Cobimetinib in Pediatric and Young Adult Patients with Relapsed or Refractory Solid Tumors (iMATRIX-cobi): A Multicenter, Phase I/II Study

Tanya Trippett, Helen Toledano, Quentin Campbell Hewson, Arnauld Verschuur, Anne-Marie Langevin, Isabelle Aerts, Lisa Howell, Soledad Gallego, Claudia Rossig, Amy Smith, Darshak Patel, Leonardo R Pereira, Sravanthi Cheeti, Luna Musib, Katherine E Hutchinson, Clare Devlin, Ronald Bernardi, Birgit Geoerger, Tanya Trippett, Helen Toledano, Quentin Campbell Hewson, Arnauld Verschuur, Anne-Marie Langevin, Isabelle Aerts, Lisa Howell, Soledad Gallego, Claudia Rossig, Amy Smith, Darshak Patel, Leonardo R Pereira, Sravanthi Cheeti, Luna Musib, Katherine E Hutchinson, Clare Devlin, Ronald Bernardi, Birgit Geoerger

Abstract

Background: The MAPK pathway is an emerging target across a number of adult and pediatric tumors. Targeting the downstream effector of MAPK, MEK1, is a proposed strategy to control the growth of MAPK-dependent tumors.

Objective: iMATRIX-cobi assessed the safety, pharmacokinetics, and anti-tumor activity of cobimetinib, a highly selective MEK inhibitor, in children and young adults with relapsed/refractory solid tumors.

Patients and methods: This multicenter Phase I/II study enrolled patients aged 6 months to < 30 years with solid tumors with known/expected MAPK pathway involvement. Patients received cobimetinib tablet or suspension formulation on Days 1-21 of a 28-day cycle. Dose escalation followed a rolling 6 design. The primary endpoint was safety; secondary endpoints were pharmacokinetics and anti-tumor activity.

Results: Of 56 enrolled patients (median age 9 years [range 3-29]), 18 received cobimetinib tablets and 38 cobimetinib suspension. Most common diagnoses were low-grade glioma (LGG; n = 32, including n = 12 in the expansion cohort) and plexiform neurofibroma within neurofibromatosis type 1 (n = 12). Six patients (11 %) experienced dose-limiting toxicities (including five ocular toxicity events), which established a pediatric recommended Phase II dose (RP2D) of 0.8 mg/kg tablet and 1.0 mg/kg suspension. Most frequently reported treatment-related adverse events were gastrointestinal and skin disorders. Steady state mean exposure (Cmax, AUC0-24) of cobimetinib at the RP2D (1.0 mg/kg suspension) was ~ 50 % lower than in adults receiving the approved 60 mg/day dose. Overall response rate was 5.4 % (3/56; all partial responses in patients with LGG).

Conclusions: The safety profile of cobimetinib in pediatrics was similar to that reported in adults. Clinical activity was observed in LGG patients with known/suspected MAPK pathway activation. Cobimetinib combination regimens may be required to improve response rates in this pediatric population.

Clinical trial registration: ClinicalTrials.gov NCT02639546, registered December 24, 2015.

Conflict of interest statement

TT reports consultancy fees from F. Hoffmann-La Roche Ltd. HT reports lecture fees from AstraZeneca and Roche. SG reports personal fees from Bayer, EUSA Pharma, and LOXO Oncology. CR reports personal fees from Amgen, BMS, Celgene, Genentech, Inc., Novartis, Pfizer, and F. Hoffmann-La Roche Ltd. DP was an employee of F. Hoffman-La Roche Ltd. at the time of study conduct. LM was an employee of Genentech, Inc. at the time of study conduct. LRP is an employee of F. Hoffmann-La Roche Ltd. SC, KEH, CD, and RB are employees of and stock-holders in F. Hoffmann-La Roche Ltd/Genentech, Inc. All other authors declare no competing interests.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
Patient status
Fig. 2
Fig. 2
Mean (±SD) plasma concentration-time profiles for cobimetinib following a single dose (cycle 1 Day 1) and b multiple dose (cycle 1 Day 21) administration of oral suspension formulations. SD standard deviation
Fig. 3
Fig. 3
Kaplan-Meier plot of PFS (per RECIST v1.1) for patients with LGG (safety-evaluable patients). CI confidence interval, LGG low-grade glioma, NE not estimable, PFS progression-free survival, RECIST Response Evaluation Criteria in Solid Tumors

References

    1. Roberts PJ, Der CJ. Targeting the Raf-MEK-ERK mitogen-activated protein kinase cascade for the treatment of cancer. Oncogene. 2007;26:3291–3310. doi: 10.1038/sj.onc.1210422.
    1. Forshew T, Tatevossian RG, Lawson AR, Ma J, Neale G, Ogunkolade BW, et al. Activation of the ERK/MAPK pathway: a signature genetic defect in posterior fossa pilocytic astrocytomas. J Pathol. 2009;218:172–181. doi: 10.1002/path.2558.
    1. Sievert AJ, Lang SS, Boucher KL, Madsen PJ, Slaunwhite E, Choudhari N, et al. Paradoxical activation and RAF inhibitor resistance of BRAF protein kinase fusions characterizing pediatric astrocytomas. Proc Natl Acad Sci USA. 2013;110:5957–5962. doi: 10.1073/pnas.1219232110.
    1. Vitagliano O, Addeo R, D’Angelo V, Indolfi C, Indolfi P, Casale F. The Bcl-2/Bax and Ras/Raf/MEK/ERK signaling pathways: implications in pediatric leukemia pathogenesis and new prospects for therapeutic approaches. Expert Rev Hematol. 2013;6:587–597. doi: 10.1586/17474086.2013.827415.
    1. Shern JF, Chen L, Chmielecki J, Wei JS, Patidar R, Rosenberg M, et al. Comprehensive genomic analysis of rhabdomyosarcoma reveals a landscape of alterations affecting a common genetic axis in fusion-positive and fusion-negative tumors. Cancer Discov. 2014;4:216–231. doi: 10.1158/-13-0639.
    1. Knight T, Irving JAE. Ras/Raf/MEK/ERK pathway activation in childhood acute lymphoblastic leukemia and its therapeutic targeting. Front Oncol. 2014;4:160. doi: 10.3389/fonc.2014.00160.
    1. Xie G, Wu H, Cai W, Chen M, Huang W, Yan W, et al. RDM1 promotes neuroblastoma growth through the RAS–Raf–MEK–ERK pathway. FEBS Open Bio. 2019;9:490–497. doi: 10.1002/2211-5463.12586.
    1. Packer RJ, Pfister S, Bouffet E, Avery R, Bandopadhayay P, Bornhorst M, et al. Pediatric low-grade gliomas: Implications of the biologic era. Neuro Oncol. 2017;19:750–761.
    1. Tateishi K, Nakamura T, Yamamoto T. Molecular genetics and therapeutic targets of pediatric low-grade gliomas. Brain Tumor Pathol. 2019;36:74–83. doi: 10.1007/s10014-019-00340-3.
    1. de Blank P, Bandopadhayay P, Haas-Kogan D, Fouladi M, Fangusaro J. Management of pediatric low-grade glioma. Curr Opin Pediatr. 2019;31:21–27. doi: 10.1097/MOP.0000000000000717.
    1. Rosen LS, LoRusso P, Wee Ma W, Goldman JW, Weise A, Colevas AD, et al. A first-in-human phase I study to evaluate the MEK1/2 inhibitor, cobimetinib, administered daily in patients with advanced solid tumors. Invest New Drugs. 2016;34:604–613. doi: 10.1007/s10637-016-0374-3.
    1. Cobimetinib SmPC. . Accessed 1 Oct 2021.
    1. FDA. Cotellic PI. . Accessed 1 Oct 2021.
    1. EMA. Cotellic EPAR summary. . Accessed 1 Oct 2021.
    1. Park JR, Bagatell R, Cohn SL, Pearson AD, Villablanca JG, Berthold F, et al. Revisions to the International Neuroblastoma Response Criteria: a consensus statement from the National Cancer Institute Clinical Trials planning meeting. J Clin Oncol. 2017;35:2580–2587. doi: 10.1200/JCO.2016.72.0177.
    1. Wen PY, Macdonald DR, Reardon DA, Cloughesy TF, Sorensen AG, Galanis E, et al. Updated response assessment criteria for high-grade gliomas: response assessment in neuro-oncology working group. J Clin Oncol. 2012;28:1963–1972. doi: 10.1200/JCO.2009.26.3541.
    1. Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, et al. New response evaluation criteria in solid tumors: revised RECIST guideline (Version 1.1). Eur J Cancer. 2009;45:228–47.
    1. Brookmeyer R, Crowley J. A confidence interval for the median survival time. Biometrics. 1982;38:29–41. doi: 10.2307/2530286.
    1. Budha NR, Ji T, Musib L, Eppler S, Dresser M, Chen Y, et al. Evaluation of cytochrome P450 3A4-mediated drug–drug interaction potential for cobimetinib using physiologically based pharmacokinetic modeling and simulation. Clin Pharmacokinet. 2016;55:1435–1445. doi: 10.1007/s40262-016-0412-5.
    1. Center for Drug Evaluation and Research. Clinical Pharmacology and Biopharmaceutics Filing for Cobimetinib. 2015. . Accessed 1 Oct 2021.
    1. Forrest SJ, Geoerger B, Janeway KA. Precision medicine in pediatric oncology. Curr Opin Pediatr. 2018;30:17–24. doi: 10.1097/MOP.0000000000000570.
    1. Banerjee A, Jakacki RI, Onar-Thomas A, Wu S, Nicolaides T, Poussaint TY, et al. A phase I trial of the MEK inhibitor selumetinib (AZD6244) in pediatric patients with recurrent or refractory low-grade glioma: A Pediatric Brain Tumor Consortium (PBTC) study. Neuro Oncol. 2017;19:1135–1144. doi: 10.1093/neuonc/now282.
    1. Fangusaro J, Onar-Thomas A, Poussaint TY, Wu S, Ligon AH, Lindeman N, et al. Selumetinib in paediatric patients with BRAF-aberrant or neurofibromatosis type 1-associated recurrent, refractory, or progressive low-grade glioma: a multicentre, phase 2 trial. Lancet Oncol. 2019;20:1011–1022. doi: 10.1016/S1470-2045(19)30277-3.
    1. Gross AM, Wolters PL, Dombi E, Baldwin A, Whitcomb P, Fisher MJ, et al. NFM-07. Selumetinib in children with inoperable plexiform neurofibromas. N Engl J Med. 2020;382:1430–42.
    1. McCowage GB, Mueller S, Pratilas CA, Hargrave DR, Moertel CL, Whitlock J, et al. Trametinib in pediatric patients with neurofibromatosis type 1 (NF-1)–associated plexiform neurofibroma: a phase I/IIa study. J Clin Oncol. 2018;36(15_Suppl):10504.
    1. Dombi E, Baldwin A, Marcus LJ, Fisher MJ, Weiss B, Kim AR, et al. Activity of selumetinib in neurofibromatosis type 1-related plexiform neurofibromas. N Engl J Med. 2016;375:2550–2560. doi: 10.1056/NEJMoa1605943.
    1. de Gooijer MC, Zhang P, Weijer R, Buil LCM, Beijnen JH, van Tellingen O. The impact of P-glycoprotein and breast cancer resistance protein on the brain pharmacokinetics and pharmacodynamics of a panel of MEK inhibitors. Int J Cancer. 2018;142:381–391. doi: 10.1002/ijc.31052.
    1. Choo EF, Ly J, Chan J, Shahidi-Latham SK, Messick K, Plise E, et al. Role of P-glycoprotein on the brain penetration and brain pharmacodynamic activity of the MEK inhibitor cobimetinib. Mol Pharm. 2014;11:4199–4207. doi: 10.1021/mp500435s.

Source: PubMed

3
Sottoscrivi