Phase 1 dose-finding study of rebastinib (DCC-2036) in patients with relapsed chronic myeloid leukemia and acute myeloid leukemia

Jorge Cortes, Moshe Talpaz, Hedy P Smith, David S Snyder, Jean Khoury, Kapil N Bhalla, Javier Pinilla-Ibarz, Richard Larson, David Mitchell, Scott C Wise, Thomas J Rutkoski, Bryan D Smith, Daniel L Flynn, Hagop M Kantarjian, Oliver Rosen, Richard A Van Etten, Jorge Cortes, Moshe Talpaz, Hedy P Smith, David S Snyder, Jean Khoury, Kapil N Bhalla, Javier Pinilla-Ibarz, Richard Larson, David Mitchell, Scott C Wise, Thomas J Rutkoski, Bryan D Smith, Daniel L Flynn, Hagop M Kantarjian, Oliver Rosen, Richard A Van Etten

Abstract

A vailable tyrosine kinase inhibitors for chronic myeloid leukemia bind in an adenosine 5'-triphosphate-binding pocket and are affected by evolving mutations that confer resistance. Rebastinib was identified as a switch control inhibitor of BCR-ABL1 and FLT3 and may be active against resistant mutations. A Phase 1, first-in-human, single-agent study investigated rebastinib in relapsed or refractory chronic or acute myeloid leukemia. The primary objectives were to investigate the safety of rebastinib and establish the maximum tolerated dose and recommended Phase 2 dose. Fifty-seven patients received treatment with rebastinib. Sixteen patients were treated using powder-in-capsule preparations at doses from 57 mg to 1200 mg daily, and 41 received tablet preparations at doses of 100 mg to 400 mg daily. Dose-limiting toxicities were dysarthria, muscle weakness, and peripheral neuropathy. The maximum tolerated dose was 150 mg tablets administered twice daily. Rebastinib was rapidly absorbed. Bioavailability was 3- to 4-fold greater with formulated tablets compared to unformulated capsules. Eight complete hematologic responses were achieved in 40 evaluable chronic myeloid leukemia patients, 4 of which had a T315I mutation. None of the 5 patients with acute myeloid leukemia responded. Pharmacodynamic analysis showed inhibition of phosphorylation of substrates of BCR-ABL1 or FLT3 by rebastinib. Although clinical activity was observed, clinical benefit was insufficient to justify continued development in chronic or acute myeloid leukemia. Pharmacodynamic analyses suggest that other kinases inhibited by rebastinib, such as TIE2, may be more relevant targets for the clinical development of rebastinib (clinicaltrials.gov Identifier:00827138).

Trial registration: ClinicalTrials.gov NCT00827138.

Copyright© Ferrata Storti Foundation.

Figures

Figure 1.
Figure 1.
Mean plasma rebastinib concentration-time profile following multiple-dose administration of rebastinib tablets to steady state. Peripheral blood samples were obtained at the indicated times following administration of an oral dose of rebastinib (tablet formulation at doses of 100 mg BID, 150 mg BID, and 200 mg BID) in patients who had received at least 1 week of continuous dosing. Plasma concentrations of rebastinib were determined by LC-MS/MS as described in Methods. IC80 values for inhibition of BCR-ABL1 phosphorylation of CRKL in ex vivo assays using whole blood from CML patients with BCR-ABL1 or BCR-ABL1 T315I are indicated. BID: twice daily; Conc: concentration; pCRKL: phosphorylated CT10 Regulator of Kinase-like.
Figure 2.
Figure 2.
Pharmacodynamic analyses of pCRKL and pSTAT5 inhibition pre- and post-rebastinib dose. Serial peripheral blood samples were obtained from patients following the first dose of rebastinib (Cycle 1 Day 1) and on Day 8 of continuous twice daily dosing (Cycle 1 Day 8). Mononuclear cells were isolated, lysed, and the indicated proteins and phosphoproteins analyzed by immunoblotting as described in Methods. (A) Patient with relapsed chronic phase CML BCR-ABL1 V299L mutation who demonstrated >75% inhibition of pCRKL 4h following the Day 8 morning dose. (B) Patient with relapsed chronic phase CML and BCR-ABL1 T315I mutation who demonstrated >90% inhibition of pSTAT5 at 1h following the Day 8 morning dose. In this patient, the degree of inhibition of pCRKL was less pronounced (~25%). PK: pharmacokinetics; pCRKL: phosphorylated CT10 regulator of kinase-like; CRKL: CT10 regulator of kinase-like; eIF4E: eukaryotic translation initiation factor 4E; pSTAT5: phosphorylated signal transducer and activator of transcription 5; STAT5: signal transducer and activator of transcription 5.

References

    1. Lugo TG, Pendergast AM, Muller AJ, Witte ON. Tyrosine kinase activity and transformation potency of bcr-abl oncogene products. Science. 1990;247(4946):1079–1082.
    1. Kantarjian HM, Talpaz M, O’Brien S, et al. Imatinib mesylate for Philadelphia Chromosome-positive, chronic-phase myeloid leukemia after failure of interferon: follow-up results. Clin Cancer Res. 2002;8(7):2177–2187.
    1. Kantarjian H, Giles F, Wunderle L, et al. Nilotinib in imatinib-resistant CML and Philadelphia chromosome-positive ALL. N Engl J Med. 2006;354(24):2542–2551.
    1. Nakao M, Yokota S, Iwai T, et al. Internal tandem duplication of the flt3 gene found in acute myeloid leukemia. Leukemia. 1996;10(12):1911–1918.
    1. Schiller GJ, Tallman MS, Goldberg SL, et al. Final results of a randomized phase 2 study showing the clinical benefit of quizartinib (AC220) in patients with FLT3-ITD positive relapsed or refractory acute myeloid leukemia. J Clin Oncol. 2014;32:5s, (suppl;abstr 7100).
    1. Wander SA, Levis MJ, Fathi AT. The evolving role of FLT3 inhibitors in acute myeloid leukemia: quizartinib and beyond. Ther Adv Hematol. 2014;5(3):65–77.
    1. Röllig C, Müller-Tidow C, Hüttmann A, et al. Addition of sorafenib versus placebo to standard therapy in patients aged 60 years or younger with newly diagnosed acute myeloid leukemia (SORAML): a multicentre, phase 2, randomised controlled trial. Lancet Oncol. 2015;16(16):1691–1699.
    1. Stone RM, Mandrekar S, Sanford BL, et al. The multi-kinase inhibitor midostaurin (M) prolongs survival compared with placebo (P) in combination with daunorubicin (D)/cytarabine(C) induction (Ind), high-dose C consolidation (consol), and as main tenance (maint) therapy in newly diagnosed acute myeloid leukemia (AML) patients (pts) age 18–60 with Flt3 mutations (muts): An international prospective randomized (rand) p-controlled double-blind trial (CALGB10603/RATIFY [Alliance]. Blood 2015;126(23):abstract 6.
    1. Gorre ME, Mohammed M, Ellwood K, Hsu N, Paquette R, Rao PN, Sawyers CL. Clinical resistance to STI-571 cancer therapy caused by BCR-ABL gene mutation or amplification. Science. 2001;293(5531):876–880.
    1. Shah NP, Nicoll JM, Nagar B, et al. Multiple BCR-ABL kinase domain mutations confer polyclonal resistance to the tyrosine kinase inhibitor imatinib (STI571) in chronic phase and blast crisis chronic myeloid leukemia. Cancer Cell. 2002;2(2):117 125.
    1. Cortes JE, Kim DW, Pinilla-Ibarz J, et al. A Phase 2 trial of ponatinib in Philadelphia chromosome–positive leukemias. N Engl J Med. 2013;369(19):1783–1796.
    1. Chan WW, Wise SC, Kaufman MD, et al. Conformational control inhibition of the BCR-ABL1 tyrosine kinase, including the gatekeeper T315I mutant, by the switch control inhibitor DCC-2036. Cancer Cell. 2011;19(4):556–568.
    1. Azam M, Seeliger MA, Gray NS, Kuriyan J, Daley GQ. Activation of tyrosine kinases by mutation of the gatekeeper threonine. Nat Struct Mol Biol. 2008;15(10):1109–1118.
    1. Roumiantsev S, Shah NP, Gorre ME, et al. Clinical resistance to the kinase inhibitor STI-571 in chronic myeloid leukemia by mutation of Tyr-253 in the Abl kinase domain P-loop. Proc Natl Acad Sci USA. 2002;99(16):10700–10705.
    1. Eide CA, Adrian LT, Tyner JW, et al. The ABL switch control inhibitor DCC-2036 is active against the chronic myeloid leukemia mutant BCR-ABLT315I and exhibits a narrow resistance profile. Cancer Research. 2011;71(9):3189–3195.
    1. Redaelli S, Mologni L, Rostagno R, et al. Three novel patient-derived BCR/ABL mutants show different sensitivity to second and third generation tyrosine kinase inhibitors. Am J Hematol. 2012;87(11): E125–128.
    1. National Cancer Institute Cancer Therapy Evaluation Program. Common Terminology Criteria for Adverse Events v3.0. Available from: Last accessed: 8th October 2008.
    1. NCCN Practice Guidelines in Oncology. Available from: Last accessed: 8th October 2008.
    1. Cheson BD, Bennett JM, Kopecky KJ, et al. Revised recommendations of the International Working Group for diagnosis, standardization of response criteria, treatment outcomes, and reporting standards for therapeutic trials in acute myeloid leukemia. J Clin Oncol. 2003;21(24):4642–4649.
    1. Renouf DJ, Velazquez-Martin JP, Simpson R, Siu LL, Bedard PL. Ocular toxicity of targeted therapies. J Clin Oncol. 2012; 30(26):3277–3286.
    1. Stjepanovic N, Velazquez-Martin JP, Bedard PL. Ocular toxicities of MEK inhibitors and other targeted therapies. Ann Oncol. 2016; 27(6):998–1005.
    1. Mellor HR, Bell AR, Valentin JP, Roberts RR. Cardiotoxicity associated with targeting kinase pathways in cancer. Toxicol Sci. 2011;120(1):14–32.
    1. Kerkala R, Grazette L, Yacobi R, et al. Cardiotoxicity of the cancer therapeutic agent imatinib mesylate. Nat Med. 2006; 12(8):908–916.
    1. Shah NP, Kasap C, Weier C, et al. Transient potent BCR-ABL inhibition is sufficient to commit chronic myeloid leukemia cells irreversibly to apoptosis. Cancer Cell. 2008; 14(6):485–493.
    1. Cortes JE, Kantarjian HM, Rea D, et al. Final analysis of the efficacy and safety of omacetaxine mepesuccinate in patients with chronic- or accelerated-phase chronic myeloid leukemia: Results with 24 months of follow-up. Cancer. 2015; 15;121(10): 1637–1644.
    1. DePrimo SE, Bello CL, Smeraglia J, et al. Circulating protein biomarkers of pharmacodynamic activity of sunitinib in patients with metastatic renal cell carcinoma: modulation of VEGF and VEGF-related proteins. J Transl Med. 2007;5:32.
    1. Lewis CE, Ferrara N. Multiple effects of angiopoietin-2 blockade on tumors. Cancer Cell. 2011;19(4):431–433.
    1. Hanahan D, Coussens LM. Accessories to the crime: functions of cells recruited to the tumor microenvironment. Cancer Cell. 2012. March 20;21(3):309–322.
    1. Coffelt SB, Chen YY, Muthana M, et al. Angiopoietin 2 stimulates TIE2-expressing monocytes to suppress T cell activation and to promote regulatory T cell expansion. J Immunol. 2011;186(7):4183–4190.
    1. Hughes R, Qian B-Z, Rowan C, et al. Perivascular M2 macrophages stimulate tumor relapse after chemotherapy. Cancer Res. 2015;75(17):3479–3491.
    1. Mazzieri R, Pucci F, Moi D, et al. Targeting the ANG2/TIE2 axis inhibits tumor growth and metastasis by impairing angiogenesis and disabling rebounds of proangiogenic myeloid cells. Cancer Cell. 2011;19(4):512–526.
    1. Ibberson M, Bron S, Guex N, et al. TIE2 and VEGFR kinase activities drive immunosuppressive function of TIE2-expressing monocytes in human breast cancer. Clin Cancer Res. 2013;19(13):3439–3449.
    1. Roussos ET, Goswami S, Balsamo M, et al. Mena invasive (Mena(INV)) and Mena11a isoforms play distinct roles in breast cancer cell cohesion and association with TMEM. Clin Exp Metastasis. 2011;28(6):515–527.
    1. Dovas A, Gligorijevic B, Chen X, Entenberg D, Condeelis J, Cox D. Visualization of actin polymerization in invasive structures of macrophages and carcinoma cells using photoconvertible beta-actin-Dendra2 fusion proteins. PLoS One. 2011;6(2): e16485.
    1. Harney AS, Arwert EN, Entenberg D, Wang Y, Guo P, Qian B-Z, et al. Real-time imaging reveals local, transient vascular permeability, and tumor cell intravasation stimulated by TIE2hi macrophage–derived VEGFA. Cancer Discov. 2015;5(9):932 943.

Source: PubMed

3
Abonnere