Elsevier

The Lancet Oncology

Volume 23, Issue 1, January 2022, Pages 53-64
The Lancet Oncology

Articles
Dabrafenib plus trametinib in patients with BRAFV600E-mutant low-grade and high-grade glioma (ROAR): a multicentre, open-label, single-arm, phase 2, basket trial

https://doi.org/10.1016/S1470-2045(21)00578-7Get rights and content

Summary

Background

Effective treatments are needed to improve outcomes for high-grade glioma and low-grade glioma. The activity and safety of dabrafenib plus trametinib were evaluated in adult patients with recurrent or progressive BRAFV600E mutation-positive high-grade glioma and low-grade glioma.

Methods

This study is part of an ongoing open-label, single-arm, phase 2 Rare Oncology Agnostic Research (ROAR) basket trial at 27 community and academic cancer centres in 13 countries (Austria, Belgium, Canada, France, Germany, Italy, Japan, the Netherlands, Norway, South Korea, Spain, Sweden, and the USA). The study enrolled patients aged 18 years or older with an Eastern Cooperative Oncology Group performance status of 0, 1, or 2. Patients with BRAFV600E mutation-positive high-grade glioma and low-grade glioma received dabrafenib 150 mg twice daily plus trametinib 2 mg once daily orally until unacceptable toxicity, disease progression, or death. In the high-grade glioma cohort, patients were required to have measurable disease at baseline using the Response Assessment in Neuro-Oncology high-grade glioma response criteria and have been treated previously with radiotherapy and first-line chemotherapy or concurrent chemoradiotherapy. Patients with low-grade glioma were required to have measurable non-enhancing disease (except pilocytic astrocytoma) at baseline using the Response Assessment in Neuro-Oncology low-grade glioma criteria. The primary endpoint, in the evaluable intention-to-treat population, was investigator-assessed objective response rate (complete response plus partial response for high-grade glioma and complete response plus partial response plus minor response for low-grade glioma). This trial is ongoing, but is closed for enrolment, NCT02034110.

Findings

Between April 17, 2014, and July 25, 2018, 45 patients (31 with glioblastoma) were enrolled into the high-grade glioma cohort and 13 patients were enrolled into the low-grade glioma cohort. The results presented here are based on interim analysis 16 (data cutoff Sept 14, 2020). In the high-grade glioma cohort, median follow-up was 12·7 months (IQR 5·4–32·3) and 15 (33%; 95% CI 20–49) of 45 patients had an objective response by investigator assessment, including three complete responses and 12 partial responses. In the low-grade glioma cohort, median follow-up was 32·2 months (IQR 25·1–47·8). Nine (69%; 95% CI 39–91) of 13 patients had an objective response by investigator assessment, including one complete response, six partial responses, and two minor responses. Grade 3 or worse adverse events were reported in 31 (53%) patients, the most common being fatigue (five [9%]), decreased neutrophil count (five [9%]), headache (three [5%]), and neutropenia (three [5%]).

Interpretation

Dabrafenib plus trametinib showed clinically meaningful activity in patients with BRAFV600E mutation-positive recurrent or refractory high-grade glioma and low-grade glioma, with a safety profile consistent with that in other indications. BRAFV600E testing could potentially be adopted in clinical practice for patients with glioma.

Funding

Novartis.

Introduction

Gliomas are the most common primary tumours of the brain, diagnosed in approximately six per 100 000 people in the USA annually.1 The 2016 WHO criteria classify tumours into subtypes and grades based on histopathology and molecular and genetic markers, especially IDH (isocitrate dehydrogenase) mutational status and 1p/19q loss.2 The revised 2021 WHO criteria present changes that advance the role of molecular diagnostics in the classification of CNS tumours and different approaches to both nomenclature and grading of CNS tumours.3 Treatment options are dependent on histological classification and include maximal surgical resection where possible, with the potential addition of radiotherapy, with or without temozolomide-based or lomustine-based chemotherapy.4, 5 Treatment of recurrent glioblastoma with temozolomide is associated with poor response rates of 5–8%.6, 7, 8 The anti-VEGF monoclonal antibody bevacizumab is approved for recurrent glioblastoma; however, an improvement in overall survival has not been shown.9 Grade II and III IDH-mutated tumours, such as astrocytomas and oligodendrogliomas, have a better outcome and typically benefit more from the addition of chemotherapy to radiotherapy than glioblastomas.5 For IDH wild-type and circumscribed low-grade gliomas, such as gangliogliomas and pleomorphic xanthoastrocytomas, the effect of chemotherapy after incomplete resection or for inaccessible tumours is less clear.10, 11 Options are scarce for patients with low-grade glioma who progress after chemotherapy, and there is a risk of transformation to high-grade glioma and a much poorer prognosis over time.12 No substantial treatment advances have been made in recent years, with progress being hampered by the rarity of gliomas, difficulty in designing treatments capable of penetrating the blood–brain barrier, and molecular heterogeneity.4, 13

Research in context

Evidence before this study

We searched PubMed for reports of clinical trials published in English using the terms “BRAF-mutant”, “BRAF V600E”, “targeted therapy”, “immunotherapy”, “refractory clinical trial”, “glioma”, “low-grade glioma”, and “high-grade glioma”. We did not restrict our search by date. Our search results showed that no notable treatment advances have been made in the field of glioma in recent years, due to the rarity of these tumours, difficulty in designing treatments capable of penetrating the blood–brain barrier, and intertumoural and intratumoural heterogeneity. There has been progress in understanding the molecular pathogenesis and biology of these tumours, but it has not led to significantly improved outcomes for patients. Moreover, the prognostic significance of the BRAFV600E mutation in adult patients with glioma is unclear and could vary with histological subtype, with no specific treatment identified for this patient population. Dabrafenib has shown meaningful clinical activity in paediatric patients with BRAFV600E-mutant high-grade glioma and low-grade glioma. Vemurafenib has also shown durable antitumour activity in patients with BRAFV600-mutant gliomas. There is also emerging evidence that acquired resistance to BRAF inhibitor monotherapy develops in gliomas, which might be mitigated by dual inhibition. Dual blockade of the MAPK pathway using a combination of a BRAF inhibitor and a MEK inhibitor is now standard of care in BRAFV600-mutant melanoma, non-small-cell lung cancer, and anaplastic thyroid cancer and has shown activity in biliary tract cancer. Case reports of dabrafenib plus trametinib combination therapy in different types of gliomas were also found in our search.

Added value of this study

To our knowledge, this study is the first time that a combination of a BRAF inhibitor (dabrafenib) and a MEK inhibitor (trametinib) have shown notable activity in these difficult-to-treat gliomas, including glioblastomas, which have historically shown resistance to therapies. It is difficult to do clinical trials in this rare subset of patients who have scarce treatment options; however, the encouraging activity of the combination of dabrafenib and trametinib in this study suggests that targeting the MAPK pathway is a promising approach in these patients.

Implications of all the available evidence

The clinically meaningful and durable anti-tumour activity suggests that dabrafenib plus trametinib is a promising treatment option for patients with BRAFV600E-mutant recurrent high-grade glioma and low-grade glioma. Routine testing for BRAFV600E mutations should be considered in clinical practice for patients with glioma.

BRAF V600 mutations that constitutively activate the MAPK pathway have been identified in 5–15% of low-grade gliomas, including pleomorphic xanthoastrocytomas (60–80%), gangliogliomas (20–70%), pilocytic astrocytomas (10%), and less frequently in high-grade gliomas, including approximately 3% of glioblastomas.14 BRAFV600E mutations are typically mutually exclusive with IDH mutations.15 The prognostic significance of BRAFV600E mutations in adult patients with glioma is unclear and can vary with histological subtype.14, 15, 16, 17 No specific treatment has been identified for this subgroup of patients.

The BRAF inhibitor dabrafenib has shown meaningful clinical activity in paediatric patients with BRAF V600 mutation-positive low-grade glioma and high-grade glioma.18, 19 Dual blockade of the MAPK pathway using dabrafenib and the MEK inhibitor trametinib has shown activity in multiple tumour types and is a standard of care in BRAF V600-mutant melanoma, non-small-cell lung cancer, and anaplastic thyroid cancer.20, 21, 22, 23 Compared with BRAF inhibitor monotherapy, combined treatment showed superior outcomes with regard to progression-free survival and overall survival and reduced the incidence of skin-related toxicities in melanoma and non-small-cell lung cancer.20, 21, 22 Furthermore, there is emerging evidence that acquired resistance to BRAF inhibitor monotherapy develops in gliomas, as in other tumour types, which might be mitigated by dual inhibition.14, 24 Taken together, these data support the investigation of dabrafenib plus trametinib in adult patients with BRAFV600E mutation-positive glioma.

With no therapy showing a promising response, a high unmet medical need exists for low-grade glioma and high-grade glioma. We did a phase 2 basket study of dabrafenib plus trametinib in patients with BRAFV600E mutation-positive rare cancers (the Rare Oncology Agnostic Research [ROAR] trial). We report the activity and safety of this treatment combination in patients with relapsed or refractory BRAFV600E mutation-positive low-grade glioma and high-grade glioma from a preplanned interim analysis.

Section snippets

Study design and participants

We conducted a multicentre, open-label, single-arm, phase 2, basket study in nine cohorts of patients with BRAFV600E mutation-positive rare cancers (appendix p 17). The study enrolled patients aged 18 years or older, with an Eastern Cooperative Oncology Group performance status of 0, 1, or 2 and adequate organ function assessed by routine chemistry, haematology, and echocardiogram. Patients in the glioma cohorts were recruited from 27 community and academic cancer centres in 13 countries

Results

Between April 17, 2014, and July 25, 2018, 206 patients with BRAFV600E mutation-positive tumours were enrolled across eight of nine cohorts (no patients enrolled in the germ cell tumour cohort; appendix p 17). At a data cutoff of Sept 14, 2020 (interim analysis 16), 45 patients were recruited to the high-grade glioma cohort and 13 patients were recruited to the low-grade glioma cohort. Baseline characteristics of patients in the high-grade glioma and low-grade glioma cohorts are presented in

Discussion

In this phase 2 study, dabrafenib plus trametinib combination therapy exhibited clinically meaningful and durable activity in patients with recurrent or progressive BRAFV600E mutation-positive high-grade glioma and low-grade glioma. Patients in these cohorts had a sufficiently long follow-up and the unmet medical need in this patient population outweighs any concern of reporting interim analysis data.

In the context of the Bayesian design and analysis, for determining power of the study, the

Data sharing

Novartis is committed to sharing with qualified external researchers access to patient-level data and supporting clinical documents from eligible studies. These requests are reviewed and approved by an independent review panel on the basis of scientific merit. All data provided are anonymised to respect the privacy of patients who have participated in the trial, in line with applicable laws and regulations. This trial data availability is according to the criteria and process described on //www.clinicalstudydatarequest.com

Declaration of interests

PYW reports research support from Agios, AstraZeneca/MedImmune, Bayer, Celgene, Eli Lilly, Genentech/Roche, Kazia Therapeutics, MediciNova, Merck, Novartis, Nuvation Bio, Oncoceutics, Vascular Biogenics, and VBI Vaccines; and has an advisory or board member role in Agios, AstraZeneca, Bayer, Black Diamond, Boston Pharmaceuticals, ElevateBio, Imvax, Karyopharm Therapeutics, Merck, Mundipharma, Novartis, Novocure, Nuvation Bio, Prelude Therapeutics, QED Therapeutics, Sapience, Vascular Biogenics,

References (45)

  • MJ van den Bent et al.

    Diffuse infiltrating oligodendroglioma and astrocytoma

    J Clin Oncol

    (2017)
  • WK Yung et al.

    A phase II study of temozolomide vs procarbazine in patients with glioblastoma multiforme at first relapse

    Br J Cancer

    (2000)
  • JR Perry et al.

    Phase II trial of continuous dose-intense temozolomide in recurrent malignant glioma: RESCUE study

    J Clin Oncol

    (2010)
  • W Wick et al.

    Lomustine and bevacizumab in progressive glioblastoma

    N Engl J Med

    (2017)
  • S Yust-Katz et al.

    Clinical and prognostic features of adult patients with gangliogliomas

    Neuro-oncol

    (2014)
  • N Shaikh et al.

    Pleomorphic xanthoastrocytoma: a brief review

    CNS Oncol

    (2019)
  • EB Claus et al.

    Survival and low-grade glioma: the emergence of genetic information

    Neurosurg Focus

    (2015)
  • AP Patel et al.

    Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma

    Science

    (2014)
  • KC Schreck et al.

    BRAF mutations and the utility of RAF and MEK inhibitors in primary brain tumors

    Cancers (Basel)

    (2019)
  • AK Chan et al.

    BRAF mutation marks out specific subgroups of glioma

    Glioma

    (2018)
  • MJ Lim-Fat et al.

    Clinical, radiological and genomic features and targeted therapy in BRAF V600E mutant adult glioblastoma

    J Neurooncol

    (2021)
  • A Aibaidula et al.

    Adult IDH wild-type lower-grade gliomas should be further stratified

    Neuro-oncol

    (2017)
  • Cited by (154)

    View all citing articles on Scopus
    View full text