A phase II study of talazoparib monotherapy in patients with wild-type BRCA1 and BRCA2 with a mutation in other homologous recombination genes

Joshua J Gruber, Anosheh Afghahi, Kirsten Timms, Alyssa DeWees, Wyatt Gross, Vasily N Aushev, Hsin-Ta Wu, Mustafa Balcioglu, Himanshu Sethi, Danika Scott, Jessica Foran, Alex McMillan, James M Ford, Melinda L Telli, Joshua J Gruber, Anosheh Afghahi, Kirsten Timms, Alyssa DeWees, Wyatt Gross, Vasily N Aushev, Hsin-Ta Wu, Mustafa Balcioglu, Himanshu Sethi, Danika Scott, Jessica Foran, Alex McMillan, James M Ford, Melinda L Telli

Abstract

Talazoparib, a PARP inhibitor, is active in germline BRCA1 and BRCA2 (gBRCA1/2)-mutant advanced breast cancer, but its activity beyond gBRCA1/2 is poorly understood. We conducted Talazoparib Beyond BRCA ( NCT02401347 ), an open-label phase II trial, to evaluate talazoparib in patients with pretreated advanced HER2-negative breast cancer (n = 13) or other solid tumors (n = 7) with mutations in homologous recombination (HR) pathway genes other than BRCA1 and BRCA2. In patients with breast cancer, four patients had a Response Evaluation Criteria in Solid Tumors (RECIST) partial response (overall response rate, 31%), and three additional patients had stable disease of ≥6 months (clinical benefit rate, 54%). All patients with germline mutations in PALB2 (gPALB2; encoding partner and localizer of BRCA2) had treatment-associated tumor regression. Tumor or plasma circulating tumor DNA (ctDNA) HR deficiency (HRD) scores were correlated with treatment outcomes and were increased in all gPALB2 tumors. In addition, a gPALB2-associated mutational signature was associated with tumor response. Thus, talazoparib has been demonstrated to have efficacy in patients with advanced breast cancer who have gPALB2 mutations, showing activity in the context of HR pathway gene mutations beyond gBRCA1/2.

Conflict of interest statement

J.J.G. had contracted research with Curis, consults for Sharma Therapeutics, LLC and Guidepoint and reports research support (to his institution) from Hummingbird Biosciences. J.M.F. has received research support (to his institution) from AstraZeneca, Genentech, Incyte, Merus, Pfizer and PUMA. M.L.T. reports research support (to her institution) from AbbVie, Arvinas, Bayer, Biothera, Calithera Biosciences, EMD Serono, Genentech, GlaxoSmithKline, Hummingbird Biosciences, Medivation, Merck, Novartis, OncoSec, Pfizer, PharmaMar, Tesaro and Vertex and consulting/advisory fees from AbbVie, Aduro Biotech, AstraZeneca, Blueprint Medicines, Celgene, Daiichi Sankyo, Genentech/Roche, Gilead, GlaxoSmithKline, G1 Therapeutics, Guardant, Immunomedics, Lilly, Merck, Natera, Novartis, OncoSec, Pfizer, RefleXion and Sanofi. Funding support was provided by Pfizer in the form of an unrestricted research grant for drug and clinical trial budget. V.N.A., H.W., M.B. and H.S. are full-time employees of Natera with stock or options to own stock in the company. K.T. is an employee and shareholder of Myriad Genetics. The remaining authors declare no competing interests.

© 2022. The Author(s).

Figures

Fig. 1. Treatment response and duration for…
Fig. 1. Treatment response and duration for therapy for all patients.
a, Waterfall plot of best change in the sum of longest diameters (SLD) of target lesions by RECIST v.1.1 for all patients (n = 20) treated in cohort B, colored by tumor type. Germline (g) or somatic (s) mutations in genes used for enrollment are indicated. The dashed line represents a 30% decrease in tumor size. b, Duration of therapy by swimmers plot indicating duration of talazoparib therapy. Tumor responses assessments are noted. n = 20 patients. Source data
Fig. 2. Tumor HRD scores and correlation…
Fig. 2. Tumor HRD scores and correlation with treatment response.
HRD score was obtained by NGS assay (Myriad myChoice) from tumor biopsies (n = 15 patients; 2 patients excluded for insufficient sample and 2 patients excluded for assay failure). a, HRD score is plotted in primary and metastatic samples for all evaluable patients with lines connecting paired samples. Horizontal dashed lines indicate an HRD score threshold of ≥33 (captures 99% of known BRCA1- and/or BRCA2-deficient ovarian cancers) or ≥42 (captures 95% of known BRCA1- and/or BRCA2-deficient ovarian cancers). Significance testing by paired t-test for primary versus metastatic HRD scores. b, Correlation of HRD score with talazoparib treatment response. HRD score is plotted against best change in SLD by RECIST. Pearson’s correlation is indicated by the solid line, with associated r and two-sided P value denoted. Vertical dashed lines indicate an HRD threshold of ≥33 or ≥42. In cases where patients had more than one HRD score (for example, due to assay of primary and metastatic tumors), the higher score was used. Source data
Fig. 3. Somatic mutations and LOH identified…
Fig. 3. Somatic mutations and LOH identified by NGS.
Heatmap of next-generation panel sequencing (108 genes interrogated) for HRD-associated and cancer-associated mutations in tumor specimens from patients treated in cohort B (n = 18 patients, 2 excluded due to insufficient sample). Germline (g) and somatic (s) mutations used for study enrollment are indicated. Hierarchical clustering is by Euclidean distance. Bar graphs depict numbers of mutations of each type detected across the cohort. Tumor type is indicated by color. See Supplementary Table 3 for a list of genes assayed. Source data
Fig. 4. ctDNA variants and ctHRD score…
Fig. 4. ctDNA variants and ctHRD score correlating with treatment outcomes.
a, Variants detected by Signatera assay of baseline and progression ctDNA samples plotted by VAF. Dashed lines represent fivefold difference cutoffs, n = 14 patients. b, pWES-identified indels (n = 18 patients) at baseline and progression. c, SNVs identified by pWES at baseline and progression (n = 18 patients). d, Deleterious SNVs enriched fivefold in progression samples (n = 6 patients) from responders (decrease of >20% in SLD) compared to baseline. Variants identified in tumor WES are shown (an anterisk indicates insufficient sample for tumor WES). e, ctDNA SNV counts are significantly increased in progression samples compared to control samples (n = 18 patients), whereas there is no significant difference in indel counts (two-way ANOVA with Tukey’s correction, *P = 0.033; median indicated by horizontal line). NS, not significant. f, scarHRD was used to calculate ctHRD score from baseline pWES and correlated with time to progression in days. The correlation coefficient (r) and two-sided P value were calculated with Pearson’s method and are depicted as a dashed line. n = 12 patients. Source data
Fig. 5. Mutational signature analysis and correlation…
Fig. 5. Mutational signature analysis and correlation with treatment outcome.
a, De novo mutational signatures were computed using all available mutation data including tumor exome and plasma ctDNA exome sequencing. The predominant signature detected in the gPALB2 samples is shown. n = 19 patients. b, Mutational signatures were binned by whether they included the gPALB2-associated signature (HRD) or other signatures and plotted as a percentage of total SNVs. n = 19 patients. c, Treatment response to talazoparib (best change in SLD by RECIST) is plotted according to percentage contribution of HRD signature derived from tumor exome and plasma ctDNA sequencing. Correlation by Pearson’s method is depicted by the dashed line, with the two-sided P value. n = 19 patients. Source data
Extended Data Fig. 1. Talazoparib Beyond BRCA…
Extended Data Fig. 1. Talazoparib Beyond BRCA study schema for cohort B.
Cohort B enrolled patients with advanced HER2-negative breast cancer or other solid tumors lacking a germline BRCA1 or BRCA2 mutation with either a germline or somatic mutation in a gene associated with HR-deficiency. Patients were treated with single-agent talazoparib in an initial cohort of 10 patients. On the basis of observed responses in the first 10 patients, an additional 10 patients were enrolled and treated.
Extended Data Fig. 2. Overlap of variant…
Extended Data Fig. 2. Overlap of variant detection by tumor WES and plasma WES.
Number of detected variants by tumor WES (red), plasma WES at baseline (green) and plasma WES at progression (blue) depicted as proportional venn diagrams. Patient study number and qualifying genetic mutation are above and below, respectively. Source data
Extended Data Fig. 3. Deleterious ctDNA SNVs…
Extended Data Fig. 3. Deleterious ctDNA SNVs enriched at progression in non-responders.
Deleterious ctDNA SNVs enriched 5-fold in progression samples from non-responders (change of SLD > -20%) compared to baseline. Deleterious variants identified in tumor WES are also shown. N = 9 patients. Source data

References

    1. Mateo J, et al. A decade of clinical development of PARP inhibitors in perspective. Ann. Oncol. 2019;30:1437–1447.
    1. Sachdev E, et al. PARP inhibition in cancer: an update on clinical development. Target Oncol. 2019;14:657–679.
    1. Tomao F, et al. Parp inhibitors as maintenance treatment in platinum sensitive recurrent ovarian cancer: an updated meta-analysis of randomized clinical trials according to BRCA mutational status. Cancer Treat. Rev. 2019;80:101909.
    1. Lord CJ, Ashworth A. BRCAness revisited. Nat. Rev. Cancer. 2016;16:110–120.
    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.
    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.
    1. Golan T, et al. Maintenance olaparib for germline BRCA-mutated metastatic pancreatic cancer. N. Engl. J. Med. 2019;381:317–327.
    1. de Bono J, et al. Olaparib for metastatic castration-resistant prostate cancer. N. Engl. J. Med. 2020;382:2091–2102.
    1. Mateo J, et al. DNA-repair defects and olaparib in metastatic prostate cancer. N. Engl. J. Med. 2015;373:1697–1708.
    1. Mateo J, et al. Olaparib in patients with metastatic castration-resistant prostate cancer with DNA repair gene aberrations (TOPARP-B): a multicentre, open-label, randomised, phase 2 trial. Lancet Oncol. 2020;21:162–174.
    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 II TRITON2 study. Clin. Cancer Res. 2020;26:2487–2496.
    1. Ryan CJ, et al. TRITON3: an international, randomized, open-label, phase III study of the PARP inhibitor rucaparib vs. physician’s choice of therapy for patients with metastatic castration-resistant prostate cancer (mCRPC) associated with homologous recombination deficiency (HRD) J. Clin. Oncol. 2018;36:TPS389.
    1. Smith MR, et al. Niraparib in patients (pts) with metastatic castration-resistant prostate cancer (mCRPC) and biallelic DNA-repair gene defects (DRD): correlative measures of tumor response in phase II GALAHAD study. J. Clin. Oncol. 2020;38:118.
    1. Bono JSD, et al. TALAPRO-1: a phase II study of talazoparib (TALA) in men with DNA damage repair mutations (DDRmut) and metastatic castration-resistant prostate cancer (mCRPC)—first interim analysis (IA) J. Clin. Oncol. 2020;38:119–119.
    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.
    1. Dieras V, et al. Veliparib with carboplatin and paclitaxel in BRCA-mutated advanced breast cancer (BROCADE3): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2020;21:1269–1282.
    1. Kurian AW, et al. Genetic testing and results in a population-based cohort of breast cancer patients and ovarian cancer patients. J. Clin. Oncol. 2019;37:1305–1315.
    1. Heeke, A.L., et al. Prevalence of homologous recombination-related gene mutations across multiple cancer types. JCO Precis. Oncol., 2018, PO.17.00286 (2018).
    1. Fraser M, et al. Genomic hallmarks of localized, non-indolent prostate cancer. Nature. 2017;541:359–364.
    1. Pritchard CC, et al. Inherited DNA-repair gene mutations in men with metastatic prostate cancer. N. Engl. J. Med. 2016;375:443–453.
    1. Robinson D, et al. Integrative clinical genomics of advanced prostate cancer. Cell. 2015;162:454.
    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.
    1. Mirza MR, et al. Niraparib maintenance therapy in platinum-sensitive, recurrent ovarian cancer. N. Engl. J. Med. 2016;375:2154–2164.
    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.
    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.
    1. Sharma P, et al. Results of a phase II randomized trial of cisplatin ± veliparib in metastatic triple-negative breast cancer (TNBC) and/or germline BRCA-associated breast cancer (SWOG S1416) J. Clin. Oncol. 2020;38:10001.
    1. Mills GB, et al. Homologous recombination deficiency score shows superior association with outcome compared with its individual score components in platinum-treated serous ovarian cancer. Gynecol. Oncol. 2016;141:2–3.
    1. Sakai W, et al. Secondary mutations as a mechanism of cisplatin resistance in BRCA2-mutated cancers. Nature. 2008;451:1116–1120.
    1. Edwards SL, et al. Resistance to therapy caused by intragenic deletion in BRCA2. Nature. 2008;451:1111–1115.
    1. Sztupinszki Z, et al. Migrating the SNP array-based homologous recombination deficiency measures to next generation sequencing data of breast cancer. NPJ Breast Cancer. 2018;4:16.
    1. Alexandrov LB, et al. Signatures of mutational processes in human cancer. Nature. 2013;500:415–421.
    1. Alexandrov LB, et al. The repertoire of mutational signatures in human cancer. Nature. 2020;578:94–101.
    1. Gruber JJ, et al. Talazoparib beyond BRCA: a phase II trial of talazoparib monotherapy in BRCA1 and BRCA2 wild-type patients with advanced HER2-negative breast cancer. J. Clin. Oncol. 2019;37:3006.
    1. Tung NM, et al. TBCRC 048: a phase II study of olaparib monotherapy in metastatic breast cancer patients with germline or somatic mutations in DNA damage response (DDR) pathway genes (Olaparib Expanded) J. Clin. Oncol. 2020;38:1002.
    1. Kass EM, et al. Double-strand break repair by homologous recombination in primary mouse somatic cells requires BRCA1 but not the ATM kinase. Proc. Natl. Acad. Sci. USA. 2013;110:5564–5569.
    1. Iyevleva AG, et al. Somatic loss of the remaining allele occurs approximately in half of CHEK2-driven breast cancers and is accompanied by a border-line increase of chromosomal instability. Breast Cancer Res. Treat. 2022;192:283–291.
    1. Qian B, et al. Clinical benefit with PARP inhibitor for pathogenic germline FANCA-mutated relapsed epithelial ovarian cancer: a case report. Front. Oncol. 2022;12:778545.
    1. Wilkes DC, et al. A germline FANCA alteration that is associated with increased sensitivity to DNA damaging agents. Cold Spring Harb. Mol. Case Stud. 2017;3:a001487.
    1. Xia B, et al. Control of BRCA2 cellular and clinical functions by a nuclear partner, PALB2. Mol. Cell. 2006;22:719–29..
    1. Sy SM, et al. PALB2 regulates recombinational repair through chromatin association and oligomerization. J. Biol. Chem. 2009;284:18302–18310.
    1. Sy SM, Huen MS, Chen J. PALB2 is an integral component of the BRCA complex required for homologous recombination repair. Proc. Natl. Acad. Sci. USA. 2009;106:7155–7160.
    1. Buisson R, Masson JY. PALB2 self-interaction controls homologous recombination. Nucleic Acids Res. 2012;40:10312–10323.
    1. Park JY, et al. Breast cancer-associated missense mutants of the PALB2 WD40 domain, which directly binds RAD51C, RAD51 and BRCA2, disrupt DNA repair. Oncogene. 2014;33:4803–4812.
    1. Buisson R, et al. Cooperation of breast cancer proteins PALB2 and piccolo BRCA2 in stimulating homologous recombination. Nat. Struct. Mol. Biol. 2010;17:1247–1254.
    1. Foo TK, et al. Compromised BRCA1–PALB2 interaction is associated with breast cancer risk. Oncogene. 2017;36:4161–4170.
    1. Reiss Binder KA, et al. A phase II, single-arm study of maintenance rucaparib in patients with platinum-sensitive advanced pancreatic cancer and a pathogenic germline or somatic mutation in BRCA1, BRCA2, or PALB2. J. Clin. Oncol. 2021;39:2497–2505.
    1. Horak P, et al. Response to olaparib in a PALB2 germline mutated prostate cancer and genetic events associated with resistance. Cold Spring Harb. Mol. Case. Stud. 2019;5:a003657.
    1. Breast Cancer Association Consortium et al. Pathology of tumors associated with pathogenic germline variants in 9 breast cancer susceptibility genes. JAMA Oncol. 2022;8:e216744.
    1. Tischkowitz M, et al. Management of individuals with germline variants in PALB2: a clinical practice resource of the American College of Medical Genetics and Genomics (ACMG) Genet. Med. 2021;23:1416–1423.
    1. von Wahlde MK, et al. Intratumor heterogeneity of homologous recombination deficiency in primary breast cancer. Clin. Cancer Res. 2017;23:1193–1199.
    1. Polak P, et al. A mutational signature reveals alterations underlying deficient homologous recombination repair in breast cancer. Nat. Genet. 2017;49:1476–1486.
    1. Eisenhauer EA, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1) Eur. J. Cancer. 2009;45:228–247.
    1. Watkins JA, et al. Genomic scars as biomarkers of homologous recombination deficiency and drug response in breast and ovarian cancers. Breast Cancer Res. 2014;16:211.
    1. Timms KM, et al. Association of BRCA1/2 defects with genomic scores predictive of DNA damage repair deficiency among breast cancer subtypes. Breast Cancer Res. 2014;16:475.
    1. Reinert T, et al. Analysis of plasma cell-free DNA by ultradeep sequencing in patients with stages I to III colorectal cancer. JAMA Oncol. 2019;5:1124–1131.
    1. Gu Z, Eils R, Schlesner M. Complex heatmaps reveal patterns and correlations in multidimensional genomic data. Bioinformatics. 2016;32:2847–2849.
    1. Favero F, et al. Sequenza: allele-specific copy number and mutation profiles from tumor sequencing data. Ann. Oncol. 2015;26:64–70.
    1. Hulsen T, de Vlieg J, Alkema W. BioVenn—a web application for the comparison and visualization of biological lists using area-proportional Venn diagrams. BMC Genomics. 2008;9:488.
    1. Fantini D, et al. MutSignatures: an R package for extraction and analysis of cancer mutational signatures. Sci. Rep. 2020;10:18217.
    1. Wickham, H. ggplot2: Elegant Graphics for Data Analysis (Springer-Verlag, 2016).

Source: PubMed

3
Sottoscrivi