Efficacy and safety results of depatuxizumab mafodotin (ABT-414) in patients with advanced solid tumors likely to overexpress epidermal growth factor receptor

Glenwood D Goss, Everett E Vokes, Michael S Gordon, Leena Gandhi, Kyriakos P Papadopoulos, Drew W Rasco, JuDee S Fischer, Katharine L Chu, William W Ames, Rajendar K Mittapalli, Ho-Jin Lee, Jiewei Zeng, Lisa A Roberts-Rapp, Lise I Loberg, Peter J Ansell, Edward B Reilly, Christopher J Ocampo, Kyle D Holen, Anthony W Tolcher, Glenwood D Goss, Everett E Vokes, Michael S Gordon, Leena Gandhi, Kyriakos P Papadopoulos, Drew W Rasco, JuDee S Fischer, Katharine L Chu, William W Ames, Rajendar K Mittapalli, Ho-Jin Lee, Jiewei Zeng, Lisa A Roberts-Rapp, Lise I Loberg, Peter J Ansell, Edward B Reilly, Christopher J Ocampo, Kyle D Holen, Anthony W Tolcher

Abstract

Background: Epidermal growth factor receptor (EGFR) alterations are associated with multiple cancers. Current EGFR-directed therapies have led to increased efficacy but are associated with specific side effects. The antibody-drug conjugate depatuxizumab mafodotin (depatux-m) targets EGFR with a monoclonal antibody linked to a cytotoxin, and is highly tumor-specific.

Methods: This phase 1/2 study evaluated the safety, pharmacokinetics, and efficacy of depatux-m in patients who had advanced solid tumors with known wild-type EGFR overexpression, amplification, or mutated EGFR variant III. A 3 + 3 dose escalation was used, and 2 dosing schedules were evaluated. Depatux-m also was manufactured under an alternate process to reduce the drug load and improve the safety profile, and it was tested at the maximum tolerated dose (MTD). In another cohort, prolonged infusion time of depatux-m was evaluated; and a cohort with confirmed EGFR amplification also was evaluated at the MTD.

Results: Fifty-six patients were treated. The MTD and the recommended phase 2 dose for depatux-m was 3.0 mg/kg. Common adverse events (AEs) were blurred vision (48%) and fatigue (41%). A majority of patients (66%) experienced 1 or more ocular AEs. Grade 3 or 4 AEs were observed in 43% of patients. One patient with EGFR-amplified, triple-negative breast cancer had a partial response. Stable disease was observed in 23% of patients. Pharmacokinetics revealed that depatux-m exposures were approximately dose-proportional.

Conclusions: Depatux-m resulted in infrequent nonocular AEs but increased ocular AEs. Patient follow-up confirmed that ocular AEs were reversible. Lowering the drug-antibody ratio did not decrease the number of ocular AEs. A partial response in 1 patient with EGFR-amplified disease provides the opportunity to study depatux-m in diseases with a high incidence of EGFR amplification. Cancer 2018;124:2174-83. © 2018 The Authors. Cancer published by Wiley Periodicals, Inc. on behalf of American Cancer Society. This is an open access article under the terms of the Creative Commons Attribution NonCommercial License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited and is not used for commercial purposes.

Trial registration: ClinicalTrials.gov NCT01800695.

Keywords: ABT-414; antibody-drug conjugate; depatuxizumab mafodotin (depatux-m); epidermal growth factor receptor (EGFR).

© 2018 The Authors. Cancer published by Wiley Periodicals, Inc. on behalf of American Cancer Society.

Figures

Figure 1
Figure 1
The study schema is illustrated. EGFR indicates epidermal growth factor receptor; MTD, maximum tolerated dose; Q3W, once every 3 weeks; RPTD, recommended phase 2 dose.
Figure 2
Figure 2
The pharmacokinetics of depatuxizumab mafodotin (depatux‐m), total depatux, and Cys‐mafodotin are illustrated. (A) Concentration‐time profiles are illustrated for each analyte after the first intravenous infusion of depatux‐m at 2 mg/kg. Mean ± standard deviation values are shown for the depatux‐m (B) maximum serum concentration (Cmax) and (C) area under the curve on day 21 (AUCday 21) versus the depatux‐m dose.
Figure 3
Figure 3
Best response and time on therapy are illustrated. The best responses, as determined by the investigator, and the time on depatuxizumab mafodotin (depatux‐m) therapy are shown for 52 of 56 patients who had data available. EGFR indicates epidermal growth factor receptor.

References

    1. Normanno N, De Luca A, Bianco C, et al. Epidermal growth factor receptor (EGFR) signaling in cancer. Gene. 2006;366:2‐16.
    1. Jonker DJ, O'Callaghan CJ, Karapetis CS, et al. Cetuximab for the treatment of colorectal cancer. N Engl J Med. 2007;357:2040‐2048.
    1. Bonner JA, Harari PM, Giralt J, et al. Radiotherapy plus cetuximab for locoregionally advanced head and neck cancer: 5‐year survival data from a phase 3 randomised trial, and relation between cetuximab‐induced rash and survival. Lancet Oncol. 2010;11:21‐28.
    1. Liang W, Wu X, Fang W, et al. Network meta‐analysis of erlotinib, gefitinib, afatinib and icotinib in patients with advanced non‐small‐cell lung cancer harboring EGFR mutations [serial online]. PLoS One. 2014;9:e85245.
    1. Reilly EB, Phillips AC, Buchanan FG, et al. Characterization of ABT‐806, a humanized tumor‐specific anti‐EGFR monoclonal antibody. Mol Cancer Ther. 2015;14:1141‐1151.
    1. Cleary JM, Reardon DA, Azad N, et al. A phase 1 study of ABT‐806 in subjects with advanced solid tumors. Invest New Drugs. 2015;33:671‐678.
    1. Agero AL, Dusza SW, Benvenuto‐Andrade C, Busam KJ, Myskowski P, Halpern AC. Dermatologic side effects associated with the epidermal growth factor receptor inhibitors. J Am Acad Dermatol. 2006;55:657‐670.
    1. Klute K, Nackos E, Tasaki S, Nguyen DP, Bander NH, Tagawa ST. Microtubule inhibitor‐based antibody‐drug conjugates for cancer therapy. Onco Targets Ther. 2014;7:2227‐2236.
    1. Phillips AC, Boghaert ER, Vaidya KS, et al. ABT‐414, an antibody‐drug conjugate targeting a tumor‐selective EGFR epitope. Mol Cancer Ther. 2016;15:661‐669.
    1. Hamblett KJ, Senter PD, Chace DF, et al. Effects of drug loading on the antitumor activity of a monoclonal antibody drug conjugate. Clin Cancer Res. 2004;10:7063‐7070.
    1. Donaghy H. Effects of antibody, drug and linker on the preclinical and clinical toxicities of antibody‐drug conjugates. MAbs. 2016;8:659‐671.
    1. Eaton JS, Miller PE, Mannis MJ, Murphy CJ. Ocular adverse events associated with antibody‐drug conjugates in human clinical trials. J Ocul Pharmacol Ther. 2015;31:589‐604.
    1. Brennan CW, Verhaak RG, McKenna A, et al. The somatic genomic landscape of glioblastoma. Cell. 2013;155:462‐477.
    1. Gan HK, Fichtel L, Lassman AB, et al. ET‐19 A phase 1 study evaluating ABT‐414 with temozolomide (TMZ) or concurrent radiotherapy (RT) and TMZ in glioblastoma (GBM) [abstract]. Neuro Oncol. 2014;16(suppl 5):v83.
    1. Fakih M, Vincent M. Adverse events associated with anti‐EGFR therapies for the treatment of metastatic colorectal cancer. Curr Oncol. 2010;17(suppl 1):S18‐S30.
    1. Moskowitz CH, Forero‐Torres A, Shah BD, et al. Interim analysis of a phase 1 study of the antibody‐drug conjugate sGN‐CD19A in relapsed or refractory B‐lineage non‐Hodgkin lymphoma [abstract]. Blood. 2014;124:1741.
    1. Fathi AT, Chen R, Trippett TM, et al. Interim analysis of a phase 1 study of the antibody‐drug conjugate SGN‐CD19A in relapsed or refractory B‐lineage acute leukemia and highly aggressive lymphoma [abstract]. Blood. 2014;124:963.
    1. Thompson JA, Forero‐Torres A, Heath EI, et al. The effect of SGN‐75, a novel antibody—drug conjugate (ADC), in treatment of patients with renal cell carcinoma (RCC) or non‐Hodgkin lymphoma (NHL): a phase I study [abstract]. J Clin Oncol. 2011;29(15 suppl):3071.
    1. Tannir NM, Forero‐Torres A, Ramchandren R, et al. Phase I dose‐escalation study of SGN‐75 in patients with CD70‐positive relapsed/refractory non‐Hodgkin lymphoma or metastatic renal cell carcinoma. Invest New Drugs. 2014;32:1246‐1257.
    1. Thompson JA, Motzer R, Molina AM, et al. Phase I studies of anti‐ENPP3 antibody drug conjugates (ADCs) in advanced refractory renal cell carcinomas (RRCC) [abstract]. J Clin Oncol. 2015;33(15 suppl):2503.
    1. Reardon DA, Lassman AB, van den Bent M, et al. Efficacy and safety results of ABT‐414 in combination with radiation and temozolomide in newly diagnosed glioblastoma. Neuro Oncol. 2017;19:965‐975.
    1. Lassman AB, van den Bent MJ, Gan HK, Reardon DA, Kumthekar Priya, Butowski NA. Efficacy analysis of ABT‐414 with or without temozolomide (TMZ) in patients (pts) with EGFR‐amplified, recurrent glioblastoma (rGBM) from a multicenter, international phase I clinical trial [abstract]. J Clin Oncol. 2017;35(15 suppl):2003.
    1. Gan HK, Reardon DA, Lassman AB, et al. Safety, pharmacokinetics, and antitumor response of depatuxizumab mafodotin as monotherapy or in combination with temozolomide in patients with glioblastoma [published online ahead of print October 25, 2017]. Neuro Oncol. doi: .
    1. van den Bent M, Gan HK, Lassman AB, et al. Efficacy of depatuxizumab mafodotin (ABT‐414) monotherapy in patients with EGFR‐amplified, recurrent glioblastoma; results from a multi‐center, international study. Cancer Chemother Pharmacol. 2017;80:1209‐1217.
    1. van den Bent M, Eoli M, Sepulveda JM, et al. LTBK‐04 First results of the randomized phase II study on depatux‐m alone, depatux‐m in combination with temozolomide and either temozolomide or lomustine in recurrent EGFR amplified glioblastoma: first report from INTELLANCE 2/EORTC trial 1410 [abstract]. Neuro Oncol. 2017;19(suppl 6):vi316.

Source: PubMed

3
Abonnere