Phase I, Open-Label, Dose-Escalation/Dose-Expansion Study of Lifirafenib (BGB-283), an RAF Family Kinase Inhibitor, in Patients With Solid Tumors

Jayesh Desai, Hui Gan, Catherine Barrow, Michael Jameson, Victoria Atkinson, Andrew Haydon, Michael Millward, Stephen Begbie, Michael Brown, Ben Markman, William Patterson, Andrew Hill, Lisa Horvath, Adnan Nagrial, Gary Richardson, Christopher Jackson, Michael Friedlander, Phillip Parente, Ben Tran, Lai Wang, Yunxin Chen, Zhiyu Tang, Wendy Huang, John Wu, Dewan Zeng, Lusong Luo, Benjamin Solomon, Jayesh Desai, Hui Gan, Catherine Barrow, Michael Jameson, Victoria Atkinson, Andrew Haydon, Michael Millward, Stephen Begbie, Michael Brown, Ben Markman, William Patterson, Andrew Hill, Lisa Horvath, Adnan Nagrial, Gary Richardson, Christopher Jackson, Michael Friedlander, Phillip Parente, Ben Tran, Lai Wang, Yunxin Chen, Zhiyu Tang, Wendy Huang, John Wu, Dewan Zeng, Lusong Luo, Benjamin Solomon

Abstract

Purpose: Lifirafenib is an investigational, reversible inhibitor of B-RAFV600E, wild-type A-RAF, B-RAF, C-RAF, and EGFR. This first-in-human, phase I, dose-escalation/dose-expansion study evaluated the safety, tolerability, and efficacy of lifirafenib in patients with B-RAF- or K-RAS/N-RAS-mutated solid tumors.

Methods: During dose escalation, adult patients with histologically/cytologically confirmed advanced solid tumors received escalating doses of lifirafenib. Primary end points were safety/tolerability during dose escalation and objective response rate in preselected patients with B-RAF and K-RAS/N-RAS mutations during dose expansion.

Results: The maximum tolerated dose was established as 40 mg/d; dose-limiting toxicities included reversible thrombocytopenia and nonhematologic toxicity. Across the entire study, the most common grade ≥ 3 treatment-emergent adverse events were hypertension (n = 23; 17.6%) and fatigue (n = 13; 9.9%). One patient with B-RAF-mutated melanoma achieved complete response, and 8 patients with B-RAF mutations had confirmed objective responses: B-RAFV600E/K melanoma (n = 5, including 1 patient treated with prior B-RAF/MEK inhibitor therapy), B-RAFV600E thyroid cancer/papillary thyroid cancer (PTC; n = 2), and B-RAFV600E low-grade serous ovarian cancer (LGSOC; n = 1). One patient with B-RAF-mutated non-small-cell lung cancer (NSCLC) had unconfirmed partial response (PR). Patients with K-RAS-mutated endometrial cancer and K-RAS codon 12-mutated NSCLC had confirmed PR (n = 1 each). No responses were seen in patients with K-RAS/N-RAS-mutated colorectal cancer (n = 20).

Conclusion: Lifirafenib is a novel inhibitor of key RAF family kinases and EGFR, with an acceptable risk-benefit profile and antitumor activity in patients with B-RAFV600-mutated solid tumors, including melanoma, PTC, and LGSOC, as well as K-RAS-mutated NSCLC and endometrial carcinoma. Future comparisons with first-generation B-RAF inhibitors and exploration of lifirafenib alone or as combination therapy in patients with selected RAS mutations who are resistant/refractory to first-generation B-RAF inhibitors are warranted.

Trial registration: ClinicalTrials.gov NCT02610361.

Figures

FIG 1.
FIG 1.
(A and B) Responses in patients with B-RAF–mutated cancers (both phases). (>) Ongoing treatment indicates ongoing response at time of data cutoff. An additional 6 patients not included in this figure had prior B-RAF inhibitor treatment (4 had melanoma, 1 had colorectal cancer, and 1 had adenocarcinoma with unknown primary origin). (*) Patients who received prior B-RAF/MEK inhibitor treatment. CR, complete response; NSCLC, non–small-cell lung cancer; PD, progressive disease; PR, partial response; SD, stable disease.
FIG 2.
FIG 2.
(A and B) Responses in patients with K-RAS–mutated cancers (excluding colorectal cancer; both phases). NE, not evaluable; NSCLC, non–small-cell lung cancer; PD, progressive disease; PR, partial response; SD, stable disease.
FIG 3.
FIG 3.
Responses across lifirafenib dose cohorts by 18F-fluorodeoxyglucose positron emission tomography-computed tomography (FDG-PET-CT) analysis. Two patients were missing a postbaseline PET scan. (*) Patients who were treated beyond progression. CR, complete response; NE, not evaluable; PD, progressive disease; PR, partial response; SD, stable disease; SUVave, average standardized uptake volume.

References

    1. Santarpia L, Lippman SM, El-Naggar AK. Targeting the MAPK-RAS-RAF signaling pathway in cancer therapy. Expert Opin Ther Targets. 2012;16:103–119.
    1. Okuda T, Sekizawa A, Purwosunu Y, et al. Genetics of endometrial cancers. Obstet Gynecol Int. 2010;2010:984013.
    1. Durrant DE, Morrison DK. Targeting the Raf kinases in human cancer: The Raf dimer dilemma. Br J Cancer. 2018;118:3–8.
    1. Long GV, Menzies AM, Nagrial AM, et al. Prognostic and clinicopathologic associations of oncogenic BRAF in metastatic melanoma. J Clin Oncol. 2011;29:1239–1246.
    1. Bösmüller H, Fischer A, Pham DL, et al. Detection of the BRAF V600E mutation in serous ovarian tumors: A comparative analysis of immunohistochemistry with a mutation-specific monoclonal antibody and allele-specific PCR. Hum Pathol. 2013;44:329–335.
    1. Singer G, Oldt R, III, Cohen Y, et al. Mutations in BRAF and KRAS characterize the development of low-grade ovarian serous carcinoma. J Natl Cancer Inst. 2003;95:484–486.
    1. Jones S, Wang TL, Kurman RJ, et al. Low-grade serous carcinomas of the ovary contain very few point mutations. J Pathol. 2012;226:413–420.
    1. Wong KK, Tsang YT, Deavers MT, et al. BRAF mutation is rare in advanced-stage low-grade ovarian serous carcinomas. Am J Pathol. 2010;177:1611–1617.
    1. Strickler JH, Wu C, Bekaii-Saab T. Targeting BRAF in metastatic colorectal cancer: Maximizing molecular approaches. Cancer Treat Rev. 2017;60:109–119.
    1. Stephen AG, Esposito D, Bagni RK, et al. Dragging ras back in the ring. Cancer Cell. 2014;25:272–281.
    1. Chapman PB, Robert C, Larkin J, et al. Vemurafenib in patients with BRAFV600 mutation-positive metastatic melanoma: Final overall survival results of the randomized BRIM-3 study. Ann Oncol. 2017;28:2581–2587.
    1. Hauschild A, Grob JJ, Demidov LV, et al. Dabrafenib in BRAF-mutated metastatic melanoma: A multicentre, open-label, phase 3 randomised controlled trial. Lancet. 2012;380:358–365.
    1. Kopetz S, Desai J, Chan E, et al: PLX4032 in metastatic colorectal cancer patients with mutant BRAF tumors. J Clin Oncol 28, 2010 (suppl; abstr 3534)
    1. Corcoran RB, Ebi H, Turke AB, et al. EGFR-mediated re-activation of MAPK signaling contributes to insensitivity of BRAF mutant colorectal cancers to RAF inhibition with vemurafenib. Cancer Discov. 2012;2:227–235.
    1. Tang Z, Yuan X, Du R, et al. BGB-283, a novel RAF kinase and EGFR inhibitor, displays potent antitumor activity in BRAF-mutated colorectal cancers. Mol Cancer Ther. 2015;14:2187–2197.
    1. Anforth R, Menzies A, Byth K, et al: Factors influencing the development of cutaneous squamous cell carcinoma in patients on BRAF inhibitor therapy. J Am Acad Dermatol 72:809-815.e1, 2015.
    1. Shi H, Moriceau G, Kong X, et al. Melanoma whole-exome sequencing identifies (V600E)B-RAF amplification-mediated acquired B-RAF inhibitor resistance. Nat Commun. 2012;3:724.
    1. Girotti MR, Pedersen M, Sanchez-Laorden B, et al. Inhibiting EGF receptor or SRC family kinase signaling overcomes BRAF inhibitor resistance in melanoma. Cancer Discov. 2013;3:158–167.
    1. Poulikakos PI, Persaud Y, Janakiraman M, et al. RAF inhibitor resistance is mediated by dimerization of aberrantly spliced BRAF(V600E) Nature. 2011;480:387–390.
    1. Rizos H, Menzies AM, Pupo GM, et al. BRAF inhibitor resistance mechanisms in metastatic melanoma: Spectrum and clinical impact. Clin Cancer Res. 2014;20:1965–1977.
    1. Mao M, Tian F, Mariadason JM, et al. Resistance to BRAF inhibition in BRAF-mutant colon cancer can be overcome with PI3K inhibition or demethylating agents. Clin Cancer Res. 2013;19:657–667.
    1. Temraz S, Mukherji D, Shamseddine A. Dual inhibition of MEK and PI3K pathway in KRAS and BRAF mutated colorectal cancers. Int J Mol Sci. 2015;16:22976–22988.
    1. Tang Z, Liu Y, Jiang B, et al: BGB-283: A novel RAF dimer inhibitor, displays potent antitumor activity in HCC patient derived xenograft models. Cancer Res 76, 2016 (suppl; abstr 1230)
    1. Desai J, Markman B, Sandhu SK, et al: Updated safety, efficacy, and pharmacokinetics (PK) results from the phase I study of BGB-A317, an antiprogrammed death-1 (PD-1) mAb in patients (pts) with advanced solid tumors. Presented at 31st Ann Meet Soc Immunother Cancer, National Harbor, MD, November 9−13, 2016.
    1. Hao H, Muniz-Medina VM, Mehta H, et al. Context-dependent roles of mutant B-Raf signaling in melanoma and colorectal carcinoma cell growth. Mol Cancer Ther. 2007;6:2220–2229.
    1. Young H, Baum R, Cremerius U, et al. Measurement of clinical and subclinical tumour response using [18F]-fluorodeoxyglucose and positron emission tomography: Review and 1999 EORTC recommendations. Eur J Cancer. 1999;35:1773–1782.
    1. Wu JH, Cohen DN, Rady PL, et al. BRAF inhibitor-associated cutaneous squamous cell carcinoma: New mechanistic insight, emerging evidence for viral involvement and perspectives on clinical management. Br J Dermatol. 2017;177:914–923.
    1. Robinson ES, Khankin EV, Karumanchi SA, et al. Hypertension induced by vascular endothelial growth factor signaling pathway inhibition: Mechanisms and potential use as a biomarker. Semin Nephrol. 2010;30:591–601.
    1. Cao H, Bhuyan AAM, Umbach AT, et al. Inhibitory effect of afatinib on platelet activation and apoptosis. Cell Physiol Biochem. 2017;43:2264–2276.
    1. Ohnishi H, Yamaguchi K, Shimada S, et al. A new approach to the treatment of atherosclerosis and trapidil as an antagonist to platelet-derived growth factor. Life Sci. 1981;28:1641–1646.
    1. Ohnishi H, Kosuzume H, Hayashi Y, et al. Effects of trapidil on thromboxane A2-induced aggregation of platelets, ischemic changes in heart and biosynthesis of thromboxane A2. Prostaglandins Med. 1981;6:269–281.
    1. McArthur GA, Puzanov I, Amaravadi R, et al. Marked, homogeneous, and early [18F]fluorodeoxyglucose-positron emission tomography responses to vemurafenib in BRAF-mutant advanced melanoma. J Clin Oncol. 2012;30:1628–1634.
    1. Chapman PB, Hauschild A, Robert C, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med. 2011;364:2507–2516.
    1. Harris SJ, Luken MJ, Perez DR, et al: Updated efficacy and safety results from the phase I study of intermittent dosing of the dual MEK/RAF inhibitor, RO5126766 in patients (pts) with RAS/RAF mutated advanced solid tumours. J Clin Oncol 34, 2016 (suppl; abstr 2582)
    1. Chenard-Poirier M, Kaiser M, Boyd K, et al: Results from the biomarker-driven basket trial of RO5126766 (CH5127566), a potent RAF/MEK inhibitor, in RAS- or RAF-mutated malignancies including multiple myeloma. J Clin Oncol 35, 2017 (suppl; abstr 2506)
    1. Canon J, Rex K, Saiki AY, et al. The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunity. Nature. 2019;575:217–223.
    1. Hallin J, Engstrom LD, Hargis L, et al: The KRASG12C inhibitor MRTX849 provides insight toward therapeutic susceptibility of KRAS-mutant cancers in mouse models and patients. Cancer Discov 10:54-71, 2020.

Source: PubMed

3
Abonneren