965
Views
7
CrossRef citations to date
0
Altmetric
Review

PARP inhibitors as single agents and in combination therapy: the most promising treatment strategies in clinical trials for BRCA-mutant ovarian and triple-negative breast cancers

ORCID Icon & ORCID Icon
Pages 607-631 | Received 30 Dec 2021, Accepted 14 Apr 2022, Published online: 03 May 2022

References

  • Anders C, Carey LA .Understanding and treating triple-negative breast cancer Oncology (Williston Park NY). 2008 Vol. 22 11 1233 Papers of special note have been highlighted as either of interest (•) or of considerable interest (••) to readers
  • Lehmann BD, et al. Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J Clin Invest. 2011;121(7):2750–2767.
  • Burstein MD, Tsimelzon A, Poage GM, et al. Comprehensive genomic analysis identifies novel subtypes and targets of triple-negative breast cancer. Clin Cancer Res. 2015;21(7):1688–1698.
  • Schmid P, et al. KEYNOTE-522: phase III study of neoadjuvant pembrolizumab D chemotherapy vs. placebo D chemotherapy, followed by adjuvant pembrolizumab vs. placebo for earlystage TNBC. In: ELSEVIER RADARWEG. Vol. 29, NX AMSTERDAM NETHERLANDS: p.1043.
  • Bardia A, et al. Biomarker analyses in the phase 3 ascent study of sacituzumab govitecan versus chemotherapy in patients with metastatic triple-negative breast cancer. Ann Oncol. 2021;32(9), 1148–1156. https://doi.org/https://doi.org/10.1016/j.annonc.2021.06.002
  • Curtin NJ. DNA repair dysregulation from cancer driver to therapeutic target. Nat Rev Cancer. 2012;12(12):801–817.
  • Lord CJ, Ashworth A. BRCAness revisited. Nat Rev Cancer. 2016;16(2):110–120.
  • Anglian breast cancer study G. Prevalence and penetrance of BRCA1 and BRCA2 mutations in a population-based series of breast cancer cases. Br J Cancer. 2000;83(10):1301.
  • Chen H, et al. Association between BRCA status and triple-negative breast cancer: a meta-analysis. Front Pharmacol. 2018;9:909.
  • Atchley DP, Albarracin CT, Lopez A, et al. Clinical and pathologic characteristics of patients with brca -positive and brca -negative breast cancer. J clin oncol. 2008;26(26):4282.
  • Daly MB, Pal T, Berry MP, et al. Genetic/familial high-risk assessment: breast, ovarian, and pancreatic, version 2.2021, NCCN clinical practice guidelines in oncology. J National Compr Cancer Network. 2021;19(1):77–102.
  • Levy-Lahad E, Catane R, Eisenberg S, et al. Founder BRCA1 and BRCA2 mutations in ashkenazi jews in Israel: frequency and differential penetrance in ovarian cancer and in breast-ovarian cancer families. Am J Hum Genet. 1997;60(5):1059.
  • Turner N, Tutt A, Ashworth A. Hallmarks of’BRCAness’ in sporadic cancers. Nat Rev Cancer. 2004;4(10):814–819.
  • Telli ML, Timms KM, Reid J, 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(15):3764–3773.
  • Kaklamani VG, et al. Phase II neoadjuvant clinical trial of carboplatin and eribulin in women with triple negative early-stage breast cancer (NCT01372579). Breast Cancer Res Treat. 2015;151(3):629–638.
  • Manasaryan G, Suplatov D, Pushkarev S, et al. Bioinformatic analysis of the nicotinamide binding site in poly (adp-ribose) polymerase family proteins. Cancers (Basel). 2021;13(6):1201.
  • Vyas S, Matic I, Uchima L, et al. Family-wide analysis of poly (ADP-ribose) polymerase activity. Nat Commun. 2014;5(1):4426.
  • Bai P. Biology of poly (ADP-ribose) polymerases: the factotums of cell maintenance. Mol Cell. 2015;58(6):947–958.
  • Wei L, Nakajima S, Hsieh C-L, et al. Damage response of XRCC1 at sites of DNA single strand breaks is regulated by phosphorylation and ubiquitylation after degradation of poly(ADP-ribose). J Cell Sci. 2013;126(Pt 19):4414–4423.
  • Isabelle M, Moreel X, Gagné J-P, et al. Investigation of PARP-1, PARP-2, and PARG interactomes by affinity-purification mass spectrometry. Proteome Sci. 2010;8(1):22.
  • Chaudhuri AR, Nussenzweig A. The multifaceted roles of PARP1 in DNA repair and chromatin remodelling. Nat Rev Mol Cell Biol. 2017;18(10):610–621.
  • Murai J, et al. Trapping of PARP1 and PARP2 by clinical PARP inhibitors. Cancer Res. 2012;72(21):5588–5599.
  • Murai J, et al. Stereospecific PARP trapping by BMN 673 and comparison with olaparib and rucaparib. Mol Cancer Ther. 2014;13(2):433–443.
  • Pommier Y, O’Connor MJ, De Bono J. Laying a trap to kill cancer cells: PARP inhibitors and their mechanisms of action. Sci Transl Med. 2016;8(362):17.
  • Hopkins TA, Shi Y, Rodriguez LE, et al. Mechanistic dissection of parp1 trapping and the impact on in vivo tolerability and efficacy of parp inhibitors. Mol Cancer Res. 2015;13(11):1465–1477.
  • Ummarino S, Hausman C, Di Ruscio A. The PARP way to epigenetic changes. Genes (Basel). 2021;12(3):446.
  • Obaji E, Maksimainen MM, Galera-Prat A, et al. Activation of PARP2/ARTD2 by DNA damage induces conformational changes relieving enzyme autoinhibition. Nat Commun. 2021;12(1):1–8.
  • Boehler C, et al., Poly (ADP-ribose) polymerase 3 (PARP3), a newcomer in cellular response to DNA damage and mitotic progression. Proceedings of the National Academy of Sciences, 2011. 108 (7): p. 2783–2788.
  • Haikarainen T, Krauss S, Lehtio L. Tankyrases: structure, function and therapeutic implications in cancer. Curr Pharm Des. 2014;20(41):6472–6488.
  • Wu J, Crowe DL. PARP5B is required for nonhomologous end joining during tumorigenesis in vivo. Mol Carcinog. 2021;61(1):85–98.
  • Tuncel H, Tanaka S, Oka S, et al. PARP6, a mono (ADP-ribosyl) transferase and a negative regulator of cell proliferation, is involved in colorectal cancer development. Int J Oncol. 2012;41(6):2079–2086.
  • Qi G, Kudo Y, Tang B, et al. PARP6 acts as a tumor suppressor via downregulating survivin expression in colorectal cancer. Oncotarget. 2016;7(14):18812.
  • Gozgit JM, Vasbinder MM, Abo RP, et al. PARP7 negatively regulates the type I interferon response in cancer cells and its inhibition triggers antitumor immunity. Cancer Cell. 2021;39(9):1214–1226.
  • Xing J, Zhang A, Du Y, et al. Identification of poly (ADP-ribose) polymerase 9 (PARP9) as a noncanonical sensor for RNA virus in dendritic cells. Nat Commun. 2021;12(1):1–17.
  • Schleicher EM, Galvan AM, Imamura-Kawasawa Y, et al. PARP10 promotes cellular proliferation and tumorigenesis by alleviating replication stress. Nucleic Acids Res. 2018;46(17):8908–8916.
  • Zhao Y, et al. PARP10 suppresses tumor metastasis through regulation of Aurora A activity. Oncogene. 2018;37(22):2921–2935.
  • Guo T, et al. ADP-ribosyltransferase PARP11 modulates the interferon antiviral response by mono-ADP-ribosylating the ubiquitin E3 ligase β-TrCP. Nat Microbiol. 2019;4(11):1872–1884.
  • Welsby I, Hutin D, Gueydan C, et al. PARP12, an interferon-stimulated gene involved in the control of protein translation and inflammation. J Biol Chem. 2014;289(38):26642–26657.
  • Todorova T, Bock FJ, Chang P. Poly (ADP-ribose) polymerase-13 and RNA regulation in immunity and cancer. Trends Mol Med. 2015;21(6):373–384.
  • Qin W, et al. Research progress on PARP14 as a drug target. Front Pharmacol. 2019;10:172.
  • Jwa M, Chang P. PARP16 is a tail-anchored endoplasmic reticulum protein required for the PERK-and IRE1α-mediated unfolded protein response. Nat Cell Biol. 2012;14(11):1223–1230.
  • Steffen JD, et al. Structural implications for selective targeting of PARPs. Front Oncol. 2013;3:301.
  • Pilié PG, Gay CM, Byers LA, et al. PARP Inhibitors: extending benefit beyond brca -mutant cancers. Clin Cancer Res. 2019;25(13):3759–3771.
  • Murai J, Pommier Y. PARP trapping beyond homologous recombination and platinum sensitivity in cancers. Annu Rev Cancer Biol. 2019;3(1):131–150.
  • Radhakrishnan SK, Jette N, Lees-Miller SP. Non-homologous end joining: emerging themes and unanswered questions. DNA Repair (Amst). 2014;17:2–8.
  • Kim G, Ison G, McKee AE, et al. FDA approval summary: olaparib monotherapy in patients with deleterious germline brca -mutated advanced ovarian cancer treated with three or more lines of chemotherapy. Clin Cancer Res. 2015;21(19):4257–4261.
  • Robson M, Im S-A, Senkus E, et al., Olaparib for metastatic breast cancer in patients with a germline brca mutation. N Engl J Med. 377(6): 523–533. 2017.
  • Golan T, Hammel P, Reni M, et al. Maintenance olaparib for germline brca -mutated metastatic pancreatic cancer. N Engl J Med. 2019;381(4):317–327.
  • Ray-Coquard I, Pautier P, Pignata S, et al. Olaparib plus bevacizumab as first-line maintenance in ovarian cancer. N Engl J Med. 2019;381(25):2416–2428.
  • de Bono J, Mateo J, Fizazi K, et al. Olaparib for metastatic castration-resistant prostate cancer. N Engl J Med. 2020;382(22):2091–2102.
  • Robson ME, Tung N, Conte P, et al., OlympiAD final overall survival and tolerability results: olaparib versus chemotherapy treatment of physician’s choice in patients with a germline BRCA mutation and HER2-negative metastatic breast cancer. Ann Oncol. 30(4): 558–566. 2019.
  • Robson M, et al. Abstract PD4-03: olympiAD extended follow-up for overall survival and safety: olaparib versus chemotherapy treatment of physician’s choice in patients with a germline BRCA mutation and HER2-negative metastatic breast cancer. Cancer Res. 2020;80(4 Supplement):4.
  • Litton JK, Rugo HS, Ettl J, et al., Talazoparib in patients with advanced breast cancer and a germline brca mutation. N Engl J Med. 379(8): 753–763. 2018.
  • Rugo HS, Ettl J, Hurvitz SA, et al. Outcomes in clinically relevant patient subgroups from the embraca study: talazoparib vs physician’s choice standard-of-care chemotherapy . JNCI Cancer Spectr. 2020; 4(1) 85.
  • Litton JK, Hurvitz SA, Mina LA, et al., Talazoparib versus chemotherapy in patients with germline BRCA1/2-mutated HER2-negative advanced breast cancer: final overall survival results from the EMBRACA trial. Ann Oncol. 31(11): 1526–1535. 2020.
  • Taylor AM, Chan DLH, Tio M, et al. PARP (Poly ADP-Ribose Polymerase) inhibitors for locally advanced or metastatic breast cancer. Cochrane Database Syst Rev. 2021;4(4):Cd011395.
  • 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(11):2487–2496.
  • 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(1):75–87.
  • Swisher EM, et al. Molecular and clinical determinants of response and resistance to rucaparib for recurrent ovarian cancer treatment in ARIEL2 (Parts 1 and 2). Nat Commun. 2021;12(1):2487.
  • Oza AM, et al. Patient-centered outcomes in ariel3, a phase iii, randomized, placebo-controlled trial of rucaparib maintenance treatment in patients with recurrent ovarian carcinoma. J Clin Oncol. 2020;38(30):3494–3505.
  • Gonzalez-Martin A, et al. Niraparib in patients with newly diagnosed advanced ovarian cancer. N Engl J Med. 2019;381(25):2391–2402.
  • Chowdhury S, et al. Pamiparib, an investigational PARP inhibitor, in patients with metastatic castration-resistant prostate cancer (mCRPC) and acirculating tumor cell (CTC) homologous recombination deficiency (HRD) phenotype or BRCA defects: atrial in progress. J Cli Oncol. 2019 ;37(15_suppl):TPS5086–TPS5086.
  • Ciardiello F, et al. A phase 3, double-blind, randomized study of pamiparib versus placebo as maintenance therapy in patients with inoperable, locally advanced, or metastatic gastric cancer that responded to platinum based first-line chemotherapy. Ann Oncol. 2018;29:viii205–viii270.
  • Sun T, et al. A phase 2 study of pamiparib in the treatment of patients with locally advanced or metastatic HER2-negative breast cancer with germline BRCA mutation. J clin oncol. 2021;39(15_suppl):1087.
  • Boussios S, et al. Veliparib in ovarian cancer: a new synthetically lethal therapeutic approach. Invest New Drugs. 2020;38(1):181–193.
  • O'Shaughnessy J, et al. Phase III study of iniparib plus gemcitabine and carboplatin versus gemcitabine and carboplatin in patients with metastatic triple-negative breast cancer. J Clin Oncol. 2014;32(34):3840–7. https://doi.org/10.1200/JCO.2014.55.2984.
  • Patel A G, et al. Failure of iniparib to inhibit poly(ADP-Ribose) polymerase in vitro. Clin Cancer Res. 2012;18(6):1655–62. https://doi.org/10.1158/1078-0432.CCR-11-2890.
  • Murai J, et al. Trapping of PARP1 and PARP2 by Clinical PARP Inhibitors. Cancer Res. 2012;72(21):5588–99. https://doi.org/10.1158/0008-5472.CAN-12-2753.
  • Schiewer MJ, et al. Dual roles of PARP-1 promote cancer growth and progression. Cancer Discov. 2012;2(12):1134–49. https://doi.org/10.1158/2159-8290.CD-12-0120.
  • Audeh MW, et al. Oral poly (ADP-ribose) polymerase inhibitor olaparib in patients with BRCA1 or BRCA2 mutations and recurrent ovarian cancer: a proof-of-concept trial. Lancet. 2010;376(9737):245–251.
  • Fong PC, et al. Poly (ADP)-ribose polymerase inhibition: frequent durable responses in BRCA carrier ovarian cancer correlating with platinum-free interval. J clin oncol. 2010;28(15):2512–2519.
  • Zhou P, et al. Recent advancements in PARP inhibitors-based targeted cancer therapy. Precision Clin Med. 2020;3(3):187–201.
  • Li H, et al. PARP inhibitor resistance: the underlying mechanisms and clinical implications. Mol Cancer. 2020;19(1):1–16.
  • Abbotts R, et al., DNA methyltransferase inhibitors induce a BRCAness phenotype that sensitizes NSCLC to PARP inhibitor and ionizing radiation Proc Natl Acad Sci USA. 2019;116( 45):22609–22618.
  • Hopkins JL, Zou L. Induction of BRCAness in triple-negative breast cancer by a CDK12/13 inhibitor improves chemotherapy. Cancer Cell. 2019;36(5):461–463.
  • Wiegmans AP, et al. Differences in expression of key DNA damage repair genes after epigenetic-induced BRCAness dictate synthetic lethality with PARP1 inhibition. Mol Cancer Ther. 2015;14(10):2321–2331.
  • Ibrahim YH, et al. PI3K inhibition impairs BRCA1/2 expression and sensitizes BRCA-proficient triple-negative breast cancer to PARP inhibition. Cancer Discov. 2012;2(11):1036–1047.
  • Mo W, et al. mTOR inhibitors suppress homologous recombination repair and synergize with PARP inhibitors via regulating SUV39H1 in BRCA-proficient triple-negative breast cancer. Clin Cancer Res. 2016;22(7):1699–1712.
  • Smith J, et al. The ATM–Chk2 and ATR–Chk1 pathways in DNA damage signaling and cancer. Adv Cancer Res. 2010;108:73–112.
  • Yazinski SA, et al. ATR inhibition disrupts rewired homologous recombination and fork protection pathways in PARP inhibitor-resistant BRCA-deficient cancer cells. Genes Dev. 2017;31(3):318–332.
  • Renwick A, et al. ATM mutations that cause ataxia-telangiectasia are breast cancer susceptibility alleles. Nat Genet. 2006;38(8):873–875.
  • Kim H, et al. Targeting the ATR/CHK1 axis with PARP inhibition results in tumor regression in BRCA-mutant ovarian cancer models. Clin Cancer Res. 2017;23(12):3097–3108.
  • Kim H, et al. Combining PARP with ATR inhibition overcomes PARP inhibitor and platinum resistance in ovarian cancer models. Nat Commun. 2020;11(1):1–16.
  • Lloyd RL, et al. Combined PARP and ATR inhibition potentiates genome instability and cell death in ATM-deficient cancer cells. Oncogene. 2020;39(25):4869–4883.
  • Smith HL, Prendergast L, Curtin NJ. Exploring the synergy between PARP and CHK1 inhibition in matched BRCA2 mutant and corrected cells. Cancers (Basel). 2020;12(4):878.
  • Gralewska P, et al. PARP inhibition increases the reliance on atr/chk1 checkpoint signaling leading to synthetic lethality—an alternative treatment strategy for epithelial ovarian cancer cells independent from hr effectiveness. Int J Mol Sci. 2020;21(24):9715.
  • Schmidt M, et al. Regulation of G2/M transition by inhibition of WEE1 and PKMYT1 kinases. Molecules. 2017;22(12):2045.
  • Ha D-H, et al. Antitumor effect of a WEE1 inhibitor and potentiation of olaparib sensitivity by DNA damage response modulation in triple-negative breast cancer. Sci Rep. 2020;10(1):1–13.
  • Smith H, Prendergast L, Curtin N. Investigating synergy between WEE1 and PARP inhibitors in BRCA2 mutant and corrected cells. Eur J Cancer. 2020;138:S36–S37.
  • Chen X, et al. Targeting replicative stress and dna repair by combining parp and wee1 kinase inhibitors is synergistic in triple negative breast cancers with cyclin e or brca1 alteration. Cancers (Basel). 2021;13(7):1656.
  • Westin SN, et al. EFFORT: efficacy of adavosertib in parp resisTance: a randomized two-arm non-comparative phase II study of adavosertib with or without olaparib in women with PARP-resistant ovarian cancer. J clin oncol. 2021;39(15_suppl):5505.
  • Hamilton E, et al. Phase Ib study of adavosertib in combination with olaparib in patients with refractory solid tumors: dose escalation. Cancer Res. 2019;79(13_Suppl):Abstract nr CT025.
  • Chow JPH, Poon RYC. The CDK1 inhibitory kinase MYT1 in DNA damage checkpoint recovery. Oncogene. 2013;32(40):4778–4788.
  • Booher RN, Holman PS, Fattaey A. Human Myt1 is a cell cycle-regulated kinase that inhibits Cdc2 but not Cdk2 activity. J Biol Chem. 1997;272(35):22300–22306.
  • Parker LL, Piwnica-Worms H. Inactivation of the p34cdc2-cyclin B complex by the human WEE1 tyrosine kinase. Science. 1992;257(5078):1955–1957.
  • Lewis CW, et al. Upregulation of myt1 promotes acquired resistance of cancer cells to wee1 inhibition. Cancer Res. 2019;79(23):5971–5985.
  • Ceccaldi R, et al. Homologous-recombination-deficient tumours are dependent on Polθ-mediated repair. Nature. 2015;518(7538):258–262.
  • Ceccaldi R, Rondinelli B, D’Andrea AD. Repair pathway choices and consequences at the double-strand break. Trends Cell Biol. 2016;26(1):52–64.
  • Zhou J, et al. A first-in-class polymerase theta inhibitor selectively targets homologous-recombination-deficient tumors. Nature Cancer. 2021;2(6):598–610. https://doi.org/https://doi.org/10.1038/s43018-021-00203-x
  • Zatreanu D, et al. Polθ inhibitors elicit BRCA-gene synthetic lethality and target PARP inhibitor resistance. Nat Commun. 2021;12(1):1–15.
  • Stecklein SR, et al., BRCA1 and HSP90 cooperate in homologous and non-homologous DNA double-strand-break repair and G2/M checkpoint activation Proc Natl Acad Sci U S A. 2012;109( 34):13650–13655.
  • Jiang J, et al. Ganetespib overcomes resistance to PARP inhibitors in breast cancer by targeting core proteins in the DNA repair machinery. Invest New Drugs. 2017;35(3):251.
  • Choi YE, et al. Sublethal concentrations of 17-AAG suppress homologous recombination DNA repair and enhance sensitivity to carboplatin and olaparib in HR proficient ovarian cancer cells. Oncotarget. 2014;5(9):2678.
  • Konstantinopoulos P, et al. In vivo synergism between PARP-inhibitor olaparib and HSP90-inhibitor AT13387 in high grade serous ovarian cancer patient derived xenografts. J Clin Oncol. 2016;34:(15_suppl):e17045–e17045.
  • Hustedt N, Durocher D. The control of DNA repair by the cell cycle. Nat Cell Biol. 2017;19(1):1–9.
  • Symington LS, Gautier J. Double-strand break end resection and repair pathway choice. Annu Rev Genet. 2011;45:247–271.
  • Liu T, Huang J. DNA End Resection: facts and Mechanisms. Genomics Proteomics Bioinformatics. 2016;14(3):126–130.
  • Tomimatsu N, et al. Phosphorylation of EXO1 by CDKs 1 and 2 regulates DNA end resection and repair pathway choice. Nat Commun. 2014;5(1):1–10.
  • Bajrami I, et al. Genome-wide profiling of genetic synthetic lethality identifies CDK12 as a novel determinant of PARP1/2 inhibitor sensitivity. Cancer Res. 2014;74(1):287–297.
  • Joshi PM, et al. Ovarian cancer-associated mutations disable catalytic activity of CDK12, a kinase that promotes homologous recombination repair and resistance to cisplatin and poly (ADP-ribose) polymerase inhibitors. J Biol Chem. 2014;289(13):9247–9253.
  • Johnson SF, et al. CDK12 inhibition reverses de novo and acquired PARP inhibitor resistance in BRCA wild-type and mutated models of triple-negative breast cancer. Cell Rep. 2016;17(9):2367–2381.
  • Militello AM, et al. Mechanism of action and clinical efficacy of CDK4/6 inhibitors in BRCA-mutated, estrogen receptor-positive breast cancers: case report and literature review. Front Oncol. 2019;9:759.
  • Aziz D, et al. Cyclin E1 protein is stabilized in BRCA1 mutated breast cancers leading to synergy between CDK2 and PARP inhibitors. npj Breast Cancer, 2021;7,111. https://doi.org/https://doi.org/10.1038/s41523-021-00312-x
  • Carey JPW, et al. Synthetic lethality of PARP inhibitors in combination with MYC blockade is independent of BRCA status in triple-negative breast cancer. Cancer Res. 2018;78(3):742–757.
  • Ivy SP, et al. Cediranib, a pan-VEGFR inhibitor, and olaparib, a PARP inhibitor, in combination therapy for high grade serous ovarian cancer. Expert Opin Investig Drugs. 2016;25(5):597–611.
  • Westin SN, et al. Phase ib dose expansion and translational analyses of olaparib in combination with capivasertib in recurrent endometrial, triple-negative breast, and ovarian cancer. Clin Cancer Res. 2021;27(23):6354–6365. https://doi.org/https://doi.org/10.1158/1078-0432.CCR-21-1656
  • Westin SN, et al. Phase Itrial of olaparib (PARP inhibitor) and vistusertib (mTORC1/2 inhibitor) in recurrent endometrial, ovarian and triple negative breast cancer. J Clin Oncol. 2018 ;36:(15_suppl):5504–5504.
  • Konstantinopoulos PA, et al. Olaparib and α-specific PI3K inhibitor alpelisib for patients with epithelial ovarian cancer: a dose-escalation and dose-expansion phase 1b trial. Lancet Oncol. 2019;20(4):570–580.
  • Kondrashova O, et al. Methylation of all BRCA1 copies predicts response to the PARP inhibitor rucaparib in ovarian carcinoma. Nat Commun. 2018;9(1):1–16.
  • Guo X, et al. Acetylation of 53BP1 dictates the DNA double strand break repair pathway. Nucleic Acids Res. 2018;46(2):689–703.
  • Du Y, et al. Blocking c-Met–mediated PARP1 phosphorylation enhances anti-tumor effects of PARP inhibitors. Nat Med. 2016;22(2):194–201.
  • Gogola E, et al. Selective loss of PARG restores PARylation and counteracts PARP inhibitor-mediated synthetic lethality. Cancer Cell. 2018;33(6):1078–1093.
  • Jasek E, Gajda M, Lis GJ, et al. Combinatorial effects of PARP inhibitor PJ34 and histone deacetylase inhibitor vorinostat on leukemia cell lines. Anticancer Res. 2014;34(4):1849–1856.
  • Ha K, Fiskus W, Choi DS, et al. Histone deacetylase inhibitor treatment induces ‘BRCAness’ and synergistic lethality with PARP inhibitor and cisplatin against human triple negative breast cancer cells. Oncotarget. 2014;5(14):5637.
  • Valdez BC, Li Y, Murray D, et al. Combination of a hypomethylating agent and inhibitors of PARP and HDAC traps PARP1 and DNMT1 to chromatin, acetylates DNA repair proteins, down-regulates NuRD and induces apoptosis in human leukemia and lymphoma cells. Oncotarget. 2018;9(3):3908.
  • Min A, Im S-A, Kim DK, et al. Histone deacetylase inhibitor, suberoylanilide hydroxamic acid (SAHA), enhances anti-tumor effects of the poly (ADP-ribose) polymerase (PARP) inhibitor olaparib in triple-negative breast cancer cells. Breast Cancer Res. 2015;17(1):1–13.
  • Pulliam N, Fang F, Ozes AR, et al. An effective epigenetic-PARP inhibitor combination therapy for breast and ovarian cancers independent of BRCA mutations. Clin Cancer Res. 2018;24(13):3163–3175.
  • Mehdipour P, Chen R, De Carvalho DD. The next generation of DNMT inhibitors. Nature Cancer. 2021;2(10):1000–1001
  • Arun BK, et al. Abstract PD4-01: first-line veliparib plus carboplatin/paclitaxel in patients with HER2-negative advanced/metastatic gBRCA-associated breast cancer: planned subgroup analysis from the phase 3 BROCADE3 trial. Cancer Res. 2020;80(4 Supplement):4.
  • Ayoub JP, Friedlander ML, Dieras VC, et al. 140O veliparib plus carboplatin-paclitaxel in patients with HER2-negative advanced/metastatic gBRCA-associated breast cancer: results in hormone receptor-positive and triple-negative breast cancer subgroups from the phase III BROCADE3 trial. Ann Oncol. 2020;31:S65.
  • Matulonis UA, Monk BJ. PARP inhibitor and chemotherapy combination trials for the treatment of advanced malignancies: does a development pathway forward exist? Ann Oncol. 2017;28(3):443–447.
  • Thallinger C, Füreder T, Preusser M, et al. Review of cancer treatment with immune checkpoint inhibitors. Wien Klin Wochenschr. 2018;130(3):85–91.
  • Thomas R, Al-Khadairi G, Decock J. Immune checkpoint inhibitors in triple negative breast cancer treatment: promising future prospects. Front Oncol. 2020;10:600573.
  • Couch FJ, et al. Inherited mutations in 17 breast cancer susceptibility genes among a large triple-negative breast cancer cohort unselected for family history of breast cancer. J clin oncol. 2015;33(4):304.
  • McCann KE, Hurvitz SA. Advances in the use of PARP inhibitor therapy for breast cancer. Drugs Context. 2018;7,212540. https://doi.org/https://doi.org/10.7573/dic.212540
  • Jiao S, et al. PARP inhibitor upregulates PD-L1 expression and enhances cancer-associated immunosuppression. Clin Cancer Res. 2017;23(14):3711–3720.
  • Vinayak S, et al. Open-label clinical trial of niraparib combined with pembrolizumab for treatment of advanced or metastatic triple-negative breast cancer. JAMA Oncol. 2019;5(8):1132–1140.
  • Thomas A, Vilimas R, Trindade C, et al. Durvalumab in combination with olaparib in patients with relapsed SCLC: results from a phase II study. J Thorac Oncol. 2019;14(8):1447–1457.
  • Lu R-M, Hwang Y-C, Liu I-J, et al. Development of therapeutic antibodies for the treatment of diseases. J Biomed Sci. 2020;27(1):1–30.
  • Singh S, Kumar NK, Dwiwedi P, et al. Monoclonal antibodies: a review. Curr Clin Pharmacol. 2018;13(2):85–99.
  • Beck A, Goetsch L, Dumontet C, et al. Strategies and challenges for the next generation of antibody–drug conjugates. Nat Rev Drug Discov. 2017;16(5):315–337.
  • Polakis P. Antibody drug conjugates for cancer therapy. Pharmacol Rev. 2016;68(1):3–19.
  • Khongorzul P, Ling CJ, Khan FU, et al. Antibody–drug conjugates: a comprehensive review. Mol Cancer Res. 2020;18(1):3–19.
  • Cardillo TM, Sharkey RM, Rossi DL, et al. Synthetic lethality exploitation by an anti–trop-2-sn-38 antibody–drug conjugate, immu-132, plus parp inhibitors in brca1/2 –wild-type triple-negative breast cancer. Clin Cancer Res. 2017;23(13):3405–3415.
  • Denkert C, von Minckwitz G, Brase JC, et al. Tumor-infiltrating lymphocytes and response to neoadjuvant chemotherapy with or without carboplatin in human epidermal growth factor receptor 2-positive and triple-negative primary breast cancers. J Clin oncol. 2015;33(9):983–991.
  • Pruneri G, Vingiani A, Bagnardi V, et al. Clinical validity of tumor-infiltrating lymphocytes analysis in patients with triple-negative breast cancer. Ann Oncol. 2016;27(2):249–256.
  • Vihervuori H, Autere TA, Repo H, et al. Tumor-infiltrating lymphocytes and CD8+ T cells predict survival of triple-negative breast cancer. J Cancer Res Clin Oncol. 2019;145(12):3105–3114.
  • Lakhani SR, Jacquemier J, Sloane JP, et al. Multifactorial analysis of differences between sporadic breast cancers and cancers involving BRCA1 and BRCA2 mutations. J Natl Cancer Inst. 1998;90(15):1138–1145.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.