J Gynecol Oncol. 2017 Jul;28(4):e51. English.
Published online May 19, 2017.
Copyright © 2017. Asian Society of Gynecologic Oncology, Korean Society of Gynecologic Oncology
Editorial

Role and clinical application of next-generation sequencing (NGS) for ovarian cancer

Myong Cheol Lim,1,2 and Leslie M. Randall2
    • 1Cancer Healthcare Research Branch, Center for Uterine Cancer and Center for Clinical Trials, Research Institute and Hospital, Department of Cancer Control and Population Health, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea.
    • 2Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Irvine Medical Center, University of California, Orange, CA, USA.
Received May 11, 2017; Accepted May 15, 2017.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (https://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

The inability to repair double stranded DNA breaks, or homologous recombination deficiency (HRD), is the first clinically actionable molecular aberration to be identified in gynecologic cancers. Germline mutations in BRCA1 and BRCA2 were the first biomarkers of this phenotype to be identified. Further investigation of these mutations revealed that they are key biomarkers for prevention in affected blood relatives, improved prognosis, response to platinum-based therapy, and, of more recent importance, maximum benefit from poly (adenosine diphosphate [ADP]-ribose) polymerase (PARP) inhibitor (PARPi) treatment [1]. Mutations in additional genes including RAD51C, RAD51D, BRIP1, ATM, CHEK2, BARD1, MRE11, NBS1, PALB2, and NBN are now also implicated in this pathway and are rationale predictors of PARPi sensitivity [2, 3, 4], as loss of their protein products impair the homologous recombination (HR) pathway's task to maintain genomic stability [5]. The Cancer Genome Atlas (TCGA) project in the USA reported up to alterations in the HR pathway in 51% of high grade serous ovarian cancers, with 16% of tumors harboring germline BRCA1 or BRCA2 mutations, 9% with somatic mutations, and 11% with epigenetic silencing of BRCA1 by way of methylation [6].

Germline BRCA mutation (gBRCAmut) has been established as a strong predictor of response rate, progression-free survival (PFS), and overall survival (OS) with PARPi. Olaparib (Lynparza™; AstraZeneca, Wilmington, DE, USA) first gained approval in Europe for maintenance following complete response to third line or greater platinum-based chemotherapy [7] and in the USA for 4th treatment or greater [8], both indications limited to gBRCAmut carriers. However, somatic mutations were also postulated to predict PARPi sensitivity [9], and in fact, rucaparib broadened the indications for PARPi when it was approved in the USA for 3rd line or greater treatment in women with either germline or somatic BRCA mutations [10]. Somatic mutations in this study, rucaparib in relapsed, platinum-sensitive high-grade ovarian carcinoma (ARIEL2 Part 1), were assessed by loss of heterozygosity (LOH). PFS was significantly longer in the gBRCAmut (hazard ratio [HR]=0.27; 95% confidence interval [CI]=0.16–0.44; p<0.001) and LOH high (HR=0.62; 95% CI=0.42–0.90; p=0.011) subgroups compared with the LOH low subgroup. This study also led to the approval of FoundationFocus™ CDx BRCA test (Foundation Medicine Inc., Cambridge, MA, USA), a tissue-based next-generation sequencing (NGS) tool to detect alteration in BRCA1 and BRCA2 genes in formalin-fixed paraffin-embedded (FFPE) ovarian tumor tissues as a companion diagnostic for rucaparib. Simplified sample collection using archived specimen 1 mm3 FFPE ovarian cancer tumor tissue specimens with >20% of malignant origin, 14-day turnaround time, and multiple cancer gene evaluations at a time is strengths of NGS. Exploratory analyses in ARIEL2 documented significant tumor responses in women with mutations in non-BRCA HR genes ATM, NBN, RAD51C, or RAD51D plus those with methylation of BRCA1 or RAD51C [11], further supporting their classification as actionable mechanisms of HRD and their inclusion in comprehensive HRD assessment.

Given these advances, and the likelihood that an additional 30% of serous and an unknown proportion of non-serous histologies harbor non-gBRCAm-associated HRD, it is now imperative to discover what other genomic alterations might predict PARPi-sensitive biology. In addition, it will be important to know if these alterations vary by ethnicity.

In this issue of Journal of Gynecologic Oncology, Zhao et al. [12] report the results of their NGS analysis of 50 matched serum and samples collected from women undergoing ovarian cancer surgery in China. These specimens were tested for germline and somatic mutations in a broad panel of HRD genes including BRCA1/2, Fanconi anemia pathway (FANCx and PALB2), plus ATM, ATR, CHEK1, CHEK2, BARD1, BRIP1, FAM175A, MRE11A, and NBN. Mutations were classified as deleterious or not based on the publicly available ClinVar database (National Library of Medicine, Bethesda, MD, USA). The prevalence of germline and somatic mutations in this sample was 18 (36%) and 5 (10%), respectively. Interestingly, BRCA2 was the most common germline mutation and RAD50 was the most frequent somatic mutation. Additional germline mutations included BRCA1 (8%), ATR (6%), RAD52 (4%), RAD54B (4%), CHEK2 (2%), and RAD50 (2%) and other somatic mutations were found in RAD50 (4%), RAD54B (2%), BRCA2 (2%), and BRCA1 (2%). This is in contrast to Western women where in 360 women from a single institution, deleterious BRCA1 (13.4%), BRCA2 (5.2%), RAD51D (1.1%), BRIP1 (1.1%), RAD51C (0.8%), CHEK2 (0.8%), PALB2 (0.5%), FAM175A (0.5%), BARD1 (0.5%), NBN (0.3%), and CHEK1 (0.3%) mutations were identified in the germline and BRCA1 (5.2%), BRCA2 (1.6%), ATM (0.8%), CHEK2 (0.8%), BRIP1 (0.5%), MRE11A (0.3%), and RAD51C (0.3%) mutations were identified in tumors. In a second analysis of BRCA1 and BRCA2 only mutations were particularly common (23%) in high-grade serous cancers. In 28 patients with available germline DNA, 9 (42.9%) of 21 and 2 (28.6%) of 7 BRCA1 and BRCA2 mutations were demonstrated to be somatic, respectively.

The current study is consistent with prior reports in that germline mutations are more common than somatic ones. However, at least in this sample, the higher incidence of BRCA2 mutation (18%) than BRCA1 mutation (10%) in Chinese women is quite different pattern compared to that in Western women who exhibit a higher incidence of BRCA1 (18.6%) than BRCA2 (6.8%) [3, 12]. This pattern in Chinese women is consistent with the previous results in Asian women from the Japanese women with ovarian, fallopian tubal, or peritoneal cancer (5.3% BRCA1 and 7.4% BRCA2) [13]. In Korean women with these cancers, BRCA1 mutations are more frequently identified than BRCA2 mutations [14, 15, 16]. Other HRD genes remain lower in prevalence but are consistently identified across studies in which they are probed. Diversity in mutational prevalence according to races or regions has been shown in founder germline mutations, and is important to explore in larger populations with comparable methodologies. These differences could further advance our understanding of ovarian carcinogenesis and treatment approach.

Current Korean Society of Gynecologic Oncology (KSGO) recommendations are to perform genetic testing for germline mutation of BRCA1 and BRCA2 in all women with a pathologically-confirmed ovarian, tubal, or peritoneal cancer [17]. NGS, as performed here, has become the standard sequencing strategy for most laboratory genetic testing due to higher throughput at a lower cost. The limitations of NGS include difficulty detecting certain mutation types (i.e., genomic rearrangements), generation of large amounts of data that require formal bioinformatics analysis, and need for investment in time and funding to validate a new system. Moreover, multigene testing creates complexity in the interpretation of variants of uncertain clinical significance and clinical significance of non-BRCA gene mutations [7, 11]. Finally, public databases, such as ClinVar (National Library of Medicine) employed in this study, have been criticized for being incomplete when compared to a proprietary, commercial database, accounting for 27% of discrepancies in variant classification across the 2 databases [18]. Database sharing across multiple institutions and/or centralization of testing would be long-term solutions to this problem.

Meanwhile, tissue-based NGS is easy and do not needs an additional invasive procedure compared to blood-based germline test. And more comprehensive genetic test from non-BRCA1/2 HR gene and somatic mutation could be achieved. Given the importance of HRD in ovarian cancer, we must continue in our efforts to understand the genetic background and share the individualized treatment outcome based on NGS test in the management of ovarian cancer. Future directions of HRD assessment include non-genomic tissue-based testing that is more phenotypic in nature such as LOH, telomeric allelic imbalance, and large-scale state transitions [19]. This is commercially available as myChoice® HRD test (Myriad Genetics, Inc., Salt Lake City, UT, USA) and is in development as a companion diagnostic to niraparib, the newest of the PARPi's to be approved in the US for maintenance in non-selected women with a complete or partial response to platinum-based chemotherapy [20].

Notes

Conflict of Interest:No potential conflict of interest relevant to this article was reported.

References

    1. Randall LM, Pothuri B. The genetic prediction of risk for gynecologic cancers. Gynecol Oncol 2016;141:10–16.
    1. Pennington KP, Swisher EM. Hereditary ovarian cancer: beyond the usual suspects. Gynecol Oncol 2012;124:347–353.
    1. Pennington KP, Walsh T, Harrell MI, Lee MK, Pennil CC, Rendi MH, 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.
    1. Walsh T, Casadei S, Lee MK, Pennil CC, Nord AS, Thornton AM, et al. Mutations in 12 genes for inherited ovarian, fallopian tube, and peritoneal carcinoma identified by massively parallel sequencing. Proc Natl Acad Sci USA 2011;108:18032–18037.
    1. Toss A, Tomasello C, Razzaboni E, Contu G, Grandi G, Cagnacci A, et al. Hereditary ovarian cancer: not only BRCA 1 and 2 genes. Biomed Res Int 2015;2015:341723.
    1. Cancer Genome Atlas Research NetworkIntegrated genomic analyses of ovarian carcinoma. Nature 2011;474:609–615.
    1. Ledermann JA, Harter P, Gourley C, Friedlander M, Vergote I, Rustin G, et al. Overall survival in patients with platinum-sensitive recurrent serous ovarian cancer receiving olaparib maintenance monotherapy: an updated analysis from a randomised, placebo-controlled, double-blind, phase 2 trial. Lancet Oncol 2016;17:1579–1589.
    1. Kaufman B, Shapira-Frommer R, Schmutzler RK, Audeh MW, Friedlander M, Balmaña J, et al. Olaparib monotherapy in patients with advanced cancer and a germline BRCA1/2 mutation. J Clin Oncol 2015;33:244–250.
    1. Hennessy BT, Timms KM, Carey MS, Gutin A, Meyer LA, Flake DD 2nd, et al. Somatic mutations in BRCA1 and BRCA2 could expand the number of patients that benefit from poly (ADP ribose) polymerase inhibitors in ovarian cancer. J Clin Oncol 2010;28:3570–3576.
    1. Swisher EM, Lin KK, Oza AM, Scott CL, Giordano H, Sun J, et al. Rucaparib in relapsed, platinum-sensitive high-grade ovarian carcinoma (ARIEL2 Part 1): an international, multicentre, open-label, phase 2 trial. Lancet Oncol 2017;18:75–87.
    1. Swisher EM, Harrell MI, Lin K, Coleman RL, Konecny GE, Tinker AV, et al. BRCA1 and RAD51C promoter hypermethylation confer sensitivity to the PARP inhibitor rucaparib in patients with relapsed, platinum-sensitive ovarian carcinoma in ARIEL2 Part 1; Presented at the 48th Annual Meeting of the Society of Gynecologic Oncology; 2017 Mar 12–15; National Harbor, MD. Chicago, IL: Society of Gynecologic Oncology; 2017.
    1. Zhao Q, Yang J, Li L, Cao D, Yu M, Shen K, et al. Germline and somatic mutations in homologous recombination genes among Chinese ovarian cancer patients detected using next-generation sequencing. J Gynecol Oncol 2017;28:e39
    1. Sakamoto I, Hirotsu Y, Nakagomi H, Ouchi H, Ikegami A, Teramoto K, et al. BRCA1 and BRCA2 mutations in Japanese patients with ovarian, fallopian tube, and primary peritoneal cancer. Cancer 2016;122:84–90.
    1. Choi MC, Heo JH, Jang JH, Jung SG, Park H, Joo WD, et al. Germline mutations of BRCA1 and BRCA2 in Korean ovarian cancer patients: finding founder mutations. Int J Gynecol Cancer 2015;25:1386–1391.
    1. Lim MC, Kang S, Seo SS, Kong SY, Lee BY, Lee SK, et al. BRCA1 and BRCA2 germline mutations in Korean ovarian cancer patients. J Cancer Res Clin Oncol 2009;135:1593–1599.
    1. Eoh KJ, Park HS, Park JS, Lee ST, Han J, Lee JY, et al. Comparison of clinical outcomes of BRCA1/2 pathologic mutation, variants of unknown significance, or wild type epithelial ovarian cancer patients. Cancer Res Treat 2017;49:408–415.
    1. Choi MC, Lim MC, Suh DH, Song YJ, Kim TJ, Chang SJ, et al. Position statements on genetic test for peritoneal, ovarian, and fallopian tubal cancers: Korean Society of Gynecologic Oncology (KSGO). J Gynecol Oncol 2016;27:e36
    1. Gradishar W, Johnson K, Brown K, Mundt E, Manley S. Clinical variant classification: a comparison of public databases and a commercial testing laboratory. Oncologist. 2017
      Forthcoming.
    1. Haluska P, Timms KM, AlHilli M, Wang Y, Hartman AM, Jones J, et al. Homologous recombination deficiency (HRD) score and niraparib efficacy in high grade ovarian cancer. Eur J Cancer 2014;50 Suppl 6:72–73.
    1. Mirza MR, Monk BJ, Herrstedt J, Oza AM, Mahner S, Redondo A, et al. Niraparib maintenance therapy in platinum-sensitive, recurrent ovarian cancer. N Engl J Med 2016;375:2154–2164.

Metrics
Share
PERMALINK