Effect of rifampin and itraconazole on the pharmacokinetics of zanubrutinib (a Bruton's tyrosine kinase inhibitor) in Asian and non-Asian healthy subjects

Song Mu, Zhiyu Tang, William Novotny, Manal Tawashi, Ta-Kai Li, Ying Ou, Srikumar Sahasranaman, Song Mu, Zhiyu Tang, William Novotny, Manal Tawashi, Ta-Kai Li, Ying Ou, Srikumar Sahasranaman

Abstract

Purpose: Zanubrutinib (BGB-3111) is a potent Bruton's tyrosine kinase inhibitor with promising clinical activity in B-cell malignancies. Zanubrutinib was shown to be mainly metabolized through cytochrome P450 3A (CYP3A) in vitro. We evaluated the effect of steady-state rifampin (a strong CYP3A inducer) and steady-state itraconazole (a strong CYP3A inhibitor) on the pharmacokinetics (PK), safety, and tolerability of zanubrutinib in healthy Asian and non-Asian subjects.

Methods: In this open-label, two-part clinical study, 20 participants received a single oral dose of zanubrutinib (320 mg) and oral rifampin (600 mg) in Part A, and 18 participants received a single oral dose of zanubrutinib (20 mg) and oral itraconazole (200 mg) in Part B. Serial blood samples were collected after administration of zanubrutinib alone and zanubrutinib in combination with rifampin or itraconazole for the measurement of PK parameters.

Results: Coadministration with rifampin decreased AUC0-∞ of zanubrutinib by 13.5-fold and Cmax by 12.6-fold. Coadministration with itraconazole increased the AUC0-∞ of zanubrutinib by 3.8-fold and Cmax by 2.6-fold. The PK of zanubrutinib was consistent between Asian and non-Asian subjects, and zanubrutinib was well tolerated in this study.

Conclusions: These results confirm that zanubrutinib is primarily metabolized by CYP3A in humans. The PK of zanubrutinib was comparable between Asian and non-Asian subjects and, therefore, no dose modifications are necessary for zanubrutinib in these ethnic populations.

Keywords: Clinical pharmacology; Clinical trials; Drug–drug interactions; Oncology; Pharmacokinetics and drug metabolism.

Conflict of interest statement

Song Mu, Zhiyu Tang, William Novotny, Manal Tawashi, Ta-Kai Li, Ying Ou, and Srikumar Sahasranaman are employees of BeiGene. The study was sponsored and funded by BeiGene, USA.

Figures

Fig. 1
Fig. 1
Arithmetic mean (+ SD) zanubrutinib plasma concentration profiles following a administration of 320 mg alone and coadministration with 600 mg rifampin or b administration of 20 mg alone and coadministration with 200 mg itraconazole. Zanubrutinib plasma concentrations on Y-axis are shown on log scale
Fig. 2
Fig. 2
Comparative box plots of area under the plasma concentration–time curve from 0 h to infinity (AUC0–∞, ng·h/mL) and maximal plasma concentration (Cmax; ng/mL) in Asian and non-Asian) subjects in a, b the absence and presence of rifampin and c, d in the absence and presence of itraconazole. The box plot represents 25th and 75th percentiles; whiskers extend to 5th and 95th percentiles. Median is indicated by a line within the box, and circles represent values for individual subject

References

    1. Ponader S, Burger JA. Bruton's tyrosine kinase: from X-linked agammaglobulinemia toward targeted therapy for B-cell malignancies. J Clin Oncol. 2014;32(17):1830–1839. doi: 10.1200/JCO.2013.53.1046.
    1. Niemann CU, Wiestner A. B-cell receptor signaling as a driver of lymphoma development and evolution. Semin Cancer Biol. 2013;23(6):410–421. doi: 10.1016/j.semcancer.2013.09.001.
    1. de Rooij MF, Kuil A, Geest CR, Eldering E, Chang BY, Buggy JJ, Pals ST, Spaargaren M. The clinically active BTK inhibitor PCI-32765 targets B-cell receptor- and chemokine-controlled adhesion and migration in chronic lymphocytic leukemia. Blood. 2012;119(11):2590–2594. doi: 10.1182/blood-2011-11-390989.
    1. Bernard S, Danglade D, Gardano L, Laguillier C, Lazarian G, Roger C, Thieblemont C, Marzec J, Gribben J, Cymbalista F, Varin-Blank N, Ledoux D, Baran-Marszak F. Inhibitors of BCR signalling interrupt the survival signal mediated by the micro-environment in mantle cell lymphoma. Int J Cancer. 2015;136(12):2761–2774. doi: 10.1002/ijc.29326.
    1. Burger JA, Tedeschi A, Barr PM, Robak T, Owen C, Ghia P, Bairey O, Hillmen P, Bartlett NL, Li J, Simpson D, Grosicki S, Devereux S, McCarthy H, Coutre S, Quach H, Gaidano G, Maslyak Z, Stevens DA, Janssens A, Offner F, Mayer J, O'Dwyer M, Hellmann A, Schuh A, Siddiqi T, Polliack A, Tam CS, Suri D, Cheng M, Clow F, Styles L, James DF, Kipps TJ, Investigators R. Ibrutinib as initial therapy for patients with chronic lymphocytic leukemia. N Engl J Med. 2015;373(25):2425–2437. doi: 10.1056/NEJMoa1509388.
    1. Byrd JC, Furman RR, Coutre SE, Flinn IW, Burger JA, Blum KA, Grant B, Sharman JP, Coleman M, Wierda WG, Jones JA, Zhao W, Heerema NA, Johnson AJ, Sukbuntherng J, Chang BY, Clow F, Hedrick E, Buggy JJ, James DF, O'Brien S. Targeting BTK with ibrutinib in relapsed chronic lymphocytic leukemia. N Engl J Med. 2013;369(1):32–42. doi: 10.1056/NEJMoa1215637.
    1. Byrd JC, Brown JR, O'Brien S, Barrientos JC, Kay NE, Reddy NM, Coutre S, Tam CS, Mulligan SP, Jaeger U, Devereux S, Barr PM, Furman RR, Kipps TJ, Cymbalista F, Pocock C, Thornton P, Caligaris-Cappio F, Robak T, Delgado J, Schuster SJ, Montillo M, Schuh A, de Vos S, Gill D, Bloor A, Dearden C, Moreno C, Jones JJ, Chu AD, Fardis M, McGreivy J, Clow F, James DF, Hillmen P, Investigators R. Ibrutinib versus ofatumumab in previously treated chronic lymphoid leukemia. N Engl J Med. 2014;371(3):213–223. doi: 10.1056/NEJMoa1400376.
    1. Farooqui MZ, Valdez J, Martyr S, Aue G, Saba N, Niemann CU, Herman SE, Tian X, Marti G, Soto S, Hughes TE, Jones J, Lipsky A, Pittaluga S, Stetler-Stevenson M, Yuan C, Lee YS, Pedersen LB, Geisler CH, Calvo KR, Arthur DC, Maric I, Childs R, Young NS, Wiestner A. Ibrutinib for previously untreated and relapsed or refractory chronic lymphocytic leukaemia with TP53 aberrations: a phase 2, single-arm trial. Lancet Oncol. 2015;16(2):169–176. doi: 10.1016/S1470-2045(14)71182-9.
    1. Maddocks K, Jones JA. Bruton tyrosine kinase inhibition in chronic lymphocytic leukemia. Semin Oncol. 2016;43(2):251–259. doi: 10.1053/j.seminoncol.2016.02.008.
    1. Wang ML, Blum KA, Martin P, Goy A, Auer R, Kahl BS, Jurczak W, Advani RH, Romaguera JE, Williams ME, Barrientos JC, Chmielowska E, Radford J, Stilgenbauer S, Dreyling M, Jedrzejczak WW, Johnson P, Spurgeon SE, Zhang L, Baher L, Cheng M, Lee D, Beaupre DM, Rule S. Long-term follow-up of MCL patients treated with single-agent ibrutinib: updated safety and efficacy results. Blood. 2015;126(6):739–745. doi: 10.1182/blood-2015-03-635326.
    1. Wang ML, Rule S, Martin P, Goy A, Auer R, Kahl BS, Jurczak W, Advani RH, Romaguera JE, Williams ME, Barrientos JC, Chmielowska E, Radford J, Stilgenbauer S, Dreyling M, Jedrzejczak WW, Johnson P, Spurgeon SE, Li L, Zhang L, Newberry K, Ou Z, Cheng N, Fang B, McGreivy J, Clow F, Buggy JJ, Chang BY, Beaupre DM, Kunkel LA, Blum KA. Targeting BTK with ibrutinib in relapsed or refractory mantle-cell lymphoma. N Engl J Med. 2013;369(6):507–516. doi: 10.1056/NEJMoa1306220.
    1. Treon SP, Tripsas CK, Meid K, Warren D, Varma G, Green R, Argyropoulos KV, Yang G, Cao Y, Xu L, Patterson CJ, Rodig S, Zehnder JL, Aster JC, Harris NL, Kanan S, Ghobrial I, Castillo JJ, Laubach JP, Hunter ZR, Salman Z, Li J, Cheng M, Clow F, Graef T, Palomba ML, Advani RH. Ibrutinib in previously treated Waldenstrom's macroglobulinemia. N Engl J Med. 2015;372(15):1430–1440. doi: 10.1056/NEJMoa1501548.
    1. Dimopoulos MA, Trotman J, Tedeschi A, Matous JV, Macdonald D, Tam C, Tournilhac O, Ma S, Oriol A, Heffner LT, Shustik C, Garcia-Sanz R, Cornell RF, de Larrea CF, Castillo JJ, Granell M, Kyrtsonis MC, Leblond V, Symeonidis A, Kastritis E, Singh P, Li J, Graef T, Bilotti E, Treon S, Buske C, i NSG, the European Consortium for Waldenstrom's M Ibrutinib for patients with rituximab-refractory Waldenstrom's macroglobulinaemia (iNNOVATE): an open-label substudy of an international, multicentre, phase 3 trial. Lancet Oncol. 2017;18(2):241–250. doi: 10.1016/S1470-2045(16)30632-5.
    1. Li N, Sun Z, Liu Y, Guo M, Zhang Y, Zhou D, Zhang B, Su D, Zhang S, Han J, Gao Y, Guo Y, Wang Z, Wei M, Luo L, Wang L. Abstract 2597: BGB-3111 is a novel and highly selective Bruton's tyrosine kinase (BTK) inhibitor. Can Res. 2015;75(15 Supplement):2597–2597. doi: 10.1158/1538-7445.Am2015-2597.
    1. Song Y, Zhou K, Zou D, Zhou J, Hu J, Yang H, Zhang H, Ji J, Xu W, Jin J, Lv F, Feng R, Gao S, Zhou D, Guo H, Wang A, Hilger J, Huang J, Novotny W, Osman M, Zhu J. Safety and activity of the investigational bruton tyrosine kinase inhibitor zanubrutinib (BGB-3111) in patients with mantle cell lymphoma from a phase 2 trial. Blood. 2018;132(Suppl 1):148–148. doi: 10.1182/blood-2018-99-117956.
    1. Tam C, Grigg AP, Opat S, Ku M, Gilbertson M, Anderson MA, Seymour JF, Ritchie DS, Dicorleto C, Dimovski B, Hedrick E, Yang J, Wang L, Luo L, Xue L, Roberts AW. The BTK inhibitor, BGB-3111, is safe, tolerable, and highly active in patients with relapsed/ refractory B-cell malignancies: initial report of a phase 1 first-in-human trial. Blood. 2015;126(23):832–832. doi: 10.1182/blood.V126.23.832.832.
    1. Tam CS, Wang M, Simpson D, Opat S, Cull G, Munoz J, Phillips TJ, Kim W-S, Hilger J, Huang J, Novotny W, Trotman J. Updated safety and activity of the investigational bruton tyrosine kinase inhibitor zanubrutinib (BGB-3111) in patients with mantle cell lymphoma. Blood. 2018;132(Suppl 1):1592–1592. doi: 10.1182/blood-2018-99-117224.
    1. Tam CS, LeBlond V, Novotny W, Owen RG, Tedeschi A, Atwal S, Cohen A, Huang J, Buske C. A head-to-head Phase III study comparing zanubrutinib versus ibrutinib in patients with Waldenstrom macroglobulinemia. Future Oncol. 2018;14(22):2229–2237. doi: 10.2217/fon-2018-0163.
    1. A Study of Zanubrutinib (BGB-3111) Versus ibrutinib in patients with relapsed/refractory chronic lymphocytic leukemia. Available from: . NLM identifier: NCT03734016. Accessed 8 May 2019
    1. Hillmen Peter, Brown Jennifer R., Kahl Brad S., Ghia Paolo, Robak Tadeusz, Marimpietri Carol, Cohen Aileen, Huang Jane, Tam Constantine Si Lun. Phase 3 zanubrutinib (BGB-3111) vs bendamustine + rituximab (BR) in patients (pts) with treatment-naïve (TN) chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL) Journal of Clinical Oncology. 2018;36(15_suppl):TPS7581–TPS7581. doi: 10.1200/JCO.2018.36.15_suppl.TPS7581.
    1. Tam CS, Trotman J, Opat S, Burger JA, Cull G, Gottlieb D, Harrup R, Johnston PB, Marlton P, Munoz J, Seymour JF, Simpson D, Tedeschi A, Elstrom R, Yu Y, Tang Z, Han L, Huang J, Novotny W, Wang L, Roberts AW. Phase 1 study of the selective BTK inhibitor zanubrutinib in B-cell malignancies and safety and efficacy evaluation in CLL. Blood. 2019;134(11):851–859. doi: 10.1182/blood.2019001160.
    1. Nosari A. Infectious complications in chronic lymphocytic leukemia. Mediterr J Hematol Infect Dis. 2012;4(1):e2012070. doi: 10.4084/MJHID.2012.070.
    1. Nucci M. Use of antifungal drugs in hematology. Rev Bras Hematol Hemoter. 2012;34(5):383–391. doi: 10.5581/1516-8484.20120095.
    1. Bruggemann RJ, Alffenaar JW, Blijlevens NM, Billaud EM, Kosterink JG, Verweij PE, Burger DM. Clinical relevance of the pharmacokinetic interactions of azole antifungal drugs with other coadministered agents. Clin Infect Dis. 2009;48(10):1441–1458. doi: 10.1086/598327.
    1. de Jong J, Skee D, Murphy J, Sukbuntherng J, Hellemans P, Smit J, de Vries R, Jiao JJ, Snoeys J, Mannaert E. Effect of CYP3A perpetrators on ibrutinib exposure in healthy participants. Pharmacol Res Perspect. 2015;3(4):e00156. doi: 10.1002/prp2.156.
    1. Srinivas NR. Pharmacokinetic interaction of rifampicin with oral versus intravenous anticancer drugs: challenges, dilemmas and paradoxical effects due to multiple mechanisms. Drugs R D. 2016;16(2):141–148. doi: 10.1007/s40268-016-0133-0.
    1. Liu L, Bello A, Dresser MJ, Heald D, Komjathy SF, O'Mara E, Rogge M, Stoch SA, Robertson SM. Best practices for the use of itraconazole as a replacement for ketoconazole in drug-drug interaction studies. J Clin Pharmacol. 2016;56(2):143–151. doi: 10.1002/jcph.562.
    1. Calquence(r) (acalabrutinib) [package insert] (2017) Acerta Pharma, South San Francisco
    1. Barr PM, Brown JR, Hillmen P, O'Brien S, Barrientos JC, Reddy NM, Coutre S, Mulligan SP, Jaeger U, Furman RR, Cymbalista F, Montillo M, Dearden C, Robak T, Moreno C, Pagel JM, Burger JA, Suzuki S, Sukbuntherng J, Cole G, James DF, Byrd JC. Impact of ibrutinib dose adherence on therapeutic efficacy in patients with previously treated CLL/SLL. Blood. 2017;129(19):2612–2615. doi: 10.1182/blood-2016-12-737346.

Source: PubMed

3
Abonnere