Safety and efficacy of depatuxizumab mafodotin + temozolomide in patients with EGFR-amplified, recurrent glioblastoma: results from an international phase I multicenter trial

Andrew B Lassman, Martin J van den Bent, Hui K Gan, David A Reardon, Priya Kumthekar, Nicholas Butowski, Zarnie Lwin, Tom Mikkelsen, Louis B Nabors, Kyriakos P Papadopoulos, Marta Penas-Prado, John Simes, Helen Wheeler, Tobias Walbert, Andrew M Scott, Erica Gomez, Ho-Jin Lee, Lisa Roberts-Rapp, Hao Xiong, Peter J Ansell, Earle Bain, Kyle D Holen, David Maag, Ryan Merrell, Andrew B Lassman, Martin J van den Bent, Hui K Gan, David A Reardon, Priya Kumthekar, Nicholas Butowski, Zarnie Lwin, Tom Mikkelsen, Louis B Nabors, Kyriakos P Papadopoulos, Marta Penas-Prado, John Simes, Helen Wheeler, Tobias Walbert, Andrew M Scott, Erica Gomez, Ho-Jin Lee, Lisa Roberts-Rapp, Hao Xiong, Peter J Ansell, Earle Bain, Kyle D Holen, David Maag, Ryan Merrell

Abstract

Background: Patients with glioblastoma (GBM) have a dismal prognosis. Nearly all will relapse with no clear standard of care for recurrent disease (rGBM). Approximately 50% of patients have tumors harboring epidermal growth factor receptor (EGFR) amplification. The antibody-drug conjugate depatuxizumab mafodotin (depatux-m) binds cells with EGFR amplification, is internalized, and releases a microtubule toxin, killing the cell. Here we report efficacy, safety and pharmacokinetics (PK) of depatux-m + temozolomide (TMZ) in patients with EGFR-amplified rGBM.

Methods: M12-356 (NCT01800695) was an open-label study encompassing patients with newly diagnosed or rGBM across 3 treatment arms. Results are reported for adults with EGFR-amplified, measurable rGBM who received depatux-m (0.5-1.5 mg/kg) on days 1 and 15, and TMZ (150-200 mg/m2) on days 1-5 in a 28-day cycle. Patients were bevacizumab and nitrosourea naïve.

Results: There were 60 patients, median age 56 years (range, 20-79). Fifty-nine patients previously received TMZ. Common adverse events (AEs) were blurred vision (63%), fatigue (38%), and photophobia (35%). Grades 3/4 AEs were split between ocular and non-ocular AEs, occurring in 22% of patients each. Systemic PK exposure of depatux-m was dose proportional. The objective response rate was 14.3%, the 6-month progression-free survival rate was 25.2%, and the 6-month overall survival rate was 69.1%.

Conclusions: Depatux-m + TMZ displayed an AE profile similar to what was described previously. Antitumor activity in this TMZ-refractory population was encouraging. Continued study of depatux-m in patients with EGFR-amplified, newly diagnosed, or recurrent GBM is ongoing in 2 global, randomized trials (NCT02573324, NCT02343406).

Figures

Fig. 1
Fig. 1
Best response and time on therapy. The best responses as determined by the investigator using RANO criteria and time on depatux-m therapy are shown for 60/60 patients (analysis included 1 patient each with SD and PD without measurable disease at baseline).
Fig. 2
Fig. 2
Best percentage change in tumor size. The percent change in target lesion from baseline are shown for 56/58 patients who had measurable disease at baseline and at least one post-baseline measurement. Best tumor percent change is defined as the maximum reduction/minimum increase from baseline in tumor size. Values were determined per investigator measurements.

References

    1. Stupp R, Hegi ME, Mason WP, et al. ; European Organisation for Research and Treatment of Cancer Brain Tumour and Radiation Oncology Groups; National Cancer Institute of Canada Clinical Trials Group Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol. 2009;10(5):459–466.
    1. Chinot OL, Wick W, Mason W, et al. . Bevacizumab plus radiotherapy-temozolomide for newly diagnosed glioblastoma. N Engl J Med. 2014;370(8):709–722.
    1. Franceschi E, Minichillo S, Brandes AA. Pharmacotherapy of glioblastoma: established treatments and emerging concepts. CNS Drugs. 2017;31(8):675–684.
    1. Taal W, Oosterkamp HM, Walenkamp AM, et al. . Single-agent bevacizumab or lomustine versus a combination of bevacizumab plus lomustine in patients with recurrent glioblastoma (BELOB trial): a randomised controlled phase 2 trial. Lancet Oncol. 2014;15(9):943–953.
    1. Seystahl K, Wick W, Weller M. Therapeutic options in recurrent glioblastoma—an update. Crit Rev Oncol Hematol. 2016;99:389–408.
    1. Wick W, Gorlia T, Bendszus M, et al. . Lomustine and bevacizumab in progressive glioblastoma. N Engl J Med. 2017;377(20):1954–1963.
    1. Brennan CW, Verhaak RG, McKenna A, et al. ; TCGA Research Network The somatic genomic landscape of glioblastoma. Cell. 2013;155(2):462–477.
    1. Gan HK, Cvrljevic AN, Johns TG. The epidermal growth factor receptor variant III (EGFRvIII): where wild things are altered. FEBS J. 2013;280(21):5350–5370.
    1. Yoshimoto K, Dang J, Zhu S, et al. . Development of a real-time RT-PCR assay for detecting EGFRvIII in glioblastoma samples. Clin Cancer Res. 2008;14(2):488–493.
    1. van den Bent MJ, Roberts-Rapp LA, Ansell P, et al. . Epidermal growth factor receptor (EGFR) amplification rates observed in screening patients for randomized clinical trials in glioblastoma. Ann Oncol. 2017; 28(suppl_5). doi:10.1093/annonc/mdx366.002.
    1. Padfield E, Ellis HP, Kurian KM. Current therapeutic advances targeting EGFR and EGFRvIII in glioblastoma. Front Oncol. 2015;5:5.
    1. Weller M, Hentschel B, Kaulich K, et al. . EGFR gene amplification and variant III (EGFRvIII) mutation in primary and recurrent glioblastoma. J Clin Oncol. 2016; 34:2042.
    1. van den Bent MJ, Gao Y, Kerkhof M, et al. . Changes in the EGFR amplification and EGFRvIII expression between paired primary and recurrent glioblastomas. Neuro Oncol. 2015;17(7):935–941.
    1. Lassman A, Dimino C, Mansukhani M, et al. . PATH-35. Maintenance of EGFR abnormalities over time and comparison of molecular methodologies in glioblastoma (GBM). Neuro Oncol. 2017;19:vi178.
    1. van den Bent MJ, Brandes AA, Rampling R, et al. . Randomized phase II trial of erlotinib versus temozolomide or carmustine in recurrent glioblastoma: EORTC brain tumor group study 26034. J Clin Oncol. 2009;27(8):1268–1274.
    1. Kreisl TN, Lassman AB, Mischel PS, et al. . A pilot study of everolimus and gefitinib in the treatment of recurrent glioblastoma (GBM). J Neurooncol. 2009;92(1):99–105.
    1. Raizer JJ, Abrey LE, Lassman AB, et al. ; North American Brain Tumor Consortium A phase II trial of erlotinib in patients with recurrent malignant gliomas and nonprogressive glioblastoma multiforme postradiation therapy. Neuro Oncol. 2010;12(1):95–103.
    1. Peereboom DM, Shepard DR, Ahluwalia MS, et al. . Phase II trial of erlotinib with temozolomide and radiation in patients with newly diagnosed glioblastoma multiforme. J Neurooncol. 2010;98(1):93–99.
    1. Raizer JJ, Giglio P, Hu J, et al. ; Brain Tumor Trials Collaborative A phase II study of bevacizumab and erlotinib after radiation and temozolomide in MGMT unmethylated GBM patients. J Neurooncol. 2016;126(1):185–192.
    1. Chakravarti A, Wang M, Robins HI, et al. . RTOG 0211: a phase ½ study of radiation therapy with concurrent gefitinib for newly diagnosed glioblastoma patients. Int J Radiat Oncol Biol Phys. 2013;85(5):1206–1211.
    1. Neyns B, Sadones J, Joosens E, et al. . Stratified phase II trial of cetuximab in patients with recurrent high-grade glioma. Ann Oncol. 2009;20(9):1596–1603.
    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(3):671–678.
    1. Krop IE, Kim SB, González-Martín A, et al. ; TH3RESA study collaborators Trastuzumab emtansine versus treatment of physician’s choice for pretreated HER2-positive advanced breast cancer (TH3RESA): a randomised, open-label, phase 3 trial. Lancet Oncol. 2014;15(7):689–699.
    1. Moskowitz CH, Nademanee A, Masszi T, et al. ; AETHERA Study Group Brentuximab vedotin as consolidation therapy after autologous stem-cell transplantation in patients with Hodgkin’s lymphoma at risk of relapse or progression (AETHERA): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2015;385(9980):1853–1862.
    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(5):1141–1151.
    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(4):661–669.
    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(4):657–670.
    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. 2016;17(7):965–975.
    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. Neuro Oncol. 2017;20(6):838–847.
    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(6):1209–1217.
    1. Wen PY, Macdonald DR, Reardon DA, et al. . Updated response assessment criteria for high-grade gliomas: Response Assessment In Neuro-Oncology working group. J Clin Oncol. 2010;28(11):1963–1972.
    1. Ohgaki H, Kleihues P. The definition of primary and secondary glioblastoma. Clin Cancer Res. 2013;19(4):764–772.
    1. Cohen AL, Holmen SL, Colman H. IDH1 and IDH2 mutations in gliomas. Curr Neurol Neurosci Rep. 2013;13(5):345.
    1. Gerber DE, Grossman SA, Zeltzman M, Parisi MA, Kleinberg L. The impact of thrombocytopenia from temozolomide and radiation in newly diagnosed adults with high-grade gliomas. Neuro Oncol. 2007;9(1):47–52.
    1. Food and Drug Administration. Safety Reporting Requirements for INDs and BA/BE Studies. U.S. Department of Health and Human Services, Food and Drug Administration, Center for Drug Evaluation and Research (CDER), Center for Biologics Evaluation and Research (CBER); 2012. . Accessed June 27, 2018.
    1. Lassman AB, van den Bent MJ, Gan HK, et al. . 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. J Clin Oncol. 2017;35:2003.
    1. van den Bent M, Eoli M, Sepulveda JM, et al. . 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. Neuro Oncol. 2017;19:vi316.

Source: PubMed

3
구독하다