A phase I study of pexidartinib, a colony-stimulating factor 1 receptor inhibitor, in Asian patients with advanced solid tumors

Jih-Hsiang Lee, Tom Wei-Wu Chen, Chih-Hung Hsu, Yu-Hsin Yen, James Chih-Hsin Yang, Ann-Lii Cheng, Shun-Ichi Sasaki, LiYin Lillian Chiu, Masahiro Sugihara, Tomoko Ishizuka, Toshihiro Oguma, Naoyuki Tajima, Chia-Chi Lin, Jih-Hsiang Lee, Tom Wei-Wu Chen, Chih-Hung Hsu, Yu-Hsin Yen, James Chih-Hsin Yang, Ann-Lii Cheng, Shun-Ichi Sasaki, LiYin Lillian Chiu, Masahiro Sugihara, Tomoko Ishizuka, Toshihiro Oguma, Naoyuki Tajima, Chia-Chi Lin

Abstract

Background Pexidartinib, a novel, orally administered small-molecule tyrosine kinase inhibitor, has strong selectivity against colony-stimulating factor 1 receptor. This phase I, nonrandomized, open-label multiple-dose study evaluated pexidartinib safety and efficacy in Asian patients with symptomatic, advanced solid tumors. Materials and Methods Patients received pexidartinib: cohort 1, 600 mg/d; cohort 2, 1000 mg/d for 2 weeks, then 800 mg/d. Primary objectives assessed pexidartinib safety and tolerability, and determined the recommended phase 2 dose; secondary objectives evaluated efficacy and pharmacokinetic profile. Results All 11 patients (6 males, 5 females; median age 64, range 23-82; cohort 1 n = 3; cohort 2 n = 8) experienced at least one treatment-emergent adverse event; 5 experienced at least one grade ≥ 3 adverse event, most commonly (18%) for each of the following: increased aspartate aminotransferase, blood alkaline phosphatase, gamma-glutamyl transferase, and anemia. Recommended phase 2 dose was 1000 mg/d for 2 weeks and 800 mg/d thereafter. Pexidartinib exposure, area under the plasma concentration-time curve from zero to 8 h (AUC0-8h), and maximum observed plasma concentration (Cmax) increased on days 1 and 15 with increasing pexidartinib doses, and time at Cmax (Tmax) was consistent throughout all doses. Pexidartinib exposure and plasma levels of adiponectin and colony-stimulating factor 1 increased following multiple daily pexidartinib administrations. One patient (13%) with tenosynovial giant cell tumor showed objective tumor response. Conclusions This was the first study to evaluate pexidartinib in Asian patients with advanced solid tumors. Pexidartinib was safe and tolerable in this population at the recommended phase 2 dose previously determined for Western patients (funded by Daiichi Sankyo; clinicaltrials.gov number, NCT02734433).

Keywords: Pexidartinib; Pharmacokinetics; Safety; Solid tumors; Tenosynovial giant cell tumor.

Conflict of interest statement

Ann-Lii Cheng has received remuneration as an officer or advisor from Bristol-Myers Squibb, Ono, Bayer, Merck Sharp & Dohme, Novartis, and Eisai Corporation and has received honoraria from Novartis, Eli Lilly, AstraZeneca, and Eisai. Shun-ichi Sasaki, LiYin (Lillian) Chiu, Masahiro Sugihara, Tomoko Ishizuka, Toshihiro Oguma, and Naoyuki Tajima have received remuneration as an officer or advisor from Daiichi Sankyo. The other authors have no conflicts of interest.

Figures

Fig. 1
Fig. 1
Schematic of overall study design and plan. DLT, dose-limiting toxicity; MTD, maximum tolerated dose; SMC, safety monitoring committee
Fig. 2
Fig. 2
CONSORT diagram
Fig. 3
Fig. 3
Percentage change in sum of longest diameters of target lesions from baseline. *The stable disease of this patient is only 53 days after first dosing date, so cannot be considered best overall response. Instead, this patient is classified as having progressive disease
Fig. 4
Fig. 4
Longest diameter for right wrist synovial cavity by magnetic resonance imaging
Fig. 5
Fig. 5
Mean (±STD) plasma concentrations of pexidartinib versus time—day 1. Cohorts 1 and 2: linear (a) and semi-logarithmic (b) scales
Fig. 6
Fig. 6
Mean (±STD) plasma concentrations of pexidartinib versus time—day 15. Cohorts 1 and 2: linear (a) and semi-logarithmic (b) scales
Fig. 7
Fig. 7
Mean (±STD) plasma concentrations of ZAAD-1006a versus time—day 1. Cohorts 1 and 2: linear (a) and semi-logarithmic (b) scales
Fig. 8
Fig. 8
Mean (±STD) plasma concentrations of ZAAD-1006a versus time—day 15. Cohorts 1 and 2: linear (a) and semi-logarithmic (b) scales
Fig. 9
Fig. 9
Individual CSF1 (a-b) and adiponectin (c-d) plasma levels by time. C, cycle; CSF1, colony-stimulating factor 1; D, day

References

    1. Tap WD, Wainberg ZA, Anthony SP, Ibrahim PN, Zhang C, Healey JH, Chmielowski B, Staddon AP, Cohn AL, Shapiro GI, Keedy VL, Singh AS, Puzanov I, Kwak EL, Wagner AJ, von Hoff DD, Weiss GJ, Ramanathan RK, Zhang J, Habets G, Zhang Y, Burton EA, Visor G, Sanftner L, Severson P, Nguyen H, Kim MJ, Marimuthu A, Tsang G, Shellooe R, Gee C, West BL, Hirth P, Nolop K, van de Rijn M, Hsu HH, Peterfy C, Lin PS, Tong-Starksen S, Bollag G. Structure-guided blockade of CSF1R kinase in tenosynovial giant-cell tumor. N Engl J Med. 2015;373(5):428–437. doi: 10.1056/NEJMoa1411366.
    1. Cannarile MA, Weisser M, Jacob W, Jegg AM, Ries CH, Ruttinger D. Colony-stimulating factor 1 receptor (CSF1R) inhibitors in cancer therapy. J Immunother Cancer. 2017;5(1):53. doi: 10.1186/s40425-017-0257-y.
    1. Fletcher JA, Rubin BP. KIT mutations in GIST. Curr Opin Genet Dev. 2007;17(1):3–7. doi: 10.1016/j.gde.2006.12.010.
    1. Tam WF, Gary Gilliland D. Can FLT3 inhibitors overcome resistance in AML? Best Pract Res Clin Haematol. 2008;21(1):13–20. doi: 10.1016/j.beha.2007.11.003.
    1. Toffalini F, Demoulin JB. New insights into the mechanisms of hematopoietic cell transformation by activated receptor tyrosine kinases. Blood. 2010;116(14):2429–2437. doi: 10.1182/blood-2010-04-279752.
    1. West RB, Rubin BP, Miller MA, Subramanian S, Kaygusuz G, Montgomery K, Zhu S, Marinelli RJ, de Luca A, Downs-Kelly E, Goldblum JR, Corless CL, Brown PO, Gilks CB, Nielsen TO, Huntsman D, van de Rijn M. A landscape effect in tenosynovial giant-cell tumor from activation of CSF1 expression by a translocation in a minority of tumor cells. Proc Natl Acad Sci U S A. 2006;103(3):690–695. doi: 10.1073/pnas.0507321103.
    1. Dammeijer F, Lievense LA, Kaijen-Lambers ME, van Nimwegen M, Bezemer K, Hegmans JP, van Hall T, Hendriks RW, Aerts JG. Depletion of tumor-associated macrophages with a CSF-1R kinase inhibitor enhances antitumor immunity and survival induced by DC immunotherapy. Cancer Immunol Res. 2017;5(7):535–546. doi: 10.1158/2326-6066.CIR-16-0309.
    1. Pollard JW. Macrophages define the invasive microenvironment in breast cancer. J Leukoc Biol. 2008;84(3):623–630. doi: 10.1189/jlb.1107762.
    1. Yang FC, Ingram DA, Chen S, Zhu Y, Yuan J, Li X, Yang X, Knowles S, Horn W, Li Y, Zhang S, Yang Y, Vakili ST, Yu M, Burns D, Robertson K, Hutchins G, Parada LF, Clapp DW. Nf1-dependent tumors require a microenvironment containing Nf1+/− and c-kit-dependent bone marrow. Cell. 2008;135(3):437–448. doi: 10.1016/j.cell.2008.08.041.
    1. Coffelt SB, Hughes R, Lewis CE. Tumor-associated macrophages: effectors of angiogenesis and tumor progression. Biochim Biophys Acta. 2009;1796(1):11–18.
    1. Lamprecht Björn, Walter Korden, Kreher Stephan, Kumar Raman, Hummel Michael, Lenze Dido, Köchert Karl, Bouhlel Mohamed Amine, Richter Julia, Soler Eric, Stadhouders Ralph, Jöhrens Korinna, Wurster Kathrin D, Callen David F, Harte Michael F, Giefing Maciej, Barlow Rachael, Stein Harald, Anagnostopoulos Ioannis, Janz Martin, Cockerill Peter N, Siebert Reiner, Dörken Bernd, Bonifer Constanze, Mathas Stephan. Derepression of an endogenous long terminal repeat activates the CSF1R proto-oncogene in human lymphoma. Nature Medicine. 2010;16(5):571–579. doi: 10.1038/nm.2129.
    1. Steidl C, Lee T, Shah SP, Farinha P, Han G, Nayar T, Delaney A, Jones SJ, Iqbal J, Weisenburger DD, Bast MA, Rosenwald A, Muller-Hermelink HK, Rimsza LM, Campo E, Delabie J, Braziel RM, Cook JR, Tubbs RR, Jaffe ES, Lenz G, Connors JM, Staudt LM, Chan WC, Gascoyne RD. Tumor-associated macrophages and survival in classic Hodgkin's lymphoma. N Engl J Med. 2010;362(10):875–885. doi: 10.1056/NEJMoa0905680.
    1. Staals EL, Ferrari S, Donati DM, Palmerini E. Diffuse-type tenosynovial giant cell tumour: current treatment concepts and future perspectives. Eur J Cancer. 2016;63:34–40. doi: 10.1016/j.ejca.2016.04.022.
    1. Tap William D., Gelderblom Hans, Stacchiotti Silvia, Palmerini Emanuela, Ferrari Stefano, Desai Jayesh, Bauer Sebastian, Blay Jean-Yves, Alcindor Thierry, Ganjoo Kristen N., Martin Broto Javier, Ryan Christopher W., Shuster Dale Edward, Zhang Ling, Wang Qiang, Hsu Henry, Lin Paul S., Tong Sandra, Wagner Andrew J. Final results of ENLIVEN: A global, double-blind, randomized, placebo-controlled, phase 3 study of pexidartinib in advanced tenosynovial giant cell tumor (TGCT) Journal of Clinical Oncology. 2018;36(15_suppl):11502–11502. doi: 10.1200/JCO.2018.36.15_suppl.11502.
    1. Cassier PA, Gelderblom H, Stacchiotti S, Thomas D, Maki RG, Kroep JR, van der Graaf WT, Italiano A, Seddon B, Dômont J, Bompas E, Wagner AJ, Blay JY. Efficacy of imatinib mesylate for the treatment of locally advanced and/or metastatic tenosynovial giant cell tumor/pigmented villonodular synovitis. Cancer. 2012;118(6):1649–1655. doi: 10.1002/cncr.26409.
    1. Gelderblom H, Cropet C, Chevreau C, Boyle R, Tattersall M, Stacchiotti S, Italiano A, Piperno-Neumann S, le Cesne A, Ferraresi V, Penel N, Duffaud F, Cassier P, Toulmonde M, Casali P, Taieb S, Guillemaut S, Metzger S, Pérol D, Blay JY. Nilotinib in locally advanced pigmented villonodular synovitis: a multicentre, open-label, single-arm, phase 2 trial. Lancet Oncol. 2018;19(5):639–648. doi: 10.1016/S1470-2045(18)30143-8.

Source: PubMed

3
Tilaa