A phase 1 dose-escalation study of veliparib with bimonthly FOLFIRI in patients with advanced solid tumours

Jordan Berlin, Ramesh K Ramanathan, John H Strickler, Deepa S Subramaniam, John Marshall, Yoon-Koo Kang, Robert Hetman, Matthew W Dudley, Jiewei Zeng, Caroline Nickner, Hao Xiong, Philip Komarnitsky, Stacie Peacock Shepherd, Herbert Hurwitz, Heinz-Josef Lenz, Jordan Berlin, Ramesh K Ramanathan, John H Strickler, Deepa S Subramaniam, John Marshall, Yoon-Koo Kang, Robert Hetman, Matthew W Dudley, Jiewei Zeng, Caroline Nickner, Hao Xiong, Philip Komarnitsky, Stacie Peacock Shepherd, Herbert Hurwitz, Heinz-Josef Lenz

Abstract

Background: Veliparib is a potent poly(ADP-ribose) polymerase inhibitor. This phase 1 study aimed to establish the maximum tolerated dose (MTD) and recommended phase 2 dose (RP2D) of veliparib combined with various FOLFIRI regimens in patients with solid tumours.

Methods: Patients received veliparib (10-270 mg BID, days 1-5, 15-19) and FOLFIRI (days 1-3, 15-17) in three regimens containing 5-fluorouracil 2,400 mg/m2: irinotecan 150 mg/m2 and folinic acid 400 mg/m2 (part 1); irinotecan 180 mg/m2, folinic acid 400 mg/m2, and 5-fluorouracil 400 mg/m2 bolus (part 2), or irinotecan 180 mg/m2 (part 3). The RP2D was further evaluated in safety expansion cohorts. Preliminary antitumour activity was also assessed.

Results: Ninety-two patients received ≥1 veliparib dose. MTD was not reached; RP2D was set at 200 mg BID veliparib plus FOLFIRI (without 5-fluorouracil bolus). Most common treatment-emergent adverse events were neutropenia (66.3%), diarrhoea, and nausea (60.9% each). Dose-limiting toxicities (n = 4) were grade 3 gastritis and grade 4 neutropenia and febrile neutropenia. Veliparib exposure was dose-proportional, with no effects on the pharmacokinetics of FOLFIRI components. Fifteen patients had a partial response (objective response rate, 17.6%).

Conclusions: The acceptable safety profile and preliminary antitumour activity of veliparib plus FOLFIRI support further evaluation of this combination.

Trial registration: ClinicalTrials.gov NCT01123876.

Conflict of interest statement

H.-J.L., R.K.R., and J.H.S.: research funding from AbbVie. J.B.: research funding from AbbVie and AbbVie ad board. S.P.S.: former employee of AbbVie. May hold AbbVie stock or options. Holds Abbott and Corcept stocks. D.S.S.: No disclosure pertaining to this study. Advisory board member for BMS, AstraZeneca, Takeda Oncology. H.H.: research funding from AbbVie. H.X., R.H., J.Z., P.K., C.N., and M.W.D.: employed by AbbVie. May hold AbbVie stock or options. The remaining authors declare that they have no competing financial interests.

Figures

Fig. 1
Fig. 1
Study design. a Veliparib was administered in all three study parts 1 h prior to FOLFIRI infusion. b Reduced FOLFIRI = reduced dose of irinotecan 150 mg/m2 (90-min infusion) + folinic acid 400 mg/m2 (2-h infusion during irinotecan administration) + 5-FU 2,400 mg/m2 (46-h continuous infusion immediately following irinotecan administration). c Standard FOLFIRI = standard dose of irinotecan 180 mg/m2 (90-min infusion) + folinic acid 400 mg/m2 (2-h infusion during irinotecan administration) + 5-FU 400 mg/m2 (bolus immediately following irinotecan administration) and 2,400 mg/m2 (46-h continuous infusion). d Modified FOLFIRI = modified dose of irinotecan 180 mg/m2 (90-min infusion) + 5-FU 2,400 mg/m2 (46-h continuous infusion immediately following irinotecan administration). e The 400 mg/m2 bolus 5-FU dose was not tolerated during the first 2 weeks of cycle 1, before veliparib administration. Therefore, in part 1 of the study, only 4/67 patients received 400 mg/m2 of 5-FU bolus infusions starting cycle 2. For the remaining 63 patients, bolus administration of 5-FU was removed to reduce the toxic effects of 5-FU. Part 2 dose escalation was consequently discontinued and, although patients could continue veliparib, they were considered not evaluable. Data from part 2 were combined with those from part 3. 5-FU 5-fluorouracil, BID twice daily, DLT dose-limiting toxicity, FOLFIRI 5-fluorouracil plus folinic acid plus irinotecan, RP2D recommended phase 2 dose
Fig. 2
Fig. 2
Evaluation of veliparib dose proportionality in part 1 of the study: mean (+SD) veliparib Cmax and AUC (top) and dose-normalized Cmax and AUC (bottom) presented vs increasing doses of veliparib (measured after the first administered dose of veliparib). Numbers of patients for each veliparib BID dose presented in the figure: n = 10 for 10 mg; n = 6 for 100 mg; n = 5 for 20 mg, 40 mg, and 200 mg; n = 4 for 80 mg and 130 mg; n = 7 for 160 mg; n = 9 for 270 mg Cmax, and n = 8 for 270 mg AUC0-8. AUC0–8 area under the plasma or serum concentration-time curve from time zero to hour 8, BID twice daily, Cmax maximum plasma concentration, SD standard deviation
Fig. 3
Fig. 3
Kaplan–Meier curves for time to disease progression in a, all dosed patients, and b, expanded safety cohort. The number and percentage of patients with disease progression and the median number of days (and 95% CIs) to disease progression are detailed per cancer type. Censored events are depicted within each graph, and the numbers of patients at risk per cancer type are listed for each depicted timepoint. CI confidence interval, NR not reached

References

    1. Chiarugi A. Poly(ADP-ribose) polymerase: killer or conspirator? The ‘suicide hypothesis’ revisited. Trends Pharmacol. Sci. 2002;23:122–129. doi: 10.1016/S0165-6147(00)01902-7.
    1. Virág L, Szabó C. The therapeutic potential of poly(ADP-ribose) polymerase inhibitors. Pharmacol. Rev. 2002;54:375–429. doi: 10.1124/pr.54.3.375.
    1. Nomura F, et al. Enhancement of poly-adenosine diphosphate-ribosylation in human hepatocellular carcinoma. J. Gastroenterol. Hepatol. 2000;15:529–535. doi: 10.1046/j.1440-1746.2000.02193.x.
    1. Ossovskaya V, Koo IC, Kaldjian EP, Alvares C, Sherman BM. Upregulation of poly (ADP-ribose) polymerase-1 (PARP1) in triple-negative breast cancer and other primary human tumour types. Genes Cancer. 2010;1:812–821. doi: 10.1177/1947601910383418.
    1. Curtin NJ, et al. Novel poly(ADP-ribose) polymerase-1 inhibitor, AG14361, restores sensitivity to temozolomide in mismatch repair-deficient cells. Clin. Cancer Res. 2004;10:881–889. doi: 10.1158/1078-0432.CCR-1144-3.
    1. Fong PC, et al. Inhibition of poly(ADP-ribose) polymerase in tumours from BRCA mutation carriers. N. Engl. J. Med. 2009;361:123–134. doi: 10.1056/NEJMoa0900212.
    1. Kaufman B, 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. Balmaña J, et al. Phase I trial of olaparib in combination with cisplatin for the treatment of patients with advanced breast, ovarian and other solid tumours. Ann. Oncol. 2014;25:1656–1663. doi: 10.1093/annonc/mdu187.
    1. Kummar S, et al. A phase I study of veliparib in combination with metronomic cyclophosphamide in adults with refractory solid tumours and lymphomas. Clin. Cancer Res. 2012;18:1726–1734. doi: 10.1158/1078-0432.CCR-11-2821.
    1. Czito BG, et al. The safety and tolerability of veliparib (V) plus capecitabine (C) and radiation (RT) in subjects with locally advanced rectal cancer: results of a phase 1b study. J. Clin. Oncol. 2014;32:abstr. 3634. doi: 10.1200/jco.2014.32.15_suppl.3634.
    1. Davidson D, Wang Y, Aloyz R, Panasci L. The PARP inhibitor ABT-888 synergizes irinotecan treatment of colon cancer cell lines. Invest. New Drugs. 2013;31:461–468. doi: 10.1007/s10637-012-9886-7.
    1. Tahara M, et al. The use of olaparib (AZD2281) potentiates SN-38 cytotoxicity in colon cancer cells by indirect inhibition of Rad51-mediated repair of DNA double-strand breaks. Mol. Cancer Ther. 2014;13:1170–1180. doi: 10.1158/1535-7163.MCT-13-0683.
    1. Ba X, Garg NJ. Signaling mechanism of poly(ADP-ribose) polymerase-1 (PARP-1) in inflammatory diseases. Am. J. Pathol. 2011;178:946–955. doi: 10.1016/j.ajpath.2010.12.004.
    1. Das SK, et al. Poly(ADP-ribose) polymers regulate DNA topoisomerase I (Top1) nuclear dynamics and camptothecin sensitivity in living cells. Nucleic Acids Res. 2016;44:8363–8375. doi: 10.1093/nar/gkw665.
    1. Murai, J. et al. Tyrosyl-DNA phosphodiesterase 1 (TDP1) repairs DNA damage induced by topoisomerases I and II and base alkylation in vertebrate cells. J. Biol. Chem. 287, 12848–12857 (2012).
    1. Pommier Y. Drugging topoisomerases: lessons and challenges. ACS Chem. Biol. 2013;8:82–95. doi: 10.1021/cb300648v.
    1. Das BB, et al. PARP1-TDP1 coupling for the repair of topoisomerase I-induced DNA damage. Nucleic Acids Res. 2014;42:4435–4449. doi: 10.1093/nar/gku088.
    1. Pommier Y, et al. Repair of topoisomerase I-mediated DNA damage. Prog. Nucleic Acid Res. Mol. Biol. 2006;81:179–229. doi: 10.1016/S0079-6603(06)81005-6.
    1. Smith LM, Willmore E, Austin CA, Curtin NJ. The novel poly(ADP-ribose) polymerase inhibitor, AG14361, sensitizes cells to topoisomerase I poisons by increasing the persistence of DNA strand breaks. Clin. Cancer Res. 2005;11:8449–8457. doi: 10.1158/1078-0432.CCR-05-1224.
    1. Santi DV, McHenry CS, Dommer H. Mechanism of interaction of thymidylate synthase with 5-fluorodeoxyuridylate. Biochemistry. 1974;13:471–481. doi: 10.1021/bi00700a012.
    1. Sommer H, Santi DV. Purification and amino acid analysis of an active site peptide from tymidylate synthetase containing covalently bound 5-fluoro-2′-deoxyuridylate and methylenetetrahydrofolate. Res. Commun. 1974;57:689–695.
    1. Guglielmi AP, Sobrero AF. Second-line therapy for advanced colorectal cancer. Gastrointest. Cancer Res. 2007;1:57–63.
    1. Kirstein MM, et al. Targeted therapies in metastatic colorectal cancer: a systematic review and assessment of currently available data. Oncologist. 2014;19:1156–1168. doi: 10.1634/theoncologist.2014-0032.
    1. Donawho CK, et al. ABT-888, an orally active poly(ADP-ribose) polymerase inhibitor that potentiates DNA-damaging agents in preclinical tumour models. Clin. Cancer Res. 2007;13:2728–2737. doi: 10.1158/1078-0432.CCR-06-3039.
    1. Tuli R. et al. Radiosensitization of pancreatic cancer cells in vitro and in vivo through poly (ADP-ribose) polymerase inhibition with ABT-888. Transl. Oncol.10.1016/j.tranon.2014.04.003 (2014).
    1. Shelton JW, et al. In vitro and in vivo enhancement of chemoradiation using the oral PARP inhibitor ABT-888 in colorectal cancer cells. Int. J. Radiat. Oncol. Biol. Phys. 2013;86:469–476. doi: 10.1016/j.ijrobp.2013.02.015.
    1. Falzacappa MV, et al. The combination of the PARP inhibitor rucaparib and 5FU is an effective strategy for treating acute leukemias. Mol. Cancer Ther. 2015;14:889–898. doi: 10.1158/1535-7163.MCT-14-0276.
    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. 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. Pujade-Lauraine E, et al. Olaparib tablets as maintenance therapy in patients with platinum-sensitive, relapsed ovarian cancer and a BRCA1/2 mutation (SOLO2/ENGOT-Ov21): a double-blind, randomised, placebo-controlled, phase 3 trial. Lancet. 2017;18:1274–1284. doi: 10.1016/S1470-2045(17)30469-2.
    1. Ledermann J, et al. Olaparib maintenance therapy in platinum-sensitive relapsed ovarian cancer. N. Engl. J. Med. 2012;366:1382–1392. doi: 10.1056/NEJMoa1105535.
    1. Oza AM, et al. Olaparib combined with chemotherapy for recurrent platinum-sensitive ovarian cancer: a randomised phase 2 trial. Lancet Oncol. 2015;16:87–97. doi: 10.1016/S1470-2045(14)71135-0.
    1. Mirza MR, et al. Niraparib maintenance therapy in platinum-sensitive, recurrent ovarian cancer. N. Engl. J. Med. 2016;375:2154–2164. doi: 10.1056/NEJMoa1611310.
    1. Coleman RL, 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. Werner TL, et al. Veliparib (ABT-888) extended-release formulations: a phase 1 study on safety, pharmacokinetics (PK), and bioavailability in patients with advanced solid tumours. J. Clin. Oncol. 2016;34:abstr. 2579. doi: 10.1200/JCO.2016.34.15_suppl.2579.
    1. Middleton MR, et al. Randomized phase II study evaluating veliparib (ABT-888) with temozolomide in patients with metastatic melanoma. Ann. Oncol. 2015;26:2173–2179. doi: 10.1093/annonc/mdv308.
    1. Pietanza MC, et al. A multi-center, randomized, double-blind phase II study comparing temozolomide (TMZ) plus either veliparib (ABT-888), a PARP inhibitor, or placebo as 2nd or 3rd-line therapy for patients (Pts) with relapsed small cell lung cancers (SCLCs) J. Clin. Oncol. 2016;34:abstr. 8512. doi: 10.1200/JCO.2016.34.15_suppl.8512.
    1. Kunos C, et al. A phase I-II evaluation of veliparib (NSC #737664), topotecan, and filgrastim or pegfilgrastim in the treatment of persistent or recurrent carcinoma of the uterine cervix: an NRG Oncology/Gynecologic Oncology Group study. Int. J. Gynecol. Cancer. 2015;25:484–492. doi: 10.1097/IGC.0000000000000380.
    1. Isakoff SJ, et al. A randomized phase II study of veliparib with temozolomide or carboplatin/paclitaxel versus placebo with carboplatin/paclitaxel in BRCA1/2 metastatic breast cancer: design and rationale. Future Oncol. 2017;13:307–320. doi: 10.2217/fon-2016-0412.
    1. LoRusso PM, et al. Phase I safety, pharmacokinetic, and pharmacodynamic study of the poly(ADP-ribose) polymerase (PARP) inhibitor veliparib (ABT-888) in combination with irinotecan in patients with advanced solid tumours. Clin. Cancer Res. 2016;22:3227–3232. doi: 10.1158/1078-0432.CCR-15-0652.
    1. LoRusso PM, et al. Combination of the PARP inhibitor veliparib (ABT888) with irinotecan (CPT-11) in patients with triple negative breast cancer: preliminary activity and signature of response. Cancer Res. 2015;75:abstr. CT325. doi: 10.1158/1538-7445.AM2015-CT325.
    1. Hwang JJ, Eisenberg SG, Marshall JL. Improving the toxicity of irinotecan/5-FU/leucovorin: a 21-day schedule. Oncology. 2003;17:37–43.
    1. Dizdar O, Arslan C, Altundag K. Advances in PARP inhibitors for the treatment of breast cancer. Expert Opin. Pharmacother. 2015;16:2751–2758. doi: 10.1517/14656566.2015.1100168.
    1. Liu X, et al. Acquired resistance to combination treatment with temozolomide and ABT-888 is mediated by both base excision repair and homologous recombination DNA repair pathways. Mol. Cancer Res. 2009;7:1686–1692. doi: 10.1158/1541-7786.MCR-09-0299.
    1. Murai J, et al. Trapping of PARP1 and PARP2 by clinical PARP inhibitors. Cancer Res. 2012;72:5588–5599. doi: 10.1158/0008-5472.CAN-12-2753.
    1. Stordal B, et al. BRCA1/2 mutation analysis in 41 ovarian cell lines reveals only one functionally deleterious BRCA1 mutation. Mol. Oncol. 2013;7:567–579. doi: 10.1016/j.molonc.2012.12.007.

Source: PubMed

3
Tilaa