Phase Ib Trial of the Combination of Imatinib and Binimetinib in Patients with Advanced Gastrointestinal Stromal Tumors

Ping Chi, Li-Xuan Qin, Niedzica Camacho, Ciara M Kelly, Sandra P D'Angelo, Mark A Dickson, Mrinal M Gounder, Mary L Keohan, Sujana Movva, Benjamin A Nacev, Evan Rosenbaum, Katherine A Thornton, Aimee M Crago, Jasmine H Francis, Moriah Martindale, Haley T Phelan, Matthew D Biniakewitz, Cindy J Lee, Samuel Singer, Sinchun Hwang, Michael F Berger, Yu Chen, Cristina R Antonescu, William D Tap, Ping Chi, Li-Xuan Qin, Niedzica Camacho, Ciara M Kelly, Sandra P D'Angelo, Mark A Dickson, Mrinal M Gounder, Mary L Keohan, Sujana Movva, Benjamin A Nacev, Evan Rosenbaum, Katherine A Thornton, Aimee M Crago, Jasmine H Francis, Moriah Martindale, Haley T Phelan, Matthew D Biniakewitz, Cindy J Lee, Samuel Singer, Sinchun Hwang, Michael F Berger, Yu Chen, Cristina R Antonescu, William D Tap

Abstract

Purpose: This phase Ib trial was designed to evaluate the safety and early efficacy signal of the combination of imatinib and binimetinib in patients with imatinib-resistant advanced gastrointestinal stromal tumors (GISTs).

Patients and methods: This trial used a standard 3 + 3 design to determine the recommended phase II dose (RP2D). Additional patients were enrolled on an expansion cohort at the RP2D enriching for succinate dehydrogenase (SDH)-deficient GISTs to explore potential efficacy.

Results: The trial enrolled nine patients in the dose-escalation cohort and 14 in the dose-expansion cohort including six with SDH-deficient GISTs. Imatinib 400 mg daily with binimetinib 45 mg twice daily was established as the RP2D. Dose-limiting toxicity (DLT) was asymptomatic grade 4 creatinine phosphokinase (CPK) elevation. The most common non-DLT grade 3/4 toxicity was asymptomatic CPK elevation (69.6%). Other common ≥grade 2 toxicities included peripheral edema (17.4%), acneiform rash (21.7%), anemia (30.4%), hypophosphatemia (39.1%), and aspartate aminotransferase (AST) increase (17.4%). Two serious adverse events occurred (grade 2 dropped head syndrome and grade 3 central retinal vein occlusion). No unexpected toxicities were observed. Limited clinical activity was observed in KIT-mutant GIST. For SDH-deficient GISTs, one of five had confirmed RECIST1.1 partial response (PR). The median progression-free survival (mPFS) in patients with SDH-deficient GIST was 45.1 months [95% confidence interval (CI), 15.8-not estimable (NE)]; the median overall survival (mOS) was not reached (95% CI, 31.6 months-NE). One patient with a refractory metastatic SDH-deficient GIST had an exceptional pathologic response and durable clinical benefit.

Conclusions: The combination of imatinib and binimetinib is safe with manageable toxicity and has encouraging activity in SDH-deficient but not imatinib-refractory KIT/PDGFRA-mutant GISTs. The observed clinical benefits provide a motivation for a larger trial of the combination strategy in SDH-deficient GISTs.

Trial registration: ClinicalTrials.gov NCT01623349.

©2022 The Authors; Published by the American Association for Cancer Research.

Figures

Figure 1.
Figure 1.
Response rates (RECIST1.1, Choi) and duration of treatment. A, Response rates by RECIST1.1 and Choi criteria. Best objective responses by RECIST1.1 (n = 22), Choi responses (n = 21) around 8 weeks (end of cycle 2, first posttreatment scan) on combination imatinib and binimetinib treatment. The best RECIST1.1 responses are shown as percentage of change from baseline for patients who received the combination of imatinib and binimetinib and with at least one postbaseline scan. The known associated primary driver mutations in KIT, SDH complex, NF1, and BRAF are shown. The best ORR for all phase Ib patients was 4.5% (1/22 confirmed PR), two-sided 95% CI, 0.1 to 22.8. The best ORR for patients with SDH-deficient GIST was 20.0% (1/5 confirmed PR), two-sided 95% CI, 0.5 to 71.6. Choi response rate was 47.6% (95% CI, 25.7–70.2) and 60.0% (95% CI, 14.7–94.7) for all phase Ib patients and patients with SDH-deficient GISTs, respectively. B, Duration of treatment. AE, adverse events; PD, progression of disease; PR, partial response; SD, stable disease.
Figure 2.
Figure 2.
Kaplan–Meier estimates of PFS and OS. PFS by RECIST1.1 (A) and OS (B) of all phase Ib patients. PFS (C) and OS (D) of patients with SDH-deficient GIST. mPFS is based on a Kaplan–Meier estimate of PFS, per investigator assessment. The mPFS was 4.2 months (95% CI, 2.8–15.8 months) and 45.1 months (95% CI, 15.8 months–NE) for all phase Ib patients and patients with SDH-deficient GISTs, respectively. The mOS was 23.7 months (95% CI, 10.0 months–NE), and not estimable (95% CI, 31.6 months–NE) for all phase Ib patients and patients with SDH-deficient GISTs, respectively.
Figure 3.
Figure 3.
An example of durable treatment response in a patient with an SDH-deficient GIST. A, Treatment timeline and duration of various treatments the patient received for a metastatic SDH-deficient GIST. B, Representative CT images of the patient's metastatic liver lesions before, approximately 11 months and approximately 22 months after receiving the imatinib plus binimetinib combination therapy. One-dimension measurement in centimeters was provided for RECIST1.1 calculation at different time points. C, Treatment response by histopathologic studies. Representative images of histology and IHC stains for SDHB and SDHA, demonstrating dual SDH-deficiency, in the pretreatment tumor samples (debulking surgery; November 20, 2012) and the histology and the proliferation index marker, Ki67 IHC. The histology from the pretrial treatment liver lesions demonstrated more than 95% viable tumor and less than 5% treatment-associated necrosis. The histology from the on-treatment tumor samples (biopsy; November 5, 2015) showed 100% pathologic response with treatment-associated fibrosis, hyalinization, and dystrophic calcification in a resected metastatic peritoneal nodule and 70% pathologic response with treatment-associated necrosis in the metastatic liver lesion and less than 10% Ki67 IHC in the residual viable tumor. H&E, hematoxylin and eosin.
Figure 4.
Figure 4.
Molecular analyses of archived GISTs and the SDH-deficient GIST patient's pre- (debulking surgery; November 20, 2012) and on-treatment (biopsy; November 5, 2015) tumor samples. A, Normalized RNA expression levels of KIT and ETV1 from U133A microarray analyses in MSKCC archived GIST samples harboring KIT (n = 29) or PDGFRA (n = 4) activating mutations, or KIT/PDGFRA WT and SDH-deficient GIST (n = 15). Each dot represents one tumor sample. Error bars: 95% CI. B, Relative RNA expression level of KIT and ETV1 compared with RPL27 (a housekeeping gene) by qRT-PCR in the patient's FFPE pretreatment tumor samples, the ETV1-expressing GIST48 human cell line, and the ETV1-negative tumor samples control. n = 3, technical replicates; error bars: mean ± S.D. Genetic alterations identified by MSK-IMPACT assays (C) and by WES (D) of the pretreatment and on-treatment FFPE tumor samples from the patient with SDH-deficient GIST shown in Fig. 4. The MSK-IMPACT assay included the mean and minimum coverage of the targeted exons and the allele frequency detected for each permutation (C). *, the mutations detected by MSK-IMPACT. SDHA nonsense mutation was not detected by WES due to lack of coverage (D). But KDR missense mutation was identified in both the WES and IMPACT.

References

    1. Corless CL, Barnett CM, Heinrich MC. Gastrointestinal stromal tumours: origin and molecular oncology. Nat Rev Cancer 2011;11:865–78.
    1. Heinrich MC, Corless CL, Duensing A, McGreevey L, Chen CJ, Joseph N, et al. . PDGFRA activating mutations in gastrointestinal stromal tumors. Science 2003;299:708–10.
    1. Hirota S, Isozaki K, Moriyama Y, Hashimoto K, Nishida T, Ishiguro S, et al. . Gain-of-function mutations of c-kit in human gastrointestinal stromal tumors. Science 1998;279:577–80.
    1. Boikos SA, Pappo AS, Killian JK, LaQuaglia MP, Weldon CB, George S, et al. . Molecular subtypes of KIT/PDGFRA wild-type gastrointestinal stromal tumors: a report from the national institutes of health gastrointestinal stromal tumor clinic. JAMA Oncol 2016;2:922–8.
    1. Killian JK, Kim SY, Miettinen M, Smith C, Merino M, Tsokos M, et al. . Succinate dehydrogenase mutation underlies global epigenomic divergence in gastrointestinal stromal tumor. Cancer Discov 2013;3:648–57.
    1. Flavahan WA, Drier Y, Johnstone SE, Hemming ML, Tarjan DR, Hegazi E, et al. . Altered chromosomal topology drives oncogenic programs in SDH-deficient GISTs. Nature 2019;575:229–33.
    1. Janeway KA, Liegl B, Harlow A, Le C, Perez-Atayde A, Kozakewich H, et al. . Pediatric KIT wild-type and platelet-derived growth factor receptor alpha-wild-type gastrointestinal stromal tumors share KIT activation but not mechanisms of genetic progression with adult gastrointestinal stromal tumors. Cancer Res 2007;67:9084–8.
    1. Chi P, Chen Y, Zhang L, Guo X, Wongvipat J, Shamu T, et al. . ETV1 is a lineage survival factor that cooperates with KIT in gastrointestinal stromal tumours. Nature 2010;467:849–53.
    1. Ran L, Sirota I, Cao Z, Murphy D, Chen Y, Shukla S, et al. . Combined inhibition of MAP kinase and KIT signaling synergistically destabilizes ETV1 and suppresses GIST tumor growth. Cancer Discov 2015;5:304–15.
    1. Hayashi Y, Bardsley MR, Toyomasu Y, Milosavljevic S, Gajdos GB, Choi KM, et al. . Platelet-derived growth factor receptor-alpha regulates proliferation of gastrointestinal stromal tumor cells with mutations in KIT by stabilizing ETV1. Gastroenterology 2015;149:420–32.
    1. Ostrowski J, Polkowski M, Paziewska A, Skrzypczak M, Goryca K, Rubel T, et al. . Functional features of gene expression profiles differentiating gastrointestinal stromal tumours according to KIT mutations and expression. BMC Cancer 2009;9:413.
    1. Xie Y, Cao Z, Wong EW, Guan Y, Ma W, Zhang JQ, et al. . COP1/DET1/ETS axis regulates ERK transcriptome and sensitivity to MAPK inhibitors. J ClinInvest 2018;128:1442–57.
    1. Wang B, Krall EB, Aguirre AJ, Kim M, Widlund HR, Doshi MB, et al. . ATXN1L, CIC, and ETS transcription factors modulate sensitivity to MAPK pathway inhibition. Cell Rep 2017;18:1543–57.
    1. Lito P, Rosen N, Solit DB. Tumor adaptation and resistance to RAF inhibitors. Nat Med 2013;19:1401–9.
    1. Chi P, Qin L, D'Angelo SP, Dickson MA, Gounder MM, Keohan ML, et al. . A phase Ib/II study of MEK162 (binimetinib [BINI]) in combination with imatinib in patients with advanced gastrointestinal stromal tumor (GIST). J Clin Oncol 33:15s, 2015(suppl; abstr 10507).
    1. Chi P, Qin LX, Nguyen B, Kelly CM, D'Angelo SP, Dickson MA, et al. . Phase II trial of imatinib plus binimetinib in patients with treatment-naive advanced gastrointestinal stromal tumor. J Clin Oncol 2022 Jan 18:JCO2102029. doi: 10.1200/JCO.21.02029. Online ahead of print. Available from: .
    1. Ji Y, Li Y, Nebiyou Bekele B. Dose-finding in phase I clinical trials based on toxicity probability intervals. Clin Trials 2007;4:235–44.
    1. Ascierto PA, Schadendorf D, Berking C, Agarwala SS, van Herpen CM, Queirolo P, et al. . MEK162 for patients with advanced melanoma harbouring NRAS or Val600 BRAF mutations: a non-randomised, open-label phase 2 study. Lancet Oncol 2013;14:249–56.
    1. Choi H, Charnsangavej C, Faria SC, Macapinlac HA, Burgess MA, Patel SR, et al. . Correlation of computed tomography and positron emission tomography in patients with metastatic gastrointestinal stromal tumor treated at a single institution with imatinib mesylate: proposal of new computed tomography response criteria. J Clin Oncol 2007;25:1753–9.
    1. Cheng DT, Mitchell TN, Zehir A, Shah RH, Benayed R, Syed A, et al. . Memorial sloan kettering-integrated mutation profiling of actionable cancer targets (MSK-IMPACT): a hybridization capture-based next-generation sequencing clinical assay for solid tumor molecular oncology. J Mol Diagn 2015;17:251–64.
    1. Won HH, Scott SN, Brannon AR, Shah RH, Berger MF. Detecting somatic genetic alterations in tumor specimens by exon capture and massively parallel sequencing. J Vis Exp 2013;80:e50710.
    1. Antonescu CR, Viale A, Sarran L, Tschernyavsky SJ, Gonen M, Segal NH, et al. . Gene expression in gastrointestinal stromal tumors is distinguished by KIT genotype and anatomic site. Clin Cancer Res 2004;10:3282–90.
    1. Francis JH, Diamond EL, Chi P, Jaben K, Hyman DM, Abramson DH. MEK inhibitor-associated central retinal vein occlusion associated with hyperhomocysteinemia and MTHFR variants. Ocul Oncol Pathol 2020;6:159–63.
    1. Chen X, Schwartz GK, DeAngelis LM, Kaley T, Carvajal RD. Dropped head syndrome: report of three cases during treatment with a MEK inhibitor. Neurology 2012;79:1929–31.
    1. Berger M, Amini-Adlé M, Maucort-Boulch D, Robinson P, Robinson P, Thomas L, et al. . Left ventricular ejection fraction decrease related to BRAF and/or MEK inhibitors in metastatic melanoma patients: a retrospective analysis. Cancer Med 2020;9:2611–20.
    1. Blay JY, Shen L, Kang YK, Rutkowski P, Qin S, Nosov D, et al. . Nilotinib versus imatinib as first-line therapy for patients with unresectable or metastatic gastrointestinal stromal tumours (ENESTg1): a randomised phase 3 trial. Lancet Oncol 2015;16:550–60.
    1. Bardsley MR, Horvath VJ, Asuzu DT, Lorincz A, Redelman D, Hayashi Y, et al. . Kitlow stem cells cause resistance to Kit/platelet-derived growth factor alpha inhibitors in murine gastrointestinal stromal tumors. Gastroenterology 2010;139:942–52.
    1. Heinrich MC, Rankin C, Blanke CD, Demetri GD, Borden EC, Ryan CW, et al. . Correlation of long-term results of imatinib in advanced gastrointestinal stromal tumors with next-generation sequencing results: analysis of phase 3 SWOG intergroup trial S0033. JAMA Oncol 2017;3:944–52.
    1. Rutkowski P, Magnan H, Chou AJ, Benson C. Treatment of gastrointestinal stromal tumours in paediatric and young adult patients with sunitinib: a multicentre case series. BMC Cancer 2017;17:717.
    1. Janeway KA, Albritton KH, Van Den Abbeele AD, D'Amato GZ, Pedrazzoli P, Siena S, et al. . Sunitinib treatment in pediatric patients with advanced GIST following failure of imatinib. Pediatr Blood Cancer 2009;52:767–71.
    1. Ben-Ami E, Barysauskas CM, von Mehren M, Heinrich MC, Corless CL, Butrynski JE, et al. . Long-term follow-up results of the multicenter phase II trial of regorafenib in patients with metastatic and/or unresectable GI stromal tumor after failure of standard tyrosine kinase inhibitor therapy. Ann Oncol 2016;27:1794–9.
    1. von Mehren M, George S, Heinrich MC, Schuetze SM, Yap JT, Yu JQ, et al. . Linsitinib (OSI-906) for the treatment of adult and pediatric wild-type gastrointestinal stromal tumors, a SARC phase II study. Clin Cancer Res 2020;26:1837–45.
    1. Glod J, Arnaldez FI, Wiener L, Spencer M, Killian JK, Meltzer P, et al. . A phase II trial of vandetanib in children and adults with succinate dehydrogenase-deficient gastrointestinal stromal tumor. Clin Cancer Res 2019;25:6302–8.

Source: PubMed

3
구독하다