Molecular and clinical determinants of response and resistance to rucaparib for recurrent ovarian cancer treatment in ARIEL2 (Parts 1 and 2)

Elizabeth M Swisher, Tanya T Kwan, Amit M Oza, Anna V Tinker, Isabelle Ray-Coquard, Ana Oaknin, Robert L Coleman, Carol Aghajanian, Gottfried E Konecny, David M O'Malley, Alexandra Leary, Diane Provencher, Stephen Welch, Lee-May Chen, Andrea E Wahner Hendrickson, Ling Ma, Prafull Ghatage, Rebecca S Kristeleit, Oliver Dorigo, Ashan Musafer, Scott H Kaufmann, Julia A Elvin, Douglas I Lin, Setsuko K Chambers, Erin Dominy, Lan-Thanh Vo, Sandra Goble, Lara Maloney, Heidi Giordano, Thomas Harding, Alexander Dobrovic, Clare L Scott, Kevin K Lin, Iain A McNeish, Elizabeth M Swisher, Tanya T Kwan, Amit M Oza, Anna V Tinker, Isabelle Ray-Coquard, Ana Oaknin, Robert L Coleman, Carol Aghajanian, Gottfried E Konecny, David M O'Malley, Alexandra Leary, Diane Provencher, Stephen Welch, Lee-May Chen, Andrea E Wahner Hendrickson, Ling Ma, Prafull Ghatage, Rebecca S Kristeleit, Oliver Dorigo, Ashan Musafer, Scott H Kaufmann, Julia A Elvin, Douglas I Lin, Setsuko K Chambers, Erin Dominy, Lan-Thanh Vo, Sandra Goble, Lara Maloney, Heidi Giordano, Thomas Harding, Alexander Dobrovic, Clare L Scott, Kevin K Lin, Iain A McNeish

Abstract

ARIEL2 (NCT01891344) is a single-arm, open-label phase 2 study of the PARP inhibitor (PARPi) rucaparib in relapsed high-grade ovarian carcinoma. In this post hoc exploratory biomarker analysis of pre- and post-platinum ARIEL2 samples, RAD51C and RAD51D mutations and high-level BRCA1 promoter methylation predict response to rucaparib, similar to BRCA1/BRCA2 mutations. BRCA1 methylation loss may be a major cross-resistance mechanism to platinum and PARPi. Genomic scars associated with homologous recombination deficiency are irreversible, persisting even as platinum resistance develops, and therefore are predictive of rucaparib response only in platinum-sensitive disease. The RAS, AKT, and cell cycle pathways may be additional modulators of PARPi sensitivity.

Conflict of interest statement

E.M.S. has received funding for clinical trials from Clovis Oncology and TESARO (paid to her institution). T.T.K., E.D., L.-T.V., S.G., L. Maloney, H.G., T.H., and K.K.L. are employees of Clovis Oncology and may own stock or have stock options in that company. A.M.O. has served on advisory boards for Clovis Oncology, Amgen, Immunovaccine, and Verastem; received support for travel or accommodation from AstraZeneca; and received honoraria from WebRx. A.V.T. has served on an advisory board for and received grants from AstraZeneca. I.R.-C. has served on advisory boards for Clovis Oncology, AstraZeneca, Genmab/Seattle Genetics, ImmunoGen, Merck Sharpe Dohme, PharmaMar, Roche, and Tesaro/GlaxoSmithKline and received support for travel or accommodation from AstraZeneca, GlaxoSmithKline, and Roche. A.O. has served on advisory boards for Clovis Oncology, AstraZeneca, ImmunoGen, Genmab/Seattle Genetics, PharmaMar, Roche, and Tesaro and received support for travel or accommodation from AstraZeneca, PharmaMar, Roche, and Tesaro. R.L.C. reports grants from Clovis Oncology, AbbVie, AstraZeneca, Esperance, Janssen, Merck, Millennium, OncoMed, and Roche/Genentech and has served as an advisor to Clovis Oncology, AbbVie, AstraZeneca, Bayer, Esperance, GamaMabs, Genmab, Gradalis, Janssen, Millennium, Merck, OncoMed, Pfizer, Roche/Genentech, and Tesaro. C.A. has served on steering committees for Clovis Oncology and Mateon Therapeutics; served on advisory boards for Clovis Oncology, Bayer, Cerulean Pharma, Tesaro, and VentiRx, and received honoraria from Clovis Oncology, Bayer, Cerulean Pharma, Mateon Therapeutics, Tesaro, and VentiRx. G.E.K. has received research funding (to the University of California outside the scope of this work) from Lilly, Merck, and Pfizer, and received honorarium from Clovis Oncology, AstraZeneca, and Tesaro. D.M.O. has served on advisory boards for Clovis Oncology, AstraZeneca, Gynecologic Oncology Group, Janssen, Myriad, and Tesaro; has served on steering committees for Clovis Oncology, Amgen, and ImmunoGen; has served as a consultant to AbbVie, Ambry, AstraZeneca, Health Analytics, and Tesaro, and his institution has received research support from Clovis Oncology, Agenus, Ajinomoto, Array BioPharma, AstraZeneca, Bristol-Myers Squibb, ERGOMED Clinical Research, Exelixis, Genentech, GlaxoSmithKline, Gynecologic Oncology Group, ImmunoGen, INC Research, inVentiv Health Clinical, Janssen Research and Development, Ludwig Institute for Cancer Research, Novartis Pharmaceuticals, PRA International, Regeneron Pharmaceuticals, Serono, Stemcentrx, Tesaro, and TRACON Pharmaceuticals. A.L. has served on advisory boards for Clovis Oncology, Ability, AstraZeneca, Biocad, Merck Serono, MSD, Pfizer, PharmaMar, Seattle Genetics, and Tesaro PharmaMar; reports institutional research grant support from GamaMabs, Inivata, Merus, and Sanofi, and reports boarding and travel expenses for congress activities from Clovis Oncology, AstraZeneca, Roche, and Tesaro. D.P. has served on advisory boards for AstraZeneca and GlaxoSmithKline, and received support for travel from AstraZeneca. S.W. has served on advisory boards for AstraZeneca and Roche; reports institutional research support from Clovis Oncology, AstraZeneca, Merck, Regeneron, and Tesaro, and has received honoraria from AstraZeneca. P.G. has served on an advisory board for AstraZeneca, GSK, and Merck, and received support for travel expenses for congress activities from AstraZeneca. R.S.K. has served on advisory boards for Clovis Oncology, Roche, and Tesaro. O.D. has served on advisory boards for Clovis Oncology, IMV, Tesaro, and Merck, and has served on the speaker bureau for AstraZeneca and Tesaro. J.A.E. and D.I.L. are employees of Foundation Medicine, Inc., which is a wholly-owned subsidiary of Roche, and may own stock or have stock options in Roche. A.D. has served on advisory boards for AstraZeneca Australia and has received travel support from Bio-Rad. C.L.S. has served on advisory boards for Clovis Oncology, AstraZeneca, Takeda, Roche Australia, MSD, Eisai Co., has received travel support from AstraZeneca, has received in kind research support from Clovis Oncology, Roche, Beigene and Funded research support from Sierra oncology and Eisai Co. I.A.M. has served on advisory boards for Clovis Oncology, AstraZeneca, Roche, Takeda, and Tesaro and receives institutional funding from AstraZeneca. The remaining authors declare no competing interests.

Figures

Fig. 1. PFS by molecular subgroup and…
Fig. 1. PFS by molecular subgroup and platinum-sensitivity status.
a PFS in patients with BRCAmut (blue), BRCAwt/LOH-high (magenta), and BRCAwt/LOH-low (teal) HGOC. b PFS in patients with BRCAmut HGOC that are platinum resistant/refractory (blue) or platinum sensitive (magenta). c PFS in patients with BRCAwt/LOH-high HGOC that are platinum resistant/refractory (blue) or platinum sensitive (magenta) and PFS in patients with BRCAwt/LOH-low HGOC that are platinum resistant/refractory (teal) or platinum sensitive (brown). P values were computed using a Cox proportional hazard model. The interaction between molecular subgroup and platinum status was also tested in the Cox proportional hazard model and found to be significant (P < 0.05). BRCABRCA1 or BRCA2, CI confidence interval, HGOC high-grade ovarian carcinoma, HR hazard ratio, LOH loss of heterozygosity, mut mutated, PFS progression-free survival, wt wild type.
Fig. 2. ORR based on post hoc…
Fig. 2. ORR based on post hoc molecular subgroups and baseline clinical characteristics.
Data are plotted as ORR (dots) with the corresponding two-sided 95% CI (error bars) based on the Clopper–Pearson method. BRCABRCA1 or BRCA2, CI confidence interval, HRR homologous recombination repair, LOH loss of heterozygosity, mut mutated, ORR objective response rate, wt wild type.
Fig. 3. High BRCA1 methylation levels at…
Fig. 3. High BRCA1 methylation levels at screening are associated with better outcomes.
a Kaplan–Meier plot showing PFS in ARIEL2 patients with BRCAwt HGOC having high (magenta), low (blue), or no methylation (unmethylated, teal) in archival biopsy. b Kaplan–Meier plot showing PFS in ARIEL2 patients with BRCAwt HGOC having high (magenta), low (blue), or no methylation (unmethylated, green) in screening biopsy. P values in panels a and b are based on Cox proportional hazard model. c Methylation status at screening as compared to the archival sample, platinum status, and best response to rucaparib of 17 HGOC with high BRCA1 methylation levels in the archival sample and an available matched screening biopsy; numbers indicate number prior lines of chemotherapy treatment; P values are based on a two-sided Fisher’s exact test testing the proportion of patients with platinum status of sensitive and best response of partial response between HGOC that maintained methylation vs decrease or loss of methylation. BRCABRCA1 or BRCA2, CI confidence interval, HGOC high-grade ovarian carcinoma, HR hazard ratio, NA not applicable, PFS progression-free survival, wt wild type.
Fig. 4. Genetic and epigenetic alteration landscape…
Fig. 4. Genetic and epigenetic alteration landscape of HGOC with confirmed best response of CR/PR (left) or PD (right).
Methylation levels shown are at screening. *Short variant include nonsense, missense, frameshift, and splice site alterations. All reported alterations are deleterious or likely deleterious. BRCABRCA1 or BRCA2, CR complete response, HGOC high-grade ovarian carcinoma, HRR homologous recombination repair, LOH loss of heterozygosity, mut mutated, PD progressive disease, PR partial response, RTK receptor tyrosine kinase, wt wild type.
Fig. 5. High BRCA1 methylation at pretreatment…
Fig. 5. High BRCA1 methylation at pretreatment or the presence of a RAD51C/D mutation result in similar PFS as the presence of BRCA mutations.
PFS of patients with BRCAmut HGOCs, excluding cases with reversion mutations (blue), patients with HGOCs harboring a RAD51C/D mutation or high methylation at pretreatment (magenta), and all other patients with known mutation and methylation status, including cases with reversion mutations (teal). P values were computed using a Cox proportional hazard model. BRCA BRCA1 or BRCA2, CI confidence interval, HGOC high-grade ovarian carcinoma, HR hazard ratio, mut mutated, PFS progression-free survival.

References

    1. Konstantinopoulos PA, Ceccaldi R, Shapiro GI, D’Andrea AD. Homologous recombination deficiency: exploiting the fundamental vulnerability of ovarian cancer. Cancer Discov. 2015;5:1137–1154. doi: 10.1158/-15-0714.
    1. Dal Molin GZ, Omatsu K, Sood AK, Coleman RL. Rucaparib in ovarian cancer: an update on safety, efficacy and place in therapy. Ther. Adv. Med. Oncol. 2018;10:1758835918778483. doi: 10.1177/1758835918778483.
    1. Pennington KP, 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. doi: 10.1158/1078-0432.CCR-13-2287.
    1. Rafii S, et al. Baseline clinical predictors of antitumor response to the PARP inhibitor olaparib in germline BRCA1/2 mutated patients with advanced ovarian cancer. Oncotarget. 2017;8:47154–47160. doi: 10.18632/oncotarget.17005.
    1. Ledermann JA, 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. doi: 10.1016/S1470-2045(16)30376-X.
    1. del Campo JM, et al. Niraparib maintenance therapy in patients with recurrent ovarian cancer after a partial response to the last platinum-based chemotherapy in the ENGOT-OV16/NOVA trial. J. Clin. Oncol. 2019;37:2968–2973. doi: 10.1200/JCO.18.02238.
    1. Coleman RL, 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. doi: 10.1016/S0140-6736(17)32440-6.
    1. Coleman RL, et al. Veliparib with first-line chemotherapy and as maintenance therapy in ovarian cancer. N. Engl. J. Med. 2019;381:2403–2415. doi: 10.1056/NEJMoa1909707.
    1. Gonzalez-Martin A, et al. Niraparib in patients with newly diagnosed advanced ovarian cancer. N. Engl. J. Med. 2019;381:2391–2402. doi: 10.1056/NEJMoa1910962.
    1. Ray-Coquard I, et al. Olaparib plus bevacizumab as first-line maintenance in ovarian cancer. N. Engl. J. Med. 2019;381:2416–2428. doi: 10.1056/NEJMoa1911361.
    1. Robson M, et al. Olaparib for metastatic breast cancer in patients with a germline BRCA mutation. N. Engl. J. Med. 2017;377:523–533. doi: 10.1056/NEJMoa1706450.
    1. Litton JK, et al. Talazoparib in patients with advanced breast cancer and a germline BRCA mutation. N. Engl. J. Med. 2018;379:753–763. doi: 10.1056/NEJMoa1802905.
    1. Golan T, et al. Maintenance olaparib for germline BRCA-mutated metastatic pancreatic cancer. N. Engl. J. Med. 2019;381:317–327. doi: 10.1056/NEJMoa1903387.
    1. Mateo J, et al. DNA-repair defects and olaparib in metastatic prostate cancer. N. Engl. J. Med. 2015;373:1697–1708. doi: 10.1056/NEJMoa1506859.
    1. de Bono J, et al. Olaparib for metastatic castration-resistant prostate cancer. N. Engl. J. Med. 2020;382:2091–2102. doi: 10.1056/NEJMoa1911440.
    1. Abida W, et al. Non-BRCA DNA damage repair gene alterations and response to the PARP Inhibitor rucaparib in metastatic castration-resistant prostate cancer: analysis from the phase 2 TRITON2 study. Clin. Cancer Res. 2020;28:2487–2496. doi: 10.1158/1078-0432.CCR-20-0394.
    1. Kondrashova O, et al. Secondary somatic mutations restoring RAD51C and RAD51D associated with acquired resistance to the PARP inhibitor rucaparib in high-grade ovarian carcinoma. Cancer Discov. 2017;7:984–998. doi: 10.1158/-17-0419.
    1. Nikkila J, et al. Heterozygous mutations in PALB2 cause DNA replication and damage response defects. Nat. Commun. 2013;4:2578. doi: 10.1038/ncomms3578.
    1. Kotsopoulos J, et al. Frequency of germline PALB2 mutations among women with epithelial ovarian cancer. Fam. Cancer. 2017;16:29–34. doi: 10.1007/s10689-016-9919-z.
    1. Castaneda A, et al. BRIP1 mutation does not confer sensitivity to PARP inhibition. Gynecol. Oncol. 2019;154:87. doi: 10.1016/j.ygyno.2019.04.205.
    1. Kawazu M, et al. Integrative analysis of genomic alterations in triple-negative breast cancer in association with homologous recombination deficiency. PLoS Genet. 2017;13:e1006853. doi: 10.1371/journal.pgen.1006853.
    1. Cancer Genome Atlas Research Network. Integrated genomic analyses of ovarian carcinoma. Nature. 2011;474:609–615. doi: 10.1038/nature10166.
    1. Ter Brugge, P. et al. Mechanisms of therapy resistance in patient-derived xenograft models of BRCA1-deficient breast cancer. J. Natl. Cancer Inst. 108, 10.1093/jnci/djw148 (2016).
    1. AlHilli MM, et al. In vivo anti-tumor activity of the PARP inhibitor niraparib in homologous recombination deficient and proficient ovarian carcinoma. Gynecol. Oncol. 2016;143:379–388. doi: 10.1016/j.ygyno.2016.08.328.
    1. Kondrashova O, et al. Methylation of all BRCA1 copies predicts response to the PARP inhibitor rucaparib in ovarian carcinoma. Nat. Commun. 2018;9:3970. doi: 10.1038/s41467-018-05564-z.
    1. Tutt A, et al. Carboplatin in BRCA1/2-mutated and triple-negative breast cancer BRCAness subgroups: the TNT Trial. Nat. Med. 2018;24:628–637. doi: 10.1038/s41591-018-0009-7.
    1. Lheureux S, et al. Long-term responders on olaparib maintenance in high-grade serous ovarian cancer: clinical and molecular characterization. Clin. Cancer Res. 2017;23:4086–4094. doi: 10.1158/1078-0432.CCR-16-2615.
    1. Abkevich V, et al. Patterns of genomic loss of heterozygosity predict homologous recombination repair defects in epithelial ovarian cancer. Br. J. Cancer. 2012;107:1776–1782. doi: 10.1038/bjc.2012.451.
    1. Davies H, et al. HRDetect is a predictor of BRCA1 and BRCA2 deficiency based on mutational signatures. Nat. Med. 2017;23:517–525. doi: 10.1038/nm.4292.
    1. Polak P, et al. A mutational signature reveals alterations underlying deficient homologous recombination repair in breast cancer. Nat. Genet. 2017;49:1476–1486. doi: 10.1038/ng.3934.
    1. Nik-Zainal S, et al. Mutational processes molding the genomes of 21 breast cancers. Cell. 2012;149:979–993. doi: 10.1016/j.cell.2012.04.024.
    1. Menghi F, et al. The tandem duplicator phenotype is a prevalent genome-wide cancer configuration driven by distinct gene mutations. Cancer Cell. 2018;34:197–210.e195. doi: 10.1016/j.ccell.2018.06.008.
    1. Nik-Zainal S, et al. Landscape of somatic mutations in 560 breast cancer whole-genome sequences. Nature. 2016;534:47. doi: 10.1038/nature17676.
    1. Macintyre G, et al. Copy number signatures and mutational processes in ovarian carcinoma. Nat. Genet. 2018;50:1262–1270. doi: 10.1038/s41588-018-0179-8.
    1. Telli ML, 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. doi: 10.1158/1078-0432.CCR-15-2477.
    1. Watkins JA, Irshad S, Grigoriadis A, Tutt AN. Genomic scars as biomarkers of homologous recombination deficiency and drug response in breast and ovarian cancers. Breast Cancer Res. 2014;16:211. doi: 10.1186/bcr3670.
    1. Swisher EM, 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. doi: 10.1016/S1470-2045(16)30559-9.
    1. Lin KK, et al. BRCA reversion mutations in circulating tumor DNA predict primary and acquired resistance to the PARP inhibitor rucaparib in high-grade ovarian carcinoma. Cancer Discov. 2019;9:210–219. doi: 10.1158/-18-0715.
    1. Oza AM, et al. Antitumor activity and safety of the PARP inhibitor rucaparib in patients with high-grade ovarian carcinoma and a germline or somatic BRCA1 or BRCA2 mutation: integrated analysis of data from Study 10 and ARIEL2. Gynecol. Oncol. 2017;147:267–275. doi: 10.1016/j.ygyno.2017.08.022.
    1. Coleman RL, et al. Refinement of prespecified cutoff for genomic loss of heterozygosity (LOH) in ARIEL2 part 1: a phase II study of rucaparib in patients (pts) with high grade ovarian carcinoma (HGOC) J. Clin. Oncol. 2016;34:5540. doi: 10.1200/JCO.2016.34.15_suppl.5540.
    1. Goodall J, et al. Circulating cell-free DNA to guide prostate cancer treatment with PARP inhibition. Cancer Discov. 2017;7:1006–1017. doi: 10.1158/-17-0261.
    1. Patch AM, et al. Whole-genome characterization of chemoresistant ovarian cancer. Nature. 2015;521:489–494. doi: 10.1038/nature14410.
    1. Patel JN, et al. Characterisation of homologous recombination deficiency in paired primary and recurrent high-grade serous ovarian cancer. Br. J. Cancer. 2018;119:1060–1066. doi: 10.1038/s41416-018-0268-6.
    1. Ahmed AA, et al. Driver mutations in TP53 are ubiquitous in high grade serous carcinoma of the ovary. J. Pathol. 2010;221:49–56. doi: 10.1002/path.2696.
    1. Etemadmoghadam D, et al. Synthetic lethality between CCNE1 amplification and loss of BRCA1. Proc. Natl Acad. Sci. USA. 2013;110:19489–19494. doi: 10.1073/pnas.1314302110.
    1. Emmanuel C, et al. Genomic classification of serous ovarian cancer with adjacent borderline differentiates RAS pathway and TP53-mutant tumors and identifies NRAS as an oncogenic driver. Clin. Cancer Res. 2014;20:6618–6630. doi: 10.1158/1078-0432.CCR-14-1292.
    1. Kaldawy A, et al. Low-grade serous ovarian cancer: a review. Gynecol. Oncol. 2016;143:433–438. doi: 10.1016/j.ygyno.2016.08.320.
    1. Mirkovic J, et al. Targeted genomic profiling reveals recurrent KRAS mutations in mesonephric-like adenocarcinomas of the female genital tract. Am. J. Surg. Pathol. 2018;42:227–233. doi: 10.1097/PAS.0000000000000958.
    1. Gershenson DM, et al. Recurrent low-grade serous ovarian carcinoma is relatively chemoresistant. Gynecol. Oncol. 2009;114:48–52. doi: 10.1016/j.ygyno.2009.03.001.
    1. Monk BJ, et al. MILO/ENGOT-ov11: binimetinib versus physician’s choice chemotherapy in recurrent or persistent low-grade serous carcinomas of the ovary, fallopian tube, or primary peritoneum. J. Clin. Oncol. 2020;38:3753–3762. doi: 10.1200/JCO.20.01164.
    1. Hahne JC, et al. Downregulation of AKT reverses platinum resistance of human ovarian cancers in vitro. Oncol. Rep. 2012;28:2023–2028. doi: 10.3892/or.2012.2041.
    1. Wang Y, et al. The BRCA1-delta11q alternative splice isoform bypasses germline mutations and promotes therapeutic resistance to PARP inhibition and cisplatin. Cancer Res. 2016;76:2778–2790. doi: 10.1158/0008-5472.CAN-16-0186.
    1. Bunting SF, et al. 53BP1 inhibits homologous recombination in Brca1-deficient cells by blocking resection of DNA breaks. Cell. 2010;141:243–254. doi: 10.1016/j.cell.2010.03.012.
    1. Hurley RM, et al. 53BP1 as a potential predictor of response in PARP inhibitor-treated homologous recombination-deficient ovarian cancer. Gynecol. Oncol. 2019;153:127–134. doi: 10.1016/j.ygyno.2019.01.015.
    1. Drost R, et al. BRCA1185delAG tumors may acquire therapy resistance through expression of RING-less BRCA1. J. Clin. Invest. 2016;126:2903–2918. doi: 10.1172/JCI70196.
    1. Park PH, et al. Amplification of the mutation-carrying BRCA2 allele promotes RAD51 loading and PARP inhibitor resistance in the absence of reversion mutations. Mol. Cancer Ther. 2020;19:602–613. doi: 10.1158/1535-7163.MCT-17-0256.
    1. Johnson N, et al. Stabilization of mutant BRCA1 protein confers PARP inhibitor and platinum resistance. Proc. Natl Acad. Sci. USA. 2013;110:17041–17046. doi: 10.1073/pnas.1305170110.
    1. Christie EL, et al. Multiple ABCB1 transcriptional fusions in drug resistant high-grade serous ovarian and breast cancer. Nat. Commun. 2019;10:1295. doi: 10.1038/s41467-019-09312-9.
    1. Hussain M, et al. Survival with olaparib in metastatic castration-resistant prostate cancer. N. Engl. J. Med. 2020;383:2345–2357. doi: 10.1056/NEJMoa2022485.
    1. Ignatov T, et al. BRCA1 promoter methylation is a marker of better response to anthracycline-based therapy in sporadic TNBC. Breast Cancer Res. Treat. 2013;141:205–212. doi: 10.1007/s10549-013-2693-9.
    1. Bernards SS, et al. Clinical characteristics and outcomes of patients with BRCA1 or RAD51C methylated versus mutated ovarian carcinoma. Gynecol. Oncol. 2018;148:281–285. doi: 10.1016/j.ygyno.2017.12.004.
    1. Moore KN, et al. Niraparib monotherapy for late-line treatment of ovarian cancer (QUADRA): a multicentre, open-label, single-arm, phase 2 trial. Lancet Oncol. 2019;20:636–648. doi: 10.1016/S1470-2045(19)30029-4.
    1. Noel EE, et al. The association of CCND1 overexpression and cisplatin resistance in testicular germ cell tumors and other cancers. Am. J. Pathol. 2010;176:2607–2615. doi: 10.2353/ajpath.2010.090780.
    1. Whicker ME, Lin ZP, Hanna R, Sartorelli AC, Ratner ES. MK-2206 sensitizes BRCA-deficient epithelial ovarian adenocarcinoma to cisplatin and olaparib. BMC Cancer. 2016;16:550. doi: 10.1186/s12885-016-2598-1.
    1. Moore K, et al. Maintenance olaparib in patients with newly diagnosed advanced ovarian cancer. N. Engl. J. Med. 2018;379:2495–2505. doi: 10.1056/NEJMoa1810858.
    1. McShane LM, et al. REporting recommendations for tumour MARKer prognostic studies (REMARK) Br. J. Cancer. 2005;93:387–391. doi: 10.1038/sj.bjc.6602678.
    1. Frampton GM, et al. Development and validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing. Nat. Biotechnol. 2013;31:1023–1031. doi: 10.1038/nbt.2696.
    1. Sun JX, et al. A computational approach to distinguish somatic vs. germline origin of genomic alterations from deep sequencing of cancer specimens without a matched normal. PLoS Comput Biol. 2018;14:e1005965. doi: 10.1371/journal.pcbi.1005965.
    1. Lanman RB, et al. Analytical and clinical validation of a digital sequencing panel for quantitative, highly accurate evaluation of cell-free circulating tumor DNA. PLoS ONE. 2015;10:e0140712. doi: 10.1371/journal.pone.0140712.
    1. Clark TA, et al. Analytical validation of a hybrid capture–based next-generation sequencing clinical assay for genomic profiling of cell-free circulating tumor DNA. J. Mol. Diagn. 2018;20:686–702. doi: 10.1016/j.jmoldx.2018.05.004.
    1. Talhouk A, et al. Single-patient molecular testing with NanoString nCounter data using a reference-based strategy for batch effect correction. PLoS ONE. 2016;11:e0153844. doi: 10.1371/journal.pone.0153844.

Source: PubMed

3
Iratkozz fel