First-in-human study of the PARP/tankyrase inhibitor E7449 in patients with advanced solid tumours and evaluation of a novel drug-response predictor

Ruth Plummer, Divyanshu Dua, Nicola Cresti, Yvette Drew, Peter Stephens, Marie Foegh, Steen Knudsen, Pallavi Sachdev, Bipin M Mistry, Vaishali Dixit, Sharon McGonigle, Nancy Hall, Mark Matijevic, Shannon McGrath, Debashis Sarker, Ruth Plummer, Divyanshu Dua, Nicola Cresti, Yvette Drew, Peter Stephens, Marie Foegh, Steen Knudsen, Pallavi Sachdev, Bipin M Mistry, Vaishali Dixit, Sharon McGonigle, Nancy Hall, Mark Matijevic, Shannon McGrath, Debashis Sarker

Abstract

Background: This phase 1 study examined the safety, maximum-tolerated dose (MTD) and antitumour activity of E7449, a novel PARP 1/2 and tankyrase 1/2 inhibitor.

Methods: E7449 was orally administered once daily in 28-day cycles to patients with advanced solid tumours (50-800-mg doses). Archival tumour samples from consenting patients were evaluated for the expression of 414 genes in a biomarker panel (2X-121 drug-response predictor [DRP]) found to be predictive of the response to E7449 in cell lines.

Results: Forty-one patients were enrolled (13 pancreatic, 5 ovarian, 4 each with breast, lung or colorectal cancer and 11 with other tumour types). The most common grade ≥3 treatment-related adverse event was fatigue (n = 7, 17.1%). Five patients experienced a dose-limiting toxicity (fatigue, n = 4, 800 mg; anaphylaxis, n = 1, 600 mg) for an MTD of 600 mg. E7449 exhibited antitumour activity in solid tumours, including 2 partial responses (PRs), and stable disease (SD) in 13 patients, which was durable (>23 weeks) for 8 patients. In 13 patients, the 2X-121 DRP identified those achieving PR and durable SD. E7449 showed good tolerability, promising antitumour activity and significant concentration-dependent PARP inhibition following 50-800-mg oral dosing.

Conclusion: The results support further clinical investigation of E7449 and its associated biomarker 2X-121 DRP.

Clinical trial registration: www.ClinicalTrials.gov code: NCT01618136.

Conflict of interest statement

Ruth Plummer has received honoraria for attending advisory boards from Pierre Faber, Genmab, Bayer, Octimet, Clovis Oncology, Novartis, Karus Therapeutics, Biosceptre, BMS, Cybrexa, Ellipses and Sanofi Aventis, and for delivery of educational talks or chairing educational meetings by AstraZeneca, Novartis, Bayer, Tesaro and BMS. R.P. is named on the patent of use for rucaparib. Yvette Drew has participated on advisory boards for Clovis Oncology, AstraZeneca, Merck, Tesaro, Inc. and Genmab. Y.D. has received research grant funding from Clovis Oncology, and was involved in the preclinical and clinical development of rucaparib. Newcastle University (Y.D.’s employer) and Y.D. have received royalties for their involvement in the development of rucaparib. Y.D. has received research grant funding from AstraZeneca and Tesaro, Inc. Y.D. is an investigator on clinical trials of olaparib and rucaparib. Marie Foegh and Steen Knudsen are employees of, and have ownership interest in Oncology Venture, which owns rights related to the contents of this article. Pallavi Sachdev, Bipin M. Mistry, Vaishali Dixit, Sharon McGonigle, Nancy Hall, Mark Matijevic and Shannon McGrath are/were full-time employees of Eisai Inc., at the time of the study. Nicola Cresti and Divyanshu Dua have no relevant conflicts of interest to declare. Dr Peter Stephens has received honoraria for serving on advisory boards for Novartis, and educational support from Eisai and Daiichi Sankyo. Dr Debashis Sarker has received honoraria from Pfizer, Bayer and Ipsen, and has had consultancy/advisory roles with Novartis, Ipsen and Eisai.

Figures

Fig. 1. Maximum percentage of inhibition of…
Fig. 1. Maximum percentage of inhibition of PARP by patient and E7449 dose during cycle 1 (any time point).
aAll patients in the 600-mg cohort exceeded the maximum % PARP inhibition measurement at 1 or more time points. Maximum inhibition was calculated when the measured PAR in peripheral blood mononuclear cells was below the assay lower detection limit. The measured value was then replaced by the assay lower detection limit. PAR, polyadenosine diphosphate–ribose; PARP, polyadenosine diphosphate–ribose polymerases.
Fig. 2. Biomarker predicted tumour sensitivity to…
Fig. 2. Biomarker predicted tumour sensitivity to E7449 versus clinical outcome (ORR).
The prediction score was corrected for differences in dose by multiplying with the dose (50–800 mg) and dividing by 518. The grey mid-line shows the prespecified cut-off score of 50 used to divide patients into sensitive and not sensitive (a). Overall survival of the two groups of patients predicted sensitive or resistant (b).

References

    1. Curtin NJ. DNA repair dysregulation from cancer driver to therapeutic target. Nat. Rev. Cancer. 2012;12:801–817. doi: 10.1038/nrc3399.
    1. Rouleau M, Patel A, Hendzel MJ, Kaufmann SH, Poirier GG. PARP inhibition: PARP1 and beyond. Nat. Rev. Cancer. 2010;10:293–301. doi: 10.1038/nrc2812.
    1. Riccio AA, Cingolani G, Pascal JM. PARP-2 domain requirements for DNA damage-dependent activation and localization to sites of DNA damage. Nucleic Acids Res. 2016;44:1691–1702. doi: 10.1093/nar/gkv1376.
    1. Yelamos J, Farres J, Llacuna L, Ampurdanes C, Martin-Caballero J. PARP-1 and PARP-2: New players in tumour development. Am. J. Cancer Res. 2011;1:328–346.
    1. Davar D, Beumer JH, Hamieh L, Tawbi H. Role of PARP inhibitors in cancer biology and therapy. Curr. Med Chem. 2012;19:3907–3921. doi: 10.2174/092986712802002464.
    1. Konecny GE, Kristeleit RS. PARP inhibitors for BRCA1/2-mutated and sporadic ovarian cancer: current practice and future directions. Br. J. Cancer. 2016;115:1157–1173. doi: 10.1038/bjc.2016.311.
    1. Césaire M, Thariat J, Candéias SM, Stefan D, Saintigny Y, Chevalier F. Combining PARP inhibition, radiation, and immunotherapy: a possible strategy to improve the treatment of cancer? Int J. Mol. Sci. 2018;19:E3793. doi: 10.3390/ijms19123793.
    1. Shen Y, Aoyagi-Scharber M, Wang B. Trapping poly(ADP-ribose) polymerase. J. Pharm. Exp. Ther. 2015;353:446–457. doi: 10.1124/jpet.114.222448.
    1. Chalmers AJ. The potential role and application of PARP inhibitors in cancer treatment. Br. Med Bull. 2009;89:23–40. doi: 10.1093/bmb/ldp005.
    1. Lynparza (olaparib) [prescribing information]. Wilmington, DE: AstraZeneca Pharmaceuticals LP, 2017.
    1. Rubraca (rucaparib) [prescribing information]. Boulder, CO: Clovis Oncology, Inc., 2016.
    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. doi: 10.1200/JCO.2014.56.2728.
    1. Shapira-Frommer R, Oza AM, Domchek SM, Balmaña J, Patel MR, Drew LC, et al. A phase II open-label, multicenter study of single-agent rucaparib in the treatment of patients with relapsed ovarian cancer and a deleterious BRCA mutation [abstract] J. Clin. Oncol. 2015;33(15 Suppl):5513. doi: 10.1200/jco.2015.33.15_suppl.5513.
    1. Zejula (niraparib) [prescribing information]. (Tesaro, Inc., Waltham, MA, 2019).
    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. doi: 10.1056/NEJMoa1611310.
    1. Moore K, Colombo N, Scambia G, Kim BG, Oaknin A, Friedlander M, 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. McGonigle S, Chen Z, Wu J, Chang P, Kolber-Simonds D, Ackermann K, et al. E7449: A dual inhibitor of PARP1/2 and tankyrase1/2 inhibits growth of DNA repair deficient tumors and antagonizes Wnt signaling. Oncotarget. 2015;6:41307–41323. doi: 10.18632/oncotarget.5846.
    1. McGonigle S, Chen Z, Wu J, Ackermann K, Tendyke K, Moniz G, et al. E7449: A novel PARP inhibitor enhances the efficacy of radiotherapy and chemotherapy and has potent single agent anticancer activity in BRCA-deficient tumors [abstract] Cancer Res. 2012;72(8 Suppl):Abstract 4688.
    1. Wang DD, Li C, Sun W, Zhang S, Shalinsky DR, Kern KA, et al. PARP activity in peripheral blood lymphocytes as a predictive biomarker for PARP inhibition in tumor tissues - A population pharmacokinetic/pharmacodynamic analysis of rucaparib. Clin. Pharm. Drug Dev. 2015;4:89–98. doi: 10.1002/cpdd.176.
    1. Jusko WJ, Ko HC. Physiologic indirect response models characterize diverse types of pharmacodynamic effects. Clin. Pharm. Ther. 1994;56:406–419. doi: 10.1038/clpt.1994.155.
    1. Buhl ASK, Christensen TD, Christensen IJ, Nelausen KM, Balslev E, Knoop AS, et al. Predicting efficacy of epirubicin by a multigene assay in advanced breast cancer within a Danish Breast Cancer Cooperative Group (DBCG) cohort: a retrospective-prospective blinded study. Breast Cancer Res. Treat. 2018;172:391–400. doi: 10.1007/s10549-018-4918-4.
    1. Buhl IK, Santoni-Rugiu E, Ravn J, Hansen A, Christensen IJ, Jensen T, et al. Molecular prediction of adjuvant cisplatin efficacy in Non-Small Cell Lung Cancer (NSCLC)-validation in two independent cohorts. PLoS ONE. 2018;13:e0194609. doi: 10.1371/journal.pone.0194609.
    1. Karn T, Pusztai L, Holtrich U, Iwamoto T, Shiang CY, Schmidt M, et al. Homogeneous datasets of triple negative breast cancers enable the identification of novel prognostic and predictive signatures. PLoS ONE. 2011;6:e28403. doi: 10.1371/journal.pone.0028403.
    1. Evans T, Matulonis U. PARP inhibitors in ovarian cancer: evidence, experience and clinical potential. Ther. Adv. Med. Oncol. 2017;9:253–267. doi: 10.1177/1758834016687254.
    1. Kristeleit R, Shapiro GI, Burris HA, Oza AM, LoRusso P, Patel MR, et al. A phase I-II study of the oral PARP inhibitor rucaparib in patients with germline BRCA1/2-mutated ovarian carcinoma or other solid tumors. Clin. Cancer Res. 2017;23:4095–4106. doi: 10.1158/1078-0432.CCR-16-2796.
    1. Dulaney C, Marcrom S, Stanley J, Yang ES. Poly(ADP-ribose) polymerase activity and inhibition in cancer. Semin Cell Dev. Biol. 2017;63:144–153. doi: 10.1016/j.semcdb.2017.01.007.
    1. O’Sullivan Coyne G, Chen AP, Meehan R, Doroshow JH. PARP inhibitors in reproductive system cancers: current use and developments. Drugs. 2017;77:113–130. doi: 10.1007/s40265-016-0688-7.
    1. Coleman RL, Sill MW, Bell-McGuinn K, Aghajanian C, Gray HJ, Tewari KS, et al. A phase II evaluation of the potent, highly selective PARP inhibitor veliparib in the treatment of persistent or recurrent epithelial ovarian, fallopian tube, or primary peritoneal cancer in patients who carry a germline BRCA1 or BRCA2 mutation - An NRG Oncology/Gynecologic Oncology Group study. Gynecol. Oncol. 2015;137:386–391. doi: 10.1016/j.ygyno.2015.03.042.
    1. Puhalla S, Beumer JH, Pahuja S, Appleman LJ, Tawbi HA, Stoller RG, et al. Final results of a phase 1 study of single-agent veliparib (V) in patients (pts) with either BRCA1/2-mutated cancer (BRCA+), platinum-refractory ovarian, or basal-like breast cancer (BRCA-wt) [abstract] J. Clin. Oncol. 2014;32(15 Suppl):2570. doi: 10.1200/jco.2014.32.15_suppl.2570.
    1. Murai J, Huang SY, Renaud A, Zhang Y, Ji J, Takeda S, et al. Stereospecific PARP trapping by BMN 673 and comparison with olaparib and rucaparib. Mol. Cancer Ther. 2014;13:433–443. doi: 10.1158/1535-7163.MCT-13-0803.
    1. Jones, P., Altamura, S., Boueres, J., Ferrigno, F., Fonsi, M., Giomini, C. et al. Discovery of 2-{4-[(3S)-piperidin-3-yl]phenyl}-2H-indazole-7-carboxamide (MK-4827): a novel oral poly(ADP-ribose)polymerase (PARP) inhibitor efficacious in BRCA-1 and -2 mutant tumors. J. Med. Chem.52, 7170–7185 (2009)..
    1. Menear, K. A., Adcock, C., Boulter, R., Cockcroft, X. L., Copsey, L., Cranston, A. et al. 4-[3-(4-cyclopropanecarbonylpiperazine-1-carbonyl)-4-fluorobenzyl]-2H-phthalazin- 1-one: a novel bioavailable inhibitor of poly(ADP-ribose) polymerase-1. J. Med. Chem.51, 6581–6591 (2008)..
    1. Mizutani A, Yashiroda Y, Muramatsu Y, Yoshida H, Chikada T, Tsumura T, et al. RK-287107, a potent and specific tankyrase inhibitor, blocks colorectal cancer cell growth in a preclinical model. Cancer Sci. 2018;109:4003–4014. doi: 10.1111/cas.13805.
    1. Li C, Zheng X, Han Y, Lv Y, Lan F, Zhao J. XAV939 inhibits the proliferation and migration of lung adenocarcinoma A549 cells through the WNT pathway. Oncol. Lett. 2018;15:8973–8982.
    1. Tentori L, Lacal PM, Muzi A, Dorio AS, Leonetti C, Scarsella M, et al. Poly(ADP-ribose) polymerase (PARP) inhibition or PARP-1 gene deletion reduces angiogenesis. Eur. J. Cancer. 2007;43:2124–2133. doi: 10.1016/j.ejca.2007.07.010.
    1. Galbis-Martínez M, Saenz L, Ramírez P, Parrilla P, Yélamos J. Poly(ADP-ribose) polymerase-1 modulates interferon-gamma-inducible protein (IP)-10 expression in murine embryonic fibroblasts by stabilizing IP-10 mRNA. Mol. Immunol. 2010;47:1492–1499. doi: 10.1016/j.molimm.2010.01.022.
    1. Cabrera R, Ararat M, Eksioglu EA, Cao M, Xu Y, Wasserfall C, et al. Influence of serum and soluble CD25 (sCD25) on regulatory and effector T-cell function in hepatocellular carcinoma. Scand. J. Immunol. 2010;72:293–301. doi: 10.1111/j.1365-3083.2010.02427.x.
    1. Liao W, Lin JX, Leonard WJ. Interleukin-2 at the crossroads of effector responses, tolerance, and immunotherapy. Immunity. 2013;38:13–25. doi: 10.1016/j.immuni.2013.01.004.
    1. Zhang P, Maruyama T, Konkel JE, Abbatiello B, Zamarron B, Wang ZQ, et al. PARP-1 controls immunosuppressive function of regulatory T cells by destabilizing Foxp3. PLoS ONE. 2013;8:e71590. doi: 10.1371/journal.pone.0071590.
    1. Luo X, Nie J, Wang S, Chen Z, Chen W, Li D, et al. Poly(ADP-ribosyl)ation of FOXP3 protein mediated by PARP-1 protein regulates the function of regulatory T cells. J. Biol. Chem. 2015;290:28675–28682. doi: 10.1074/jbc.M115.661611.
    1. Nasta F, Laudisi F, Sambucci M, Rosado MM, Pioli C. Increased Foxp3+ regulatory T cells in poly(ADP-Ribose) polymerase-1 deficiency. J. Immunol. 2010;184:3470–3477. doi: 10.4049/jimmunol.0901568.
    1. Ahmad SF, Zoheir KM, Bakheet SA, Ashour AE, Attia SM. Poly(ADP-ribose) polymerase-1 inhibitor modulates T regulatory and IL-17 cells in the prevention of adjuvant induced arthritis in mice model. Cytokine. 2014;68:76–85. doi: 10.1016/j.cyto.2014.04.006.

Source: PubMed

3
Tilaa