Entrectinib in children and young adults with solid or primary CNS tumors harboring NTRK, ROS1, or ALK aberrations (STARTRK-NG)

Ami V Desai, Giles W Robinson, Karen Gauvain, Ellen M Basu, Margaret E Macy, Luke Maese, Nicholas S Whipple, Amit J Sabnis, Jennifer H Foster, Suzanne Shusterman, Janet Yoon, Brian D Weiss, Mohamed S Abdelbaki, Amy E Armstrong, Thomas Cash, Christine A Pratilas, Nadège Corradini, Lynley V Marshall, Mufiza Farid-Kapadia, Saibah Chohan, Clare Devlin, Georgina Meneses-Lorente, Alison Cardenas, Katherine E Hutchinson, Guillaume Bergthold, Hubert Caron, Edna Chow Maneval, Amar Gajjar, Elizabeth Fox, Ami V Desai, Giles W Robinson, Karen Gauvain, Ellen M Basu, Margaret E Macy, Luke Maese, Nicholas S Whipple, Amit J Sabnis, Jennifer H Foster, Suzanne Shusterman, Janet Yoon, Brian D Weiss, Mohamed S Abdelbaki, Amy E Armstrong, Thomas Cash, Christine A Pratilas, Nadège Corradini, Lynley V Marshall, Mufiza Farid-Kapadia, Saibah Chohan, Clare Devlin, Georgina Meneses-Lorente, Alison Cardenas, Katherine E Hutchinson, Guillaume Bergthold, Hubert Caron, Edna Chow Maneval, Amar Gajjar, Elizabeth Fox

Abstract

Background: Entrectinib is a TRKA/B/C, ROS1, ALK tyrosine kinase inhibitor approved for the treatment of adults and children aged ≥12 years with NTRK fusion-positive solid tumors and adults with ROS1 fusion-positive non-small-cell lung cancer. We report an analysis of the STARTRK-NG trial, investigating the recommended phase 2 dose (RP2D) and activity of entrectinib in pediatric patients with solid tumors including primary central nervous system tumors.

Methods: STARTRK-NG (NCT02650401) is a phase 1/2 trial. Phase 1, dose-escalation of oral, once-daily entrectinib, enrolled patients aged <22 years with solid tumors with/without target NTRK1/2/3, ROS1, or ALK fusions. Phase 2, basket trial at the RP2D, enrolled patients with intracranial or extracranial solid tumors harboring target fusions or neuroblastoma. Primary endpoints: phase 1, RP2D based on toxicity; phase 2, objective response rate (ORR) in patients harboring target fusions. Safety-evaluable patients: ≥1 dose of entrectinib; response-evaluable patients: measurable/evaluable baseline disease and ≥1 dose at RP2D.

Results: At data cutoff, 43 patients, median age of 7 years, were response-evaluable. In phase 1, 4 patients experienced dose-limiting toxicities. The most common treatment-related adverse event was weight gain (48.8%). Nine patients experienced bone fractures (20.9%). In patients with fusion-positive tumors, ORR was 57.7% (95% CI 36.9-76.7), median duration of response was not reached, and median (interquartile range) duration of treatment was 10.6 months (4.2-18.4).

Conclusions: Entrectinib resulted in rapid and durable responses in pediatric patients with solid tumors harboring NTRK1/2/3 or ROS1 fusions.

Keywords: CNS tumors; entrectinib; pediatric; recommended phase 2 dose; solid tumors.

© The Author(s) 2022. Published by Oxford University Press on behalf of the Society for Neuro-Oncology.

Figures

Fig. 1
Fig. 1
CONSORT diagram. aFollowing implementation of protocol version 5 (October 25, 2018), enrollment in cohorts C and E was closed; cohort A was completed following dose-escalation recruitment. Enrollment continued in cohorts B (for patients with primary CNS tumors with gene fusions) and D (for patients with extracranial tumors with gene fusions). Following closure of cohort E, new patients that were unable to swallow capsules were enrolled in cohorts B and D, depending on their tumor type. Molecular testing was required, prior to enrollment in cohorts B or D.  bPatients in cohorts B, C, and D received entrectinib at the MTD (F1 formulation at 550 mg/m2 or F06 formulation at 300 mg/m2), and patients in cohort E received entrectinib at 400 mg/m2 (F1 formulation) mixed with soft foods. cPrimary CNS tumors, n = 5; extracranial solid tumors, n = 3; neuroblastoma, n = 1. The patient with neuroblastoma was not merged into cohorts B or D; they were followed up separately. Abbreviations: CNS, central nervous system; F1, F1 formulation; F06, F06 formulation; MTD, maximum tolerated dose; NTRK, neurotrophic tyrosine receptor kinase; ROS1, ROS proto-oncogene 1.
Fig. 2
Fig. 2
Responses to entrectinib as assessed by BICR in patients with tumors harboring target gene fusions (n = 26). (A) Waterfall plot of BICR-assessed maximum percentage change in tumor size from baseline as measured by RECIST or RANO in patients with measurable target lesions. Plot includes 21 patients with both baseline and post-baseline measurements available for SLD or SPD. Five patients were excluded due to the presence of non-target lesions only (n = 3) or non-evaluable response (n = 2). Best overall confirmed responses per BICR assessment are also indicated; note that confirmed response does not align with best percentage improvement from baseline in SLD/SPD in 5 patients due to consideration of response in non-target lesions (n = 2), development of new lesions (n = 1), and requirement for confirmation of response after ≥28 days (n = 2; initial PR unconfirmed due to subsequent surgical resection [n = 1] or new lesion [n = 1]). (B) Swimmer plot of BICR-assessed best overall response from start of therapy to time of last therapy. Abbreviations: ALK, anaplastic lymphoma kinase; BICR, blinded independent central review; CNS, central nervous system; CR, complete response; HGG, high-grade glioma; IMT, inflammatory myofibroblastic tumors; NOS, not otherwise specified; NTRK, neurotrophic tyrosine receptor kinase; PD, progressive disease; PR, partial response; RANO, Response Assessment in Neuro-Oncology; RECIST, Response Evaluation Criteria in Solid Tumors; ROS1, ROS proto-oncogene 1; SD, stable disease; SLD, sum of longest diameter; SPD, sum of products of diameters.
Fig. 3
Fig. 3
Example MRI scans showing measurable and durable responses to entrectinib. (A) Primary CNS tumors. (B) An infant with a primary CNS tumor determined to be an anaplastic astrocytoma harboring an ETV6-NTRK3 fusion. Patient remains on treatment after 1 year. (C) Extracranial solid tumors. Per protocol, entrectinib was administered in 28-day cycles. Abbreviations: CNS, central nervous system; IMT, inflammatory myofibroblastic tumor; MRI, magnetic resonance imaging; NTRK, neurotrophic tyrosine receptor kinase; ROS1, ROS proto-oncogene 1.

References

    1. Dziadziuszko R, Krebs MG, De Braud F, et al. Updated integrated analysis of the efficacy and safety of entrectinib in locally advanced or metastatic ROS1 fusion-positive non–small-cell lung cancer. J Clin Oncol. 2021;39(11):1253–1263.
    1. Dupain C, Harttrampf AC, Urbinati G, Geoerger B, Massaad-Massade L. Relevance of fusion genes in pediatric cancers: toward precision medicine. Mol Ther Nucleic Acids. 2017;6:315–326.
    1. Okamura R, Boichard A, Kato S, et al. Analysis of NTRK alterations in pan-cancer adult and pediatric malignancies: implications for NTRK-targeted therapeutics. JCO Precis Oncol. 2018;2:PO.18.00183.
    1. Albert CM, Davis JL, Federman N, Casanova M, Laetsch TW. TRK fusion cancers in children: a clinical review and recommendations for screening. J Clin Oncol. 2019;37(6):513–524.
    1. Cocco E, Scaltriti M, Drilon A. NTRK fusion-positive cancers and TRK inhibitor therapy. Nat Rev Clin Oncol. 2018;15(12):731–747.
    1. Guerreiro Stucklin AS, Ryall S, Fukuoka K, et al. Alterations in ALK/ROS1/NTRK/MET drive a group of infantile hemispheric gliomas. Nat Commun. 2019;10(1):4343.
    1. Yamamoto H, Yoshida A, Taguchi K, et al. ALK, ROS1 and NTRK3 gene rearrangements in inflammatory myofibroblastic tumours. Histopathology. 2016;69(1):72–83.
    1. Ross JS, Ali SM, Fasan O, et al. ALK fusions in a wide variety of tumor types respond to anti-ALK targeted therapy. Oncologist. 2017;22(12):1444–1450.
    1. Trigg RM, Turner SD. ALK in neuroblastoma: biological and therapeutic implications. Cancers (Basel). 2018;10(4):113.
    1. Brodeur GM, Minturn JE, Ho R, et al. Trk receptor expression and inhibition in neuroblastomas. Clin Cancer Res. 2009;15(10):3244–3250.
    1. Fischer H, Ullah M, de la Cruz CC, et al. Entrectinib, a TRK/ROS1 inhibitor with anti-CNS tumor activity: differentiation from other inhibitors in its class due to weak interaction with P-glycoprotein. Neuro Oncol. 2020;22(6):819–829.
    1. EMA. ROZLYTREK (Entrectinib). 2020. Available at . Accessed January 17, 2022.
    1. FDA. ROZLYTREK Prescribing Information. 2019. Available at . Accessed January 17, 2022.
    1. Doebele RC, Drilon A, Paz-Ares L, et al. Entrectinib in patients with advanced or metastatic NTRK fusion-positive solid tumours: integrated analysis of three phase 1–2 trials. Lancet Oncol. 2020;21(2):271–282.
    1. Drilon A, Siena S, Dziadziuszko R, et al. Entrectinib in ROS1 fusion-positive non–small-cell lung cancer: integrated analysis of three phase 1–2 trials. Lancet Oncol. 2020;21(2):261–270.
    1. Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45(2):228–247.
    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. Ady N, Zucker JM, Asselain B, et al. A new 123I-MIBG whole body scan scoring method—application to the prediction of the response of metastases to induction chemotherapy in stage IV neuroblastoma. Eur J Cancer. 1995;31A(2):256–261.
    1. Fox E, Maris JM, Widemann BC, et al. A phase I study of ABT-751, an orally bioavailable tubulin inhibitor, administered daily for 21 days every 28 days in pediatric patients with solid tumors. Clin Cancer Res. 2008;14(4):1111–1115.
    1. Gonzalez-Sales M, Djebli N, Meneses-Lorente G, et al. Population pharmacokinetic analysis of entrectinib in pediatric and adult patients with advanced/metastatic solid tumors: support of new drug application submission. Cancer Chemother Pharmacol. 2021;88(6):997–1007.
    1. Djebli N, Buchheit V, Parrott N, et al. Physiologically-based pharmacokinetic modelling of entrectinib parent and active metabolite to support regulatory decision-making. Eur J Drug Metab Pharmacokinet. 2021;46(6):779–791.
    1. Parrott N, Stillhart C, Lindenberg M, et al. Physiologically based absorption modelling to explore the impact of food and gastric pH changes on the pharmacokinetics of entrectinib. AAPS J. 2020;22(4):78.
    1. Shaw AT, Solomon BJ, Chiari R, et al. Lorlatinib in advanced ROS1-positive non–small-cell lung cancer: a multicentre, open-label, single-arm, phase 1-2 trial. Lancet Oncol. 2019;20(12):1691–1701.
    1. Su Y, Zhou X, Foster B, et al. Roles of neurotrophins in skeletal tissue formation and healing. J Cell Physiol. 2018;233(3):2133–2145.
    1. Meneses-Lorente G, Bentley D, Guerini E. Characterization of the pharmacokinetics of entrectinib and its active M5 metabolite in healthy volunteers and patients with solid tumors. Invest New Drugs. 2021;39(3):803–811.
    1. Djebli N, Parrot N, Mercier F, et al. Combined use of population and physiologically-based PK to support pediatric dose recommendation of entrectinib. Presented at the American Society for Clinical Pharmacology and Therapeutics 2020 (Poster P1-020). 2020. Available at . Accessed 13 April 2022.
    1. Hong DS, DuBois SG, Kummar S, et al. Larotrectinib in patients with TRK fusion-positive solid tumours: a pooled analysis of three phase 1/2 clinical trials. Lancet Oncol. 2020;21(4):531–540.
    1. Pacenta HL, Macy ME. Entrectinib and other ALK/TRK inhibitors for the treatment of neuroblastoma. Drug Des Devel Ther. 2018;12:3549–3561.
    1. Sturm D, Pfister SM, Jones DTW. Pediatric gliomas: current concepts on diagnosis, biology, and clinical management. J Clin Oncol. 2017;35(21):2370–2377.
    1. Karremann M, Gielen GH, Hoffmann M, et al. Diffuse high-grade gliomas with H3 K27M mutations carry a dismal prognosis independent of tumor location. Neuro Oncol. 2018;20(1):123–131.
    1. Zhao X, Kotch C, Fox E, et al. NTRK fusions identified in pediatric tumors: the frequency, fusion partners, and clinical outcome. JCO Precis Oncol. 2021;5:204–214.
    1. Clarke M, Mackay A, Ismer B, et al. Infant high-grade gliomas comprise multiple subgroups characterized by novel targetable gene fusions and favorable outcomes. Cancer Discov. 2020;10(7):942–963.

Source: PubMed

3
Abonnieren