INTELLANCE 2/EORTC 1410 randomized phase II study of Depatux-M alone and with temozolomide vs temozolomide or lomustine in recurrent EGFR amplified glioblastoma

Martin Van Den Bent, Marica Eoli, Juan Manuel Sepulveda, Marion Smits, Annemiek Walenkamp, Jean-Sebastian Frenel, Enrico Franceschi, Paul M Clement, Olivier Chinot, Filip De Vos, Nicolas Whenham, Paul Sanghera, Michael Weller, H J Dubbink, Pim French, Jim Looman, Jyotirmoy Dey, Scott Krause, Pete Ansell, Sarah Nuyens, Maarten Spruyt, Joana Brilhante, Corneel Coens, Thierry Gorlia, Vassilis Golfinopoulos, Martin Van Den Bent, Marica Eoli, Juan Manuel Sepulveda, Marion Smits, Annemiek Walenkamp, Jean-Sebastian Frenel, Enrico Franceschi, Paul M Clement, Olivier Chinot, Filip De Vos, Nicolas Whenham, Paul Sanghera, Michael Weller, H J Dubbink, Pim French, Jim Looman, Jyotirmoy Dey, Scott Krause, Pete Ansell, Sarah Nuyens, Maarten Spruyt, Joana Brilhante, Corneel Coens, Thierry Gorlia, Vassilis Golfinopoulos

Abstract

Background: Depatuxizumab mafodotin (Depatux-M) is a tumor-specific antibody-drug conjugate consisting of an antibody (ABT-806) directed against activated epidermal growth factor receptor (EGFR) and the toxin monomethylauristatin-F. We investigated Depatux-M in combination with temozolomide or as a single agent in a randomized controlled phase II trial in recurrent EGFR amplified glioblastoma.

Methods: Eligible were patients with centrally confirmed EGFR amplified glioblastoma at first recurrence after chemo-irradiation with temozolomide. Patients were randomized to either Depatux-M 1.25 mg/kg every 2 weeks intravenously, or this treatment combined with temozolomide 150-200 mg/m2 day 1-5 every 4 weeks, or either lomustine or temozolomide. The primary endpoint of the study was overall survival.

Results: Two hundred sixty patients were randomized. In the primary efficacy analysis with 199 events (median follow-up 15.0 mo), the hazard ratio (HR) for the combination arm compared with the control arm was 0.71 (95% CI = 0.50, 1.02; P = 0.062). The efficacy of Depatux-M monotherapy was comparable to that of the control arm (HR = 1.04, 95% CI = 0.73, 1.48; P = 0.83). The most frequent toxicity in Depatux-M treated patients was a reversible corneal epitheliopathy, occurring as grades 3-4 adverse events in 25-30% of patients. In the long-term follow-up analysis with median follow-up of 28.7 months, the HR for the comparison of the combination arm versus the control arm was 0.66 (95% CI = 0.48, 0.93).

Conclusion: This trial suggests a possible role for the use of Depatux-M in combination with temozolomide in EGFR amplified recurrent glioblastoma, especially in patients relapsing well after the end of first-line adjuvant temozolomide treatment. (NCT02343406).

Keywords: Antibody drug conjugate; EGFR; depatux-m; glioblastoma; recurrent.

© The Author(s) 2019. Published by Oxford University Press on behalf of the Society for Neuro-Oncology.

Figures

Fig. 1
Fig. 1
Consolidated Standards of Reporting Trials (CONSORT) flow diagram of EORTC study 1410, at the time of primary analysis.
Fig. 2
Fig. 2
(A) Overall survival (Kaplan–Meier) curve for the comparison between Depatux-M with temozolomide versus the control arm (lomustine or temozolomide) at the time of long-term follow-up. (B) Overall survival (Kaplan–Meier) curve for the comparison between Depatux-M monotherapy versus the control arm (lomustine or TMZ) at the time of long-term follow-up.
Fig. 2
Fig. 2
(A) Overall survival (Kaplan–Meier) curve for the comparison between Depatux-M with temozolomide versus the control arm (lomustine or temozolomide) at the time of long-term follow-up. (B) Overall survival (Kaplan–Meier) curve for the comparison between Depatux-M monotherapy versus the control arm (lomustine or TMZ) at the time of long-term follow-up.

References

    1. Weller M, van den Bent M, Tonn JC, . et al; European Association for Neuro-Oncology (EANO) Task Force on Gliomas. European Association for Neuro-Oncology (EANO) guideline on the diagnosis and treatment of adult astrocytic and oligodendroglial gliomas. Lancet Oncol. 2017;18(6):e315–e329.
    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. Batchelor TT, Mulholland P, Neyns B, et al. . Phase III randomized trial comparing the efficacy of cediranib as monotherapy, and in combination with lomustine, versus lomustine alone in patients with recurrent glioblastoma. J Clin Oncol. 2013;31(26):3212–3218.
    1. Weller M, Tabatabai G, Kästner B, et al. ; DIRECTOR Study Group MGMT Promoter methylation is a strong prognostic biomarker for benefit from dose-intensified temozolomide rechallenge in progressive glioblastoma: the DIRECTOR Trial. Clin Cancer Res. 2015;21(9):2057–2064.
    1. Perry JR, Bélanger K, Mason WP, et al. . Phase II trial of continuous dose-intense temozolomide in recurrent malignant glioma: RESCUE study. J Clin Oncol. 2010;28(12):2051–2057.
    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. Han SJ, Rolston JD, Molinaro AM, et al. . Phase II trial of 7 days on/7 days off temozolmide for recurrent high-grade glioma. Neuro Oncol. 2014;16(9):1255–1262.
    1. van den Bent MJ, Dubbink HJ, Sanson M, et al. . MGMT promoter methylation is prognostic but not predictive for outcome to adjuvant PCV chemotherapy in anaplastic oligodendroglial tumors: a report from EORTC Brain Tumor Group Study 26034. J Clin Oncol. 2009;27(35):5881–5886.
    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. Reardon DA, Nabors LB, Mason WP, et al. ; BI 1200 36 Trial Group and the Canadian Brain Tumour Consortium Phase I/randomized phase II study of afatinib, an irreversible ErbB family blocker, with or without protracted temozolomide in adults with recurrent glioblastoma. Neuro Oncol. 2015;17(3):430–439.
    1. Sepúlveda-Sánchez JM, Vaz MÁ, Balañá C, et al. . Phase II trial of dacomitinib, a pan-human EGFR tyrosine kinase inhibitor, in recurrent glioblastoma patients with EGFR amplification. Neuro Oncol. 2017;19(11):1522–1531.
    1. Hegi ME, Diserens AC, Bady P, et al. . Pathway analysis of glioblastoma tissue after preoperative treatment with the EGFR tyrosine kinase inhibitor gefitinib—a phase II trial. Mol Cancer Ther. 2011;10(6):1102–1112.
    1. Lynch TJ, Bell DW, Sordella R, et al. . Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med. 2004;350(21):2129–2139.
    1. Verma S, Miles D, Gianni L, et al. ; EMILIA Study Group Trastuzumab emtansine for HER2-positive advanced breast cancer. N Engl J Med. 2012;367(19):1783–1791.
    1. Younes A, Bartlett NL, Leonard JP, et al. . Brentuximab vedotin (SGN-35) for relapsed CD30-positive lymphomas. N Engl J Med. 2010;363(19):1812–1821.
    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. Lassman AB, van den Bent MJ, Gan HK, et al. . Safety and efficacy of depatuxizumab mafodotin + temozolomide in patients with EGFR-amplified, recurrent glioblastoma: results from an international phase I multicenter trial. Neuro Oncol. 2019;21(1):106–114.
    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. Felsberg J, Hentschel B, Kaulich K, et al. ; German Glioma Network Epidermal growth factor receptor variant III (EGFRvIII) positivity in EGFR-amplified glioblastomas: prognostic role and comparison between primary and recurrent tumors. Clin Cancer Res. 2017;23(22):6846–6855.
    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(7):965–975.
    1. Esteller M, Garcia-Foncillas J, Andion E, et al. . Inactivation of the DNA-repair gene MGMT and the clinical response of gliomas to alkylating agents. N Engl J Med. 2000;343(19):1350–1354.
    1. Ellingson BM, Bendszus M, Boxerman J, et al. ; Jumpstarting Brain Tumor Drug Development Coalition Imaging Standardization Steering Committee Consensus recommendations for a standardized brain tumor imaging protocol in clinical trials. Neuro Oncol. 2015;17(9): 1188–1198.
    1. van den Bent MJ, Klein M, Smits M, et al. . Final results of the EORTC Brain Tumor Group randomized phase II TAVAREC trial on temozolomide with or without bevacizumab in 1st recurrence grade II/III glioma without 1p/19q co-deletion. JCO 2017;35:2009
    1. Pocock SJ, Simon R. Sequential treatment assignment with balancing for prognostic factors in the controlled clinical trial. Biometrics. 1975;31(1):103–115.
    1. Freedman LS, White SJ. On the use of Pocock and Simon’s method for balancing treatment numbers over prognostic factors in the controlled clinical trial. Biometrics. 1976;32(3):691–694.
    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. Masters JC, Nickens DJ, Xuan D, Shazer RL, Amantea M. Clinical toxicity of antibody drug conjugates: a meta-analysis of payloads. Invest New Drugs. 2018;36(1):121–135.

Source: PubMed

3
Prenumerera