A Phase Ib Dose-Escalation Study of the Safety, Tolerability, and Pharmacokinetics of Cobimetinib and Duligotuzumab in Patients with Previously Treated Locally Advanced or Metastatic Cancers with Mutant KRAS

Christopher H Lieu, Manuel Hidalgo, Jordan D Berlin, Andrew H Ko, Andres Cervantes, Patricia LoRusso, David E Gerber, J Paul Eder, S Gail Eckhardt, Amy V Kapp, Amy Tsuhako, Bruce McCall, Andrea Pirzkall, Anne Uyei, Josep Tabernero, Christopher H Lieu, Manuel Hidalgo, Jordan D Berlin, Andrew H Ko, Andres Cervantes, Patricia LoRusso, David E Gerber, J Paul Eder, S Gail Eckhardt, Amy V Kapp, Amy Tsuhako, Bruce McCall, Andrea Pirzkall, Anne Uyei, Josep Tabernero

Abstract

Lessons learned: Cobimetinib and duligotuzumab were well tolerated as single agents and in combination with other agents.The cobimetinib and duligotuzumab combination was associated with increased toxicity, most notably gastrointestinal, and limited efficacy in the patient population tested.

Background: KRAS-mutant tumors possess abnormal mitogen-activated protein kinases (MAPK) pathway signaling, leading to dysregulated cell proliferation. Cobimetinib blocks MAPK signaling. The dual-action antibody duligotuzumab (MEHD7945A) inhibits ligand binding to both epidermal growth factor receptor (EGFR) and human epidermal growth factor receptor 3 (HER3). Blockade of EGFR/HER3 and inhibition of mitogen-activated protein kinase (MEK) in KRAS-mutant tumors may provide additive benefit.

Methods: Patients with KRAS-mutant solid tumors were eligible for this phase Ib dose-escalation study with a planned expansion phase. Duligotuzumab was given intravenously (IV) at 1,100 mg every 2 weeks (q2w), while cobimetinib was given orally in a standard 3 + 3 design to identify the recommended phase II dose (RP2D). The primary objective was to evaluate the safety and tolerability of this combination.

Results: Twenty-three patients were enrolled. Dose-limiting toxicities (DLTs) included grade 4 hypokalemia and grade 3 mucosal inflammation, asthenia, and dermatitis acneiform. Seventy percent of patients experienced grade 3 or worse adverse events (AEs). Five (22%) and 12 (52%) patients missed at least 1 dose of duligotuzumab and cobimetinib, respectively, and 9 (39%) patients required a cobimetinib dose reduction. Three (13%) patients discontinued due to an AE. Best response was limited to 9 patients with stable disease and 13 patients with progressive disease.

Conclusion: Given the limited tolerability and efficacy of this combination, the study did not proceed to expansion stage and closed for enrollment.

Trial registration: ClinicalTrials.gov NCT01986166.

© AlphaMedPress; the data published online to support this summary is the property of the authors.

References

    1. Sergina NV, Moasser MM. The HER family and cancer: Emerging molecular mechanisms and therapeutic targets. Trends Mol Med 2007;13:527–534.
    1. Carracedo A, Ma L, Teruya‐Feldstein J et al. Inhibition of mTORC1 leads to MAPK pathway activation through a PI3K‐dependent feedback loop in human cancer. J Clin Invest 2008;118:3065–3074.
    1. Turke AB, Song Y, Costa C et al. MEK inhibition leads to PI3K/AKT activation by relieving a negative feedback on ERBB receptors. Cancer Res 2012;72:3228–3237.
    1. Mirzoeva OK, Das D, Heiser LM et al. Basal subtype and MAPK/ERK kinase (MEK)‐phosphoinositide 3‐kinase feedback signaling determine susceptibility of breast cancer cells to MEK inhibition. Cancer Res 2009;69:565–572.
    1. Diep CH, Munoz RM, Choudhary A et al. Synergistic effect between erlotinib and MEK inhibitors in KRAS wild‐type human pancreatic cancer cells. Clin Cancer Res 2011;17:2744–2756.
    1. Young A, Lou D, McCormick F. Oncogenic and wild‐type Ras play divergent roles in the regulation of mitogen‐activated protein kinase signaling. Cancer Discov 2013;3:112–123.
    1. Sunaga N, Kaira K, Imai H et al. Oncogenic KRAS‐induced epiregulin overexpression contributes to aggressive phenotype and is a promising therapeutic target in non‐small‐cell lung cancer. Oncogene 2013;32:4034–4042.
    1. Sun C, Hobor S, Bertotti A et al. Intrinsic resistance to MEK inhibition in KRAS mutant lung and colon cancer through transciptional induction of ERBB3. Cell Rep 2014;7:86–93.
    1. Fayette J, Wirth L, Oprean C et al. Randomized phase II study of duligotuzumab (MEHD7945A) vs. cetuximab in squamous cell carcinoma of the head and neck (MEHGAN Study). Front Oncol 2016;6:232.
    1. Johnson L, Mercer K, Greenbaum D et al. Somatic activation of the K‐ras oncogene causes early onset lung cancer in mice. Nature 2001;410:1111–1116.
    1. Hynes NE, MacDonald G. ErbB receptors and signaling pathways in cancer. Curr Opin Cell Biol 2009;21:177–184.
    1. Holbro T, Civenni, G, Hynes NE. The ErbB receptors and their role in cancer progression. Exp Cell Res 2003;284:99–110.
    1. Moasser MM. Targeting the function of the HER2 oncogene in human cancer therapeutics. Oncogene 2007;26:6577–6592.
    1. Ciardiello F, Tortora G. EGFR antagonists in cancer treatment. N Engl J Med 2008;358:1160–1174.
    1. Riese DJ 2nd, van Raaij TM, Plowman GD et al. The cellular response to neuregulins is governed by complex interactions of the erbB receptor family. Mol Cell Biol 1995;15:5770–5776.
    1. Pinkas‐Kramarski R, Soussan L, Waterman H et al. Diversification of Neu differentiation factor and epidermal growth factor signaling by combinatorial receptor interactions. EMBO J 1996;15:2452–2467.
    1. Jura N, Shan Y, Cao X et al. Structural analysis of the catalytically inactive kinase domain of the human EGF receptor 3. Proc Natl Acad Sci USA 2009;106:21608–21613.
    1. Shi F, Telesco SE, Liu Y et al. ErbB3/HER3 intracellular domain is competent to bind ATP and catalyze autophosphorylation. Proc Natl Acad Sci USA 2010;107:7692–7697.
    1. Sergina NV, Rausch M, Wang D et al. Escape from HER‐family tyrosine kinase inhibitor therapy by the kinase‐inactive HER3. Nature 2007;445:437–441.

Source: PubMed

3
Se inscrever