants (germline-first), or alternatively, only patients with a tumour BRCA1/2 pathogenic variant undergo subsequent germline testing to determine whether the variant is constitutional or somatically acquired (tumour-first). The choice of using a parallel versus sequential model of testing is determined by the local treating multi-disciplinary team and is often based on financial costs (including reimbursement agreements) and availability of regional diagnostic services [12]. The national genomic test directory for NHS England allows all patients diagnosed with epithelial ovarian cancer to undergo germline BRCA1/2 testing, and all patients eligible for first-line treatment, who have been diagnosed with high-grade ovarian cancer, to undergo tumour BRCA1/2 testing (as part of ‘tumour HRD testing’) [20,22].
Following validation of Myriad’s myChoice® CDx in the randomised, Phase 3 trial, PAOLA-1 reporting that patients with HRD-positive tumours had longer median progression-free survival and overall survival with first-line bevacizumab and olaparib therapy [10,29]; this commercial assay has been adopted by many cancer centres to detect tumour BRCA1/2 pathogenic variants [30,31]. Our study shows that for cancer centres that use a tumour-first BRCA1/2 testing approach using the myChoice® CDx, around 5–10% of germline BRCA1/2 pathogenic variants may not be reported, all of which are likely to be pathogenic large rearrangements. Our real-world data show a higher rate of unreported germline BRCA1/2 pathogenic variants compared to myChoice® CDx test data from breast and ovarian tumours tested in the randomised, Phase 3 PARPi trials OlympiAD and SOLO2, where the missed rate was much lower at 0.7% and 1.7%, respectively [15,16]. All the missed pathogenic variants in OlympiAD and SOLO2 were pathogenic large rearrangements [15,16]. The higher miss rate in our study is probably due to one of the unreported large rearrangements (BRCA1 Exon 13 duplication) being a founder mutation in the North of England, from where our cohort is derived [32]. Moreover, the size of our study population (n = 56) was lower than that tested in OlympiAD (n = 143) and SOLO2 (n = 241) [15,16].
It is unclear why both BRCA1 Exon 13 duplications and a BRCA2 14–16 deletion were unreported in our study. The three FFPE samples for these patients contained more than 30% tumour content, preferred by Myriad for testing. Moreover, two out of three of the FFPE samples had a GIS reported, suggesting that the quality and quantity of tumour DNA extracted reached the required threshold for full assay completion (the GIS for the two tumours with a BRCA1 Exon 13 duplication were 68 and 70). In addition, a germline BRCA2 Exon 14–16 deletion was reported in tumour DNA from another patient in our study, confirming the ability of the myChoice® CDx to detect this pathogenic large rearrangement. Unfortunately, we were unable to perform repeat HRD testing of other tumour samples from the three patients with these unreported pathogenic variants because the positive germline mutant status had already been reported.
One possible explanation why BRCA1 Exon 13 duplication was not reported in our study is that this large rearrangement may often exist outside the limit of detection of the myChoice® CDx. In the Phase 3 trial, SOLO2, Myriad reported that both patients with a germline BRCA1 Exon 13 duplication were not reported by the myChoice® CDx because “it was not possible to confirm that these large rearrangements were intragenic” [15]. The wider implication of possible under reporting of BRCA1 Exon 13 duplication can be seen in two recent publications in which only one case of this large rearrangement was reported in over 22,000 ovarian cancer tissue samples tested using the myChoice® CDx [33,34]. This unexpectedly low prevalence of BRCA1 Exon 13 duplication contradicts previous prevalence data from North American/European cohorts of patients at risk of hereditary breast and/or ovarian cancer, where this large rearrangement was the fourth most common germline BRCA1 pathogenic variant [35,36,37]. Our group is particularly interested in BRCA1 Exon 13 duplication because it accounts for 3.1% (85/2709) of all germline BRCA1/2 pathogenic variants in our regional dataset (as of 11 October 2023). Cancer centres located in other geographical regions where germline BRCA1 Exon 13 duplication occurs frequently should be aware of the possible under reporting of this large rearrangement by the myChoice® CDx [32]. It may also be useful to repeat our study in a different population where BRCA1 Exon 13 duplications are less common and/or in a population with alternative large BRCA1/2 founder duplications.
Matched tumour-normal BRCA1/2 testing can be costly and logistically challenging [12,38]. However, the implications of missed germline pathogenic variants are significant, especially for unaffected family members in which risk-reduction strategies can prevent breast and/or ovarian cancer from occurring [39]. To limit costs of genetic testing, we have proposed a testing strategy that involves paired germline and tumour BRCA1/2 testing in all patients diagnosed with epithelial ovarian cancer aged < 79 years old, while reserving tumour-first testing for those aged ≥ 80 years old [40]. Our latest study further supports this testing strategy, should the myChoice® CDx be used. The only two BRCA1/2 pathogenic variants detected in patients diagnosed at ≥80 years old were small sequencing variants that were detected in tumour DNA (BRCA1:c.68_69del and BRCA2:c.4478_4481del). No large rearrangements occurred in any patient aged > 77 years old. By adopting our age-defined BRCA1/2 testing strategy, we estimate a 15% reduction in the total number of patients being tested, while maintaining near universal detection of all BRCA1/2 pathogenic variants.
The main limitations with this study are the modest sample size and single-centre status. We also acknowledge the fact that almost 4% (15/382) of the study group did not undergo germline BRCA1/2 testing. A review of the clinical notes of these patients showed that almost all either declined germline testing or had poor prognostic clinical factors and died before seeing an oncologist to consent for germline testing. We also recognize that a minority (12/367) of the study group did not have a diagnosis of high-grade ovarian cancer and were therefore ineligible for tumour HRD testing. Only one patient with non-high-grade ovarian cancer had a tumour BRCA1/2 pathogenic variant. The inclusion of these patients provides a real-world perspective of genetic testing practice at a large tertiary referral centre. It is also important to note that no tumour BRCA1/2 test has been validated to distinguish between germline and somatic pathogenic variants, so all patients with a tumour BRCA1/2 pathogenic variant, irrespective of the tumour test used, should undergo germline testing.

5. Conclusions

We report the concordance between germline and tumour BRCA1/2 pathogenic variants in a large, real-world cohort of patients diagnosed with epithelial ovarian cancer. If the Myriad myChoice® CDx is used for tumour HRD testing, our data support a testing model of germline and tumour BRCA1/2 testing in all patients diagnosed with epithelial ovarian cancer aged < 79 years old, with germline BRCA1/2 testing only necessary for patients aged ≥ 80 years old with a tumour BRCA1/2 pathogenic variant.

Author Contributions

Conceptualization, R.D.M. and D.G.R.E.; methodology, R.D.M. 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. and D.G.R.E.; writing—review and editing, R.D.M., G.J.B., H.S., A.R.C., J.H., C.L.M., Z.S., J.S., S.D., G.C.J., E.R.W. and D.G.R.E. All authors have read and agreed to the published version of the manuscript.

Funding

This research study received no external funding. E.R.W. and D.G.R.E. are supported by the Manchester National Institute for Health Research (NIHR) Manchester Biomedical Research Centre (IS-BRC-1215–20007).

Institutional Review Board Statement

This 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 patients involved in this study.

Data Availability Statement

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

Acknowledgments

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 [41].

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. 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. Lheureux, S.; Gourley, C.; Vergote, I.; Oza, A.M. Epithelial ovarian cancer. Lancet 2019, 393, 1240–1253. [Google Scholar] [CrossRef] [PubMed]
  3. Peres, L.C.; Cushing-Haugen, K.L.; Kobel, M.; Harris, H.R.; Berchuck, A.; Rossing, M.A.; Schildkraut, J.M.; Doherty, J.A. Invasive Epithelial Ovarian Cancer Survival by Histotype and Disease Stage. J. Natl. Cancer Inst. 2019, 111, 60–68. [Google Scholar] [CrossRef]
  4. 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]
  5. Mirza, M.R.; Monk, B.J.; Herrstedt, J.; Oza, A.M.; Mahner, S.; Redondo, A.; Fabbro, M.; Ledermann, J.A.; Loruosso, 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]
  6. Pujade-Lauraine, E.; Ledermann, J.A.; Selle, F.; Gebski, V.; Penson, R.T.; Oza, A.M.; Korach, J.; Huzarski, T.; Poveda, A.; Pignata, S.; et al. Olaparib tablets as maintenance therapy in patients with platinum-sensitive, relapsed ovarian cancer and a BRCA1/2 mutation (SOLO2/ENGOT-Ov21): A double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Oncol. 2017, 18, 1274–1284. [Google Scholar] [CrossRef] [PubMed]
  7. Coleman, R.; 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]
  8. Moore, K.; Colombo, N.; Scambia, G.; Kim, B.G.; Oaknin, A.; Friedlander, M.; Lisyanskaya, A.; Floquet, A.; Leary, A.; Sonke, G.S.; et al. Maintenance Olaparib in Patients with Newly Diagnosed Advanced Ovarian Cancer. N. Engl. J. Med. 2018, 379, 2495–2505. [Google Scholar] [CrossRef]
  9. González-Martín, 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]
  10. Ray-Coquard, I.; Pautier, P.; Pignata, S.; Pérol, D.; González-Martín, A.; Berger, R.; Fujiwara, K.; Vergote, I.; Colombo, N.; Mäenpää, J.; et al. Olaparib plus Bevacizumab as First-Line Maintenance in Ovarian Cancer. N. Engl. J. Med. 2019, 381, 2416–2428. [Google Scholar] [CrossRef]
  11. 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]
  12. Mandelker, D.; Zhang, L. The emerging significance of secondary germline testing in cancer genomics. J. Pathol. 2018, 244, 610–615. [Google Scholar] [CrossRef] [PubMed]
  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] [PubMed]
  14. Sessa, C.; Balmaña, J.; Bober, S.L.; Cardoso, M.J.; Colombo, N.; Curigliano, G.; Domchek, S.M.; 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. 2023, 34, 33–47. [Google Scholar] [CrossRef] [PubMed]
  15. Hodgson, D.R.; Brown, J.S.; Dearden, S.P.; Lai, Z.; Elks, C.E.; Milenkova, T.; Doughtery, B.A.; Lanchbury, J.S.; Perry, M.; Timms, K.M.; et al. Concordance of BRCA mutation detection in tumor versus blood, and frequency of bi-allelic loss of BRCA in tumors from patients in the phase III SOLO2 trial. Gynecol. Oncol. 2021, 163, 563–568. [Google Scholar] [CrossRef] [PubMed]
  16. Hodgson, D.; Lai, Z.; Dearden, S.; Barrett, J.C.; Harrington, E.A.; Timms, K.; Lanchbury, J.; Wu, W.; Allen, A.; Senkus, E.; et al. Analysis of mutation status and homologous recombination deficiency in tumors of patients with germline BRCA1 or BRCA2 mutations and metastatic breast cancer: OlympiAD. Ann. Oncol. 2021, 32, 1582–1589. [Google Scholar] [CrossRef] [PubMed]
  17. Blum, J.L.; Laird, A.D.; Litton, J.K.; Rugo, H.S.; Ettl, J.; Hurvitz, S.A.; Martin, M.; Roché, H.H.; Lee, K.-H.; Goodwin, A.; et al. Determinants of Response to Talazoparib in Patients with HER2-Negative, Germline BRCA1/2-Mutated Breast Cancer. Clin. Cancer Res. 2022, 28, 1383–1390. [Google Scholar] [CrossRef] [PubMed]
  18. Turner, N.C.; Laird, A.D.; Telli, M.L.; Rugo, H.S.; Mailliez, A.; Ettl, J.; Grischke, E.-M.; Mina, L.A.; Balmaña, J.; Fasching, P.A.; et al. Genomic analysis of advanced breast cancer tumors from talazoparib-treated gBRCA1/2mut carriers in the ABRAZO study. NPJ Breast Cancer 2023, 9, 81. [Google Scholar] [CrossRef]
  19. Callens, C.; Vaur, D.; Soubeyran, I.; Rouleau, E.; Just, P.A.; Guillerm, E.; Golmard, L.; Goardon, N.; Sevenet, N.; Cabaret, O.; et al. Concordance Between Tumor and Germline BRCA Status in High-Grade Ovarian Carcinoma Patients in the Phase III PAOLA-1/ENGOT-ov25 Trial. J. Natl. Cancer Inst. 2021, 113, 917–923. [Google Scholar] [CrossRef]
  20. National Genomic Test Directory for Cancer, Version 5.1 (Updated 1 June 2023). Available online: https://www.england.nhs.uk/publication/national-genomic-test-directories (accessed on 1 November 2023).
  21. 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]
  22. MyChoice® CDx Technical Information (PRD-0762 rv 6). Available online: https://myriad.com (accessed on 1 November 2023).
  23. National Genomic Test Directory for Rare and Inherited Disease, Version 7.3 (Updated 20 September 2023). Available online: https://www.england.nhs.uk/publication/national-genomic-test-directories (accessed on 1 November 2023).
  24. Richards, S.; Aziz, N.; Bale, S.; Bick, D.; Das, S.; Gastier-Foster, J.; Grody, W.W.; Hedge, M.; Lyon, E.; Spector, E.; et al. Standards and guidelines for the interpretation of sequence variants: A joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet. Med. 2015, 17, 405–424. [Google Scholar] [CrossRef] [PubMed]
  25. Garrett, A.; Callaway, A.; Durkie, M.; Cubuk, C.; Alikian, M.; Burghel, G.J.; Robinson, R.; Izatt, L.; Talukdar, S.; Side, L.; et al. Cancer Variant Interpretation Group UK (CanVIG-UK): An exemplar national subspecialty multidisciplinary network. J. Med. Genet. 2020, 57, 829–834. [Google Scholar] [CrossRef] [PubMed]
  26. 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] [PubMed]
  27. Kwon, J.S.; Tinker, A.V.; Santos, J.; Compton, K.; Sun, S.; Schrader, K.A.; Karsan, A. Germline Testing and Somatic Tumor Testing for BRCA1/2 Pathogenic Variants in Ovarian Cancer: What Is the Optimal Sequence of Testing? JCO Precis. Oncol. 2022, 6, e2200033. [Google Scholar] [CrossRef]
  28. Witjes, V.M.; Ligtenberg, M.J.L.; Vos, J.R.; Braspenning, J.C.C.; Ausems, M.; Mourits, M.J.E.; de Hullu, J.A.; Adang, E.M.M.; Hoogerbrugge, N. The most efficient and effective BRCA1/2 testing strategy in epithelial ovarian cancer: Tumor-First or Germline-First? Gynecol. Oncol. 2023, 174, 121–128. [Google Scholar] [CrossRef]
  29. Ray-Coquard, I.; Leary, A.; Pignata, S.; Cropet, C.; González-Martín, A.; Marth, C.; Nagao, S.; Vergote, I.; Colombo, N.; Mäenpää, J.; et al. Olaparib plus bevacizumab first-line maintenance in ovarian cancer: Final overall survival results from the PAOLA-1/ENGOT-ov25 trial. Ann. Oncol. 2023, 34, 681–692. [Google Scholar] [CrossRef]
  30. Denkert, C.; Romey, M.; Swedlund, B.; Hattesohl, A.; Teply-Szymanski, J.; Kommoss, S.; Kaiser, K.; Staebler, A.; du Bois, A.; Grass, A.; et al. Homologous Recombination Deficiency as an Ovarian Cancer Biomarker in a Real-World Cohort: Validation of Decentralized Genomic Profiling. J. Mol. Diagn. 2022, 24, 1254–1263. [Google Scholar] [CrossRef]
  31. Heitz, F.; Ataseven, B.; Staniczok, C.; Denkert, C.; Rhiem, K.; Hahnen, E.; Heikaus, S.; Moubarak, M.; Weiz, J.; Dagres, T.; et al. Implementing HRD Testing in Routine Clinical Practice on Patients with Primary High-Grade Advanced Ovarian Cancer. Cancers 2023, 15, 818. [Google Scholar] [CrossRef]
  32. The BRCA1 Exon 13 Duplication Screening Group. The exon 13 duplication in the BRCA1 gene is a founder mutation present in geographically diverse populations. The BRCA1 Exon 13 Duplication Screening Group. Am. J. Hum. Genet. 2000, 67, 207–212. [Google Scholar] [CrossRef]
  33. Morgan, R.D.; Clamp, A.R.; Barnes, B.M.; Timms, K.; Schlecht, H.; Yarram-Smith, L.; Wallis, Y.; Valganon-Petrizan, M.; MacMahon, S.; White, R.; et al. Homologous recombination deficiency in newly diagnosed FIGO stage III/IV high-grade epithelial ovarian cancer: A multi-national observational study. Int. J. Gynecol. Cancer 2023, 33, 1253–1259. [Google Scholar] [CrossRef]
  34. Jones, M.A.; Timms, K.M.; Hatcher, S.; Cogan, E.S.; Comeaux, M.S.; Perry, M.; Morris, B.; Swedlund, B.; Elks, C.E.; Lao-Sirieix, P.; et al. The landscape of BRCA1 and BRCA2 large rearrangements in an international cohort of over 20,000 ovarian tumors identified using next-generation sequencing. Genes Chromosomes Cancer 2023, 62, 589–596. [Google Scholar] [CrossRef] [PubMed]
  35. Hendrickson, B.C.; Judkins, T.; Ward, B.D.; Eliason, K.; Deffenbaugh, A.E.; Burbidge, L.A.; Eliason, K.; Deffenbaugh, A.E.; Burbidge, L.A.; Pyne, K.; et al. Prevalence of five previously reported and recurrent BRCA1 genetic rearrangement mutations in 20,000 patients from hereditary breast/ovarian cancer families. Genes Chromosomes Cancer 2005, 43, 309–313. [Google Scholar] [CrossRef] [PubMed]
  36. Judkin, T.; Rosenthal, E.; Arnell, C.; Burbidge, L.A.; Geary, W.; Barrus, T.; Schoenberger, J.; Trost, J.; Wenstrup, R.J.; Roa, B.B. Clinical significance of large rearrangements in BRCA1 and BRCA2. Cancer 2012, 118, 5210–5216. [Google Scholar] [CrossRef] [PubMed]
  37. Zhang, S.; Royer, R.; Li, S.; McLaughlin, J.R.; Rosen, B.; Risch, H.A.; Fan, I.; Bradley, L.; Shaw, P.A.; Narod, S.A. Frequencies of BRCA1 and BRCA2 mutations among 1,342 unselected patients with invasive ovarian cancer. Gynecol. Oncol. 2011, 121, 353–357. [Google Scholar] [CrossRef] [PubMed]
  38. De Paolis, E.; Marchetti, C.; Concolino, P.; Scambia, G.; Urbani, A.; Fagotti, A.; Minucci, A. A commentary on the discrepancy between blood and tumour BRCA testing: An open question. BJOG 2022, 129, 1422–1426. [Google Scholar] [CrossRef]
  39. Terraf, P.; Pareja, F.; Brown, D.N.; Ceyhan-Birsoy, O.; Misyura, M.; Rana, S.; O’Reilly, E.; Carlo, M.I.; Aghajanian, C.; Liu, Y.; et al. Comprehensive assessment of germline pathogenic variant detection in tumor-only sequencing. Ann. Oncol. 2022, 33, 426–433. [Google Scholar] [CrossRef]
  40. 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. [Google Scholar] [CrossRef]
  41. ISO 15189; Medical Laboratories—Requirements for Quality and Competence. ISO: Geneva, Switzerland, 2022.
Table 1. Demographic data for study group.
Table 1. Demographic data for study group.
CharacteristicStudy Group (367 Patients)
Age at diagnosis—years
Median66
Range24–90
Histology—number (%)
High-grade serous326 (89)
Low-grade serous7 (2)
Endometrioid *10 (3)
Clear cell14 (4)
Mucinous0
Mixed 1 (<1)
Carcinosarcoma8 (2)
Adenocarcinoma, NOS1 (<1)
FIGO stage—number (%)
I10 (3)
II11 (3)
III243 (66)
IV103 (28)
Key: FIGO, International Federation of Gynecology and Obstetrics (2014) staging system; NOS, not otherwise specified. * Five patients diagnosed with grade 1 or 2 (low-grade) endometrioid ovarian adenocarcinoma and five patients diagnosed with grade 3 (high-grade) endometrioid ovarian adenocarcinoma. One patient was diagnosed with clear cell and grade 1 (low-grade) endometrioid ovarian adenocarcinoma.
Table 2. Types of BRCA1/2 pathogenic variants.
Table 2. Types of BRCA1/2 pathogenic variants.
Variant TypeGermline BRCA1/2
(36 Variants)
Somatic BRCA1/2
(22 Variants)
Nucleotide level—number (%) *
Single nucleotide variants9 (25)6 (27)
Small deletions13 (36)7 (32)
Small insertions1 (3)0
Small duplications5 (14)3 (14)
Small indels02 (9)
Large rearrangements8 (22)4 (18)
Protein level—number (%)
Nonsense7 (19)5 (14)
Frameshift19 (53)11 (31)
Missense1 (3)0
Unknown 9 (25)6 (17)
Key: Indels, insertion-deletions; * The denominator is the number of variants (36 germline, 22 somatic); Includes splice site variants and large rearrangements.
Table 3. Germline and tumour BRCA1/2 pathogenic variants.
Table 3. Germline and tumour BRCA1/2 pathogenic variants.
Germline BRCA1/2 Status
MutantWild-Type
Tumour BRCA1/2 statusMutant3320 *
Wild-type3 311
Key: Data presented as number of patients; * Somatic BRCA1/2 pathogenic variants; False negative germline BRCA1/2 result.
Table 4. Pathogenic large rearrangements detected in germline and tumour DNA.
Table 4. Pathogenic large rearrangements detected in germline and tumour DNA.
Large RearrangementGermline DNA—Number
of Pathogenic Variants Detected
Tumour DNA—Number
of Pathogenic Variants Detected
BRCA1
Exon 1–2 deletion11
Exon 3 deletion01 *
Exon 9–12 deletion11
Exon 13–24 deletion01 *
Exon 17 deletion11
Exon 20 deletion11
Exon 13 duplication20
BRCA2
Exon 1–24 deletion01 *
Exon 1–27 deletion01 *
Exon 14–16 deletion21
Key: * Somatic BRCA1/2 pathogenic variants; Discordant germline and tumour BRCA1/2 results.
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.; Schlecht, H.; Clamp, A.R.; Hasan, J.; Mitchell, C.L.; Salih, Z.; Shaw, J.; Desai, S.; Jayson, G.C.; et al. Real-World Concordance between Germline and Tumour BRCA1/2 Status in Epithelial Ovarian Cancer. Cancers 2024, 16, 177. https://doi.org/10.3390/cancers16010177

AMA Style

Morgan RD, Burghel GJ, Schlecht H, Clamp AR, Hasan J, Mitchell CL, Salih Z, Shaw J, Desai S, Jayson GC, et al. Real-World Concordance between Germline and Tumour BRCA1/2 Status in Epithelial Ovarian Cancer. Cancers. 2024; 16(1):177. https://doi.org/10.3390/cancers16010177

Chicago/Turabian Style

Morgan, Robert D., George J. Burghel, Helene Schlecht, Andrew R. Clamp, Jurjees Hasan, Claire L. Mitchell, Zena Salih, Joseph Shaw, Sudha Desai, Gordon C. Jayson, and et al. 2024. "Real-World Concordance between Germline and Tumour BRCA1/2 Status in Epithelial Ovarian Cancer" Cancers 16, no. 1: 177. https://doi.org/10.3390/cancers16010177

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