Selumetinib in combination with dexamethasone for the treatment of relapsed/refractory RAS-pathway mutated paediatric and adult acute lymphoblastic leukaemia (SeluDex): study protocol for an international, parallel-group, dose-finding with expansion phase I/II trial

Tobias Menne, Daniel Slade, Joshua Savage, Sarah Johnson, Julie Irving, Pamela Kearns, Ruth Plummer, Geoff Shenton, Gareth J Veal, Britta Vormoor, Josef Vormoor, Lucinda Billingham, Tobias Menne, Daniel Slade, Joshua Savage, Sarah Johnson, Julie Irving, Pamela Kearns, Ruth Plummer, Geoff Shenton, Gareth J Veal, Britta Vormoor, Josef Vormoor, Lucinda Billingham

Abstract

Introduction: Event-free survival rates at 15 years for paediatric patients with relapsed/refractory acute lymphoblastic leukaemia (ALL) are 30%-50%, with 5-year survival for adult patients only 20%. Many patients with newly diagnosed and relapsed ALL harbour somatic RAS-signalling activation mutations. Induction therapy for ALL involves steroids, with preclinical data suggesting the combination of dexamethasone with the MEK1/2 inhibitor, selumetinib (ARRY-142886) has a synergistic anticancer effect.

Methods and analysis: The SeluDex trial is an international, parallel-group, dose-finding with expansion, phase I/II trial to assess the selumetinib/dexamethasone combination in adult and paediatric patients with relapsed/refractory, RAS pathway mutant ALL. The Cancer Research UK Clinical Trials Unit at University of Birmingham is the UK Coordinating Centre, with national hubs in Copenhagen, Denmark; Monza, Italy; Münster, Germany; Paris, France; and Utrecht, Netherlands. Patients with morphologically proven relapsed/refractory or progressive B-cell precursor or T-cell ALL, with demonstrated RAS pathway activating mutations are eligible. Adult patients are >18 years old, ECOG <2 and paediatric <18 years old, Lansky play scale ≥60% or Karnofsky score ≥60%. Phase I primary objective is the recommended phase II dose of selumetinib as defined by occurrence/non-occurrence of dose limiting toxicities using the continual reassessment method; phase II will evaluate preliminary antileukaemic activity of the combination, as defined by morphological response 28 days post-treatment using a Bayesian approach. Target recruitment is between 26 and 42 patients (minimum 13 and maximum 21 per group), depending the number of phase I patients included in phase II.

Ethics and dissemination: Medical ethical committees of all the participating countries have approved the study protocol; initial (UK) ethics approval (17/YH/0123) was granted by Yorkshire & The Humber-Leeds West Research Ethics Committee. Participants are required to provide written informed consent/assent. Results will be disseminated through national and international presentations and peer-reviewed publications.

Trial registration number: ISRCTN92323261.

Keywords: adult oncology; clinical trials; leukaemia; paediatric oncology.

Conflict of interest statement

Competing interests: JI has received research funding from F. Hoffmann-La Roche.

© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY. Published by BMJ.

Figures

Figure 1
Figure 1
SeluDex trial schema SeluDex trial schema showing the patient pathway highlighting screening, trial entry, treatment and follow-up. ALL, acute lymphoblastic leukaemia; BD, two times a day; PD, pharmacodynamic; PIS, patient information sheet; PK, pharmacokinetic.

References

    1. Kaatsch P. Epidemiology of childhood cancer. Cancer Treat Rev 2010;36:277–85. 10.1016/j.ctrv.2010.02.003
    1. Einsiedel HG, von Stackelberg A, Hartmann R, et al. . Long-term outcome in children with relapsed ALL by risk-stratified salvage therapy: results of trial acute lymphoblastic leukemia-relapse study of the Berlin-Frankfurt-Münster group 87. J Clin Oncol 2005;23:7942–50. 10.1200/JCO.2005.01.1031
    1. Bassan R, Hoelzer D. Modern therapy of acute lymphoblastic leukemia. J Clin Oncol 2011;29:532–43. 10.1200/JCO.2010.30.1382
    1. Geyer MB, Hsu M, Devlin SM, et al. . Overall survival among older US adults with ALL remains low despite modest improvement since 1980: SEER analysis. Blood 2017;129:1878–81. 10.1182/blood-2016-11-749507
    1. Irving J, Matheson E, Minto L, et al. . Ras pathway mutations are prevalent in relapsed childhood acute lymphoblastic leukemia and confer sensitivity to MEK inhibition. Blood 2014;124:3420–30. 10.1182/blood-2014-04-531871
    1. Liu Y, Easton J, Shao Y, et al. . The genomic landscape of pediatric and young adult T-lineage acute lymphoblastic leukemia. Nat Genet 2017;49:1211–8. 10.1038/ng.3909
    1. Ryan SL, Matheson E, Grossmann V, et al. . The role of the RAS pathway in iAMP21-ALL. Leukemia 2016;30:1824–31. 10.1038/leu.2016.80
    1. Zhang J, Ding L, Holmfeldt L, et al. . The genetic basis of early T-cell precursor acute lymphoblastic leukaemia. Nature 2012;481:157–63. 10.1038/nature10725
    1. Zhang J, Mullighan CG, Harvey RC, et al. . Key pathways are frequently mutated in high-risk childhood acute lymphoblastic leukemia: a report from the Children’s Oncology Group. Blood 2011;118:3080–7. 10.1182/blood-2011-03-341412
    1. Roberts KG, Li Y, Payne-Turner D, et al. . Targetable kinase-activating lesions in Ph-like acute lymphoblastic leukemia. N Engl J Med 2014;371:1005–15. 10.1056/NEJMoa1403088
    1. Holmfeldt L, Wei L, Diaz-Flores E, et al. . The genomic landscape of hypodiploid acute lymphoblastic leukemia. Nat Genet 2013;45:242–52. 10.1038/ng.2532
    1. Shochat C, Tal N, Bandapalli OR, et al. . Gain-of-function mutations in interleukin-7 receptor-α (IL7R) in childhood acute lymphoblastic leukemias. J Exp Med 2011;208:901–8. 10.1084/jem.20110580
    1. Zenatti PP, Ribeiro D, Li W, et al. . Oncogenic IL7R gain-of-function mutations in childhood T-cell acute lymphoblastic leukemia. Nat Genet 2011;43:932–9. 10.1038/ng.924
    1. Jeong EG, Kim MS, Nam HK, et al. . Somatic mutations of JAK1 and JAK3 in acute Leukemias and solid cancers. Clin Cancer Res 2008;14:3716–21. 10.1158/1078-0432.CCR-07-4839
    1. Goldinger SM, Zimmer L, Schulz C, et al. . Upstream mitogen-activated protein kinase (MAPK) pathway inhibition: MEK inhibitor followed by a BRAF inhibitor in advanced melanoma patients. Eur J Cancer 2014;50:406–10. 10.1016/j.ejca.2013.09.014
    1. Carvajal RD, Piperno-Neumann S, Kapiteijn E, et al. . Selumetinib in combination with dacarbazine in patients with metastatic uveal melanoma: a phase III, multicenter, randomized trial (SUMIT). J Clin Oncol 2018;36:1232–9. 10.1200/JCO.2017.74.1090
    1. Chung V, McDonough S, Philip PA, et al. . Effect of selumetinib and MK-2206 vs oxaliplatin and fluorouracil in patients with metastatic pancreatic cancer after prior therapy. JAMA Oncol 2017;3:516–22. 10.1001/jamaoncol.2016.5383
    1. Krishnamurthy A, Dasari A, Noonan AM, et al. . Phase Ib results of the rational combination of selumetinib and cyclosporin A in advanced solid tumors with an expansion cohort in metastatic colorectal cancer. Cancer Res 2018;78:5398–407. 10.1158/0008-5472.CAN-18-0316
    1. Jänne PA, Shaw AT, Pereira JR, et al. . Selumetinib plus docetaxel for KRAS-mutant advanced non-small-cell lung cancer: a randomised, multicentre, placebo-controlled, phase 2 study. Lancet Oncol 2013;14:38–47. 10.1016/S1470-2045(12)70489-8
    1. Matheson EC, Thomas H, Case M, et al. . Glucocorticoids and selumetinib are highly synergistic in RAS pathway-mutated childhood acute lymphoblastic leukemia through upregulation of BIM. Haematologica 2019;104:1804–11. 10.3324/haematol.2017.185975
    1. McCune J, et al. . In vivo and in vitro induction of human cytochrome P4503A4 by dexamethasone. Clin Pharmacol Ther 2000;68:356–66. 10.1067/mcp.2000.110215
    1. Banerjee A, Jakacki RI, Onar-Thomas A, 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;19:1135–44. 10.1093/neuonc/now282
    1. Dombi E, Baldwin A, Marcus LJ, et al. . Activity of selumetinib in neurofibromatosis type 1–Related plexiform neurofibromas. N Engl J Med 2016;375:2550–60. 10.1056/NEJMoa1605943
    1. O'Quigley J, Pepe M, Fisher L. Continual reassessment method: a practical design for phase 1 clinical trials in cancer. Biometrics 1990;46:33–48. 10.2307/2531628
    1. Program CTE . Common terminology criteria for adverse events (CTCAE), version 4.03. US Department of Health and Human Services, NIH, NCI, 2010.
    1. O'Connor L, et al. . Bim: a novel member of the Bcl-2 family that promotes apoptosis. Embo J 1998;17:384–95. 10.1093/emboj/17.2.384
    1. Goodman SN, Zahurak ML, Piantadosi S. Some practical improvements in the continual reassessment method for phase I studies. Stat Med 1995;14:1149–61. 10.1002/sim.4780141102
    1. RH K, Ji L, Barnette P, et al. . Outcome of patients treated for relapsed or refractory acute lymphoblastic leukemia: a therapeutic advances in childhood leukemia Consortium study. J Clin Oncol 2010;28:648–54.
    1. Nie Y, Lu W, Chen D, et al. . Mechanisms underlying CD19-positive ALL relapse after anti-CD19 CAR T cell therapy and associated strategies. Biomarker Research 2020;8:18. 10.1186/s40364-020-00197-1
    1. Wheeler GM, Mander AP, Bedding A, et al. . How to design a dose-finding study using the continual reassessment method. BMC Med Res Methodol 2019;19:18. 10.1186/s12874-018-0638-z

Source: PubMed

3
Se inscrever