Trametinib in Patients With NF1-, GNAQ-, or GNA11-Mutant Tumors: Results From the NCI-MATCH ECOG-ACRIN Trial (EAY131) Subprotocols S1 and S2

Kari B Wisinski, Yael Flamand, Melissa A Wilson, Jason J Luke, Hussein A Tawbi, Fangxin Hong, Edith P Mitchell, James A Zwiebel, Helen Chen, Robert J Gray, Shuli Li, Lisa M McShane, Lawrence V Rubinstein, David Patton, P Mickey Williams, Stanley R Hamilton, Robert J Behrens, Kathryn P Pennington, Barbara A Conley, Carlos L Arteaga, Lyndsay N Harris, Peter J O'Dwyer, Alice P Chen, Keith T Flaherty, Kari B Wisinski, Yael Flamand, Melissa A Wilson, Jason J Luke, Hussein A Tawbi, Fangxin Hong, Edith P Mitchell, James A Zwiebel, Helen Chen, Robert J Gray, Shuli Li, Lisa M McShane, Lawrence V Rubinstein, David Patton, P Mickey Williams, Stanley R Hamilton, Robert J Behrens, Kathryn P Pennington, Barbara A Conley, Carlos L Arteaga, Lyndsay N Harris, Peter J O'Dwyer, Alice P Chen, Keith T Flaherty

Abstract

Purpose: NCI-MATCH is a precision medicine trial using genomic testing to allocate patients with advanced malignancies to targeted treatment subprotocols. This report combines two subprotocols evaluating trametinib, a MEK1/2 inhibitor, in patients with Neurofibromatosis 1 (NF1[S1] or GNA11/Q [S2]) altered tumors.

Methods: Eligible patients had tumors with deleterious inactivating NF1 or GNA11/Q mutations by the customized Oncomine AmpliSeq panel. Prior MEK inhibitor treatment was excluded. Glioblastomas (GBMs) were permitted, including malignancies associated with germline NF1 mutations (S1 only). Trametinib was administered at 2 mg once daily over 28-day cycles until toxicity or disease progression. Primary end point was objective response rate (ORR). Secondary end points included progression-free survival (PFS) at 6 months, PFS, and overall survival. Exploratory analyses included co-occurring genomic alterations and PTEN loss.

Results: Fifty patients were eligible and started therapy: 46 with NF1 mutations (S1) and four with GNA11 mutations (S2). In the NF1 cohort, nonsense single-nucleotide variants were identified in 29 and frameshift deletions in 17 tumors. All in S2 had nonuveal melanoma and GNA11 Q209L variant. Two partial responses (PR) were noted in S1, one patient each with advanced lung cancer and GBM for an ORR of 4.3% (90% CI, 0.8 to 13.1). One patient with melanoma in S2 had a PR (ORR, 25%; 90% CI, 1.3 to 75.1). Prolonged stable disease (SD) was also noted in five patients (four in S1 and one in S2) with additional rare histologies. Adverse events were as previously described with trametinib. Comutations in TP53 and PIK3CA were common.

Conclusion: Although these subprotocols did not meet the primary end point for ORR, significant responses or prolonged SD noted in some disease subtypes warrants further investigation.

Trial registration: ClinicalTrials.gov NCT02465060.

Conflict of interest statement

The following represents disclosure information provided by authors of this manuscript. All relationships are considered compensated unless otherwise noted. Relationships are self-held unless noted. I = Immediate Family Member, Inst = My Institution. Relationships may not relate to the subject matter of this manuscript. For more information about ASCO's conflict of interest policy, please refer to www.asco.org/rwc or ascopubs.org/po/author-center.

Open Payments is a public database containing information reported by companies about payments made to US-licensed physicians (Open Payments).

Figures

FIG 1.
FIG 1.
Diagrams for subprotocols (A) S1 and (B) S2. aReasons for not receiving an assignment: study suspended (n = 15), assigned to another arm for which the qualifying variant’s level of evidence was higher (n = 7), before study activation (n = 6), assigned to another arm for which accrual was lower (n = 2), and prior treatment of trametinib (n = 1). bReasons not enrolled after receiving an assignment: active infection and abnormal brain MRI (n = 1), deteriorating performance status (n = 6), eye problems (n = 1), in hospice care (n = 1), inadequate organ/marrow function (n = 2), interstitial lung disease/pneumonitis (n = 2), intervening news/targeted treatment (n = 2), other investigational treatment (n = 1), patient died (n = 6), patient refusal (n = 4), receiving other therapy (n = 2), and unknown (n = 3). cReasons for being nonevaluable: no disease assessment done before death (n = 4), no disease assessment done before NPT (n = 1), baseline scans done outside of specified window (n = 5), withdrew consent (n = 2), different methods of evaluation used (n = 1), no evaluation during treatment (n = 1). dReasons for not receiving an assignment: ineligible histology—uveal melanoma (n = 13). eReasons not enrolled after receiving an assignment: central screening cohort: ineligible histology (n = 1), ineligible histology and inadequate organ/marrow function (n = 1), and unknown (n = 1). NPT, nonprotocol therapy.
FIG 1.
FIG 1.
Diagrams for subprotocols (A) S1 and (B) S2. aReasons for not receiving an assignment: study suspended (n = 15), assigned to another arm for which the qualifying variant’s level of evidence was higher (n = 7), before study activation (n = 6), assigned to another arm for which accrual was lower (n = 2), and prior treatment of trametinib (n = 1). bReasons not enrolled after receiving an assignment: active infection and abnormal brain MRI (n = 1), deteriorating performance status (n = 6), eye problems (n = 1), in hospice care (n = 1), inadequate organ/marrow function (n = 2), interstitial lung disease/pneumonitis (n = 2), intervening news/targeted treatment (n = 2), other investigational treatment (n = 1), patient died (n = 6), patient refusal (n = 4), receiving other therapy (n = 2), and unknown (n = 3). cReasons for being nonevaluable: no disease assessment done before death (n = 4), no disease assessment done before NPT (n = 1), baseline scans done outside of specified window (n = 5), withdrew consent (n = 2), different methods of evaluation used (n = 1), no evaluation during treatment (n = 1). dReasons for not receiving an assignment: ineligible histology—uveal melanoma (n = 13). eReasons not enrolled after receiving an assignment: central screening cohort: ineligible histology (n = 1), ineligible histology and inadequate organ/marrow function (n = 1), and unknown (n = 1). NPT, nonprotocol therapy.
FIG 2.
FIG 2.
Subprotocol 1 response and Kaplan-Meier curves for PFS. (A) Waterfall plot (N = 30) of best % change from baseline responses. This includes the 32 evaluable patients, excluding two patients who had a best response of PD with new lesions that had no recorded measurements. (B) Treatment duration in the 15 patients who achieved a best response of SD or PR. (C) Kaplan-Meier curve for PFS. aNew lesions. Cholangio, cholangiocarcinoma; endo, endocrine cancer; gall, gallbladder cancer; GBM, glioblastoma; neuro, peripheral nerve sheath tumor or neurofibroma; neuroendo, neuroendocrine carcinoma; ovar/fallop, ovarian/fallopian tube cancer; PD, progressive disease; PFS, progression-free survival; PR, partial response; stom/small int, gastric or small intestine carcinoma; SD, stable disease.
FIG 3.
FIG 3.
Subprotocol 2 response and Kaplan-Meier curves for PFS. (A) Waterfall plot (N = 4) of best % change from baseline responses. (B) Treatment duration in the three patients who achieved a best response of SD or PR. (C) Kaplan-Meier curve for PFS. NA, not applicable; PD, progressive disease; PFS, progression-free survival; PR, partial response; SD, stable disease.
FIG 4.
FIG 4.
Co-occurring genomic alterations in subprotocols S1 and S2. Co-occurring genomic alterations using the NCI-MATCH assay color-coded by: left: variant type; top: copy-number variant (pink), SNV (red), indel (purple), and unified gene fusion (magenta) for each eligible patient; and bottom: information regarding histology and best response on treatment: PR, SD, PD, and UE. Cholangio, cholangiocarcinoma; endo, endocrine cancer; gall, gallbladder cancer; GBM, glioblastoma; neuro, peripheral nerve sheath tumor or neurofibroma; neuroendo, neuroendocrine carcinoma; ovar/fallop, ovarian/fallopian tube cancer; PD, progressive disease; PFS, progression-free survival; PR, partial response; SD, stable disease; SNV, single-nucleotide variant; stom/small int, gastric or small intestine carcinoma; UE, unevaluable.
FIG A1.
FIG A1.
Lollipop plot showing location and sequence of NF1 mutations in S1 cohort.

References

    1. Downward J. Targeting RAS signalling pathways in cancer therapy. Nat Rev Cancer. 2003;3:11–22.
    1. Imperial R, Toor OM, Hussain A, et al. Comprehensive pancancer genomic analysis reveals (RTK)-RAS-RAF-MEK as a key dysregulated pathway in cancer: Its clinical implications. Semin Cancer Biol. 2019;54:14–28.
    1. Flaherty KT, Robert C, Hersey P, et al. Improved survival with MEK inhibition in BRAF-mutated melanoma. N Engl J Med. 2012;367:107–114.
    1. Patil S, Chamberlain RS. Neoplasms associated with germline and somatic NF1 gene mutations. Oncologist. 2012;17:101–116.
    1. Philpott C, Tovell H, Frayling IM, et al. The NF1 somatic mutational landscape in sporadic human cancers. Hum Genomics. 2017;11:13.
    1. Denayer E, de Ravel T, Legius E. Clinical and molecular aspects of RAS related disorders. J Med Genet. 2008;45:695–703.
    1. See WL, Tan IL, Mukherjee J, et al. Sensitivity of glioblastomas to clinically available MEK inhibitors is defined by neurofibromin 1 deficiency. Cancer Res. 2012;72:3350–3359.
    1. Jessen WJ, Miller SJ, Jousma E, et al. MEK inhibition exhibits efficacy in human and mouse neurofibromatosis tumors. J Clin Invest. 2013;123:340–347.
    1. Dombi E, Baldwin A, Marcus LJ, et al. Activity of selumetinib in neurofibromatosis type 1-related plexiform neurofibromas. N Engl J Med. 2016;375:2550–2560.
    1. Van Raamsdonk CD, Bezrookove V, Green G, et al. Frequent somatic mutations of GNAQ in uveal melanoma and blue naevi. Nature. 2009;457:599–602.
    1. Van Raamsdonk CD, Griewank KG, Crosby MB, et al. Mutations in GNA11 in uveal melanoma. N Engl J Med. 2010;363:2191–2199.
    1. Chen X, Wu Q, Depeille P, et al. RasGRP3 mediates MAPK pathway activation in GNAQ mutant uveal melanoma. Cancer Cell. 2017;31:685–696.e6.
    1. O'Hayre M, Vazquez-Prado J, Kufareva I, et al. The emerging mutational landscape of G proteins and G-protein-coupled receptors in cancer. Nat Rev Cancer. 2013;13:412–424.
    1. Ambrosini G, Pratilas CA, Qin LX, et al. Identification of unique MEK-dependent genes in GNAQ mutant uveal melanoma involved in cell growth, tumor cell invasion, and MEK resistance. Clin Cancer Res. 2012;18:3552–3561.
    1. Carvajal RD, Sosman JA, Quevedo JF, et al. Effect of selumetinib vs chemotherapy on progression-free survival in uveal melanoma: A randomized clinical trial. JAMA. 2014;311:2397–2405.
    1. Livingstone E, Zaremba A, Horn S, et al. GNAQ and GNA11 mutant nonuveal melanoma: A subtype distinct from both cutaneous and uveal melanoma. Br J Dermatol. 2020;183:928–939.
    1. Khoury JD, Wang WL, Prieto VG, et al. Validation of immunohistochemical assays for integral biomarkers in the NCI-MATCH EAY131 clinical trial. Clin Cancer Res. 2018;24:521–531.
    1. Lih CJ, Harrington RD, Sims DJ, et al. Analytical validation of the next-generation sequencing assay for a nationwide signal-finding clinical trial: Molecular analysis for therapy choice clinical trial. J Mol Diagn. 2017;19:313–327.
    1. Conley BA, Flaherty KT. Molecular analysis for therapy choice (NCI-MATCH): A precision medicine signal-seeking trial in oncology. Personalized Med Oncol. 2016;5
    1. Flaherty KT, Gray R, Chen A, et al. The molecular analysis for therapy choice (NCI-MATCH) trial: Lessons for genomic trial design. J Natl Cancer Inst. 2020;112:1021–1029.
    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;45:228–247.
    1. Lin NU, Lee EQ, Aoyama H, et al. Response assessment criteria for brain metastases: Proposal from the RANO group. Lancet Oncol. 2015;16:e270–e278.
    1. Vogel CL, Cobleigh MA, Tripathy D, et al. Efficacy and safety of trastuzumab as a single agent in first-line treatment of HER2-overexpressing metastatic breast cancer. J Clin Oncol. 2002;20:719–726.
    1. Ramalingam SS, Vansteenkiste J, Planchard D, et al. Overall survival with osimertinib in untreated, EGFR-mutated advanced NSCLC. N Engl J Med. 2020;382:41–50.
    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;21:531–540.
    1. Seeber A, Chahine G, Nasr F, et al. Treatment according to a comprehensive molecular profiling can lead to a better outcome in heavily pretreated patients with metastatic cancer: Data of a pooled analysis. Cancer J. 2019;25:73–79.
    1. Lake D, Corrêa SAL, Muller J. Negative feedback regulation of the ERK1/2 MAPK pathway. Cell Mol Life Sci. 2016;73:4397–4413.
    1. Chen X, Wu Q, Tan L, et al. Combined PKC and MEK inhibition in uveal melanoma with GNAQ and GNA11 mutations. Oncogene. 2014;33:4724–4734.
    1. Carvajal RD, Schwartz GK, Tezel T, et al. Metastatic disease from uveal melanoma: Treatment options and future prospects. Br J Ophthalmol. 2017;101:38–44.
    1. Zhao Y, Adjei AA. The clinical development of MEK inhibitors. Nat Rev Clin Oncol. 2014;11:385–400.
    1. Zhu Y, Guignard F, Zhao D, et al. Early inactivation of p53 tumor suppressor gene cooperating with NF1 loss induces malignant astrocytoma. Cancer Cell. 2005;8:119–130.
    1. Herting CJ, Chen Z, Pitter KL, et al. Genetic driver mutations define the expression signature and microenvironmental composition of high-grade gliomas. Glia. 2017;65:1914–1926.
    1. Sintupisut N, Liu PL, Yeang CH. An integrative characterization of recurrent molecular aberrations in glioblastoma genomes. Nucleic Acids Res. 2013;41:8803–8821.
    1. The Cancer Genome Atlas Research Network Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature. 2008;455:1061–1068.
    1. Cerami E, Gao J, Dogrusoz U, et al. The cBio cancer genomics portal: An open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012;2:401–404.
    1. Ameratunga M, McArthur G, Gan H, et al. Prolonged disease control with MEK inhibitor in neurofibromatosis type I-associated glioblastoma. J Clin Pharm Ther. 2016;41:357–359.
    1. Khan I, Waqas M, Shamim MS. Prognostic significance of IDH 1 mutation in patients with glioblastoma multiforme. J Pak Med Assoc. 2017;67:816–817.
    1. Blumenschein GR, Jr, Smit EF, Planchard D, et al. A randomized phase II study of the MEK1/MEK2 inhibitor trametinib (GSK1120212) compared with docetaxel in KRAS-mutant advanced non-small-cell lung cancer (NSCLC) Ann Oncol. 2015;26:894–901.
    1. Redig AJ, Capelletti M, Dahlberg SE, et al. Clinical and molecular characteristics of NF1-mutant lung cancer. Clin Cancer Res. 2016;22:3148–3156.
    1. Gross AM, Wolters PL, Dombi E, et al. Selumetinib in children with inoperable plexiform neurofibromas. N Engl J Med. 2020;382:1430–1442.
    1. McCowage GB, Mueller S, Pratilas CA, et al. Trametinib in pediatric patients with neurofibromatosis type 1 (NF-1)–associated plexiform neurofibroma: A phase I/IIa study. J Clin Oncol. 2018;36:10504.
    1. Chapman PB, Hauschild A, Robert C, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med. 2011;364:2507–2516.
    1. Shoushtari AN, Kudchadkar RR, Panageas K, et al. A randomized phase 2 study of trametinib with or without GSK2141795 in patients with advanced uveal melanoma. J Clin Oncol. 2016;34:9511.

Source: PubMed

3
Abonnieren