Alterations in the MAPK pathway: promising biomarkers for MEK1/2 inhibition
Editorial Commentary

Alterations in the MAPK pathway: promising biomarkers for MEK1/2 inhibition

Thaïs Baert1,2, Tine Ottenbourgs1,2, Toon Van Gorp1,2, Els Van Nieuwenhuysen1,2

1Gynaecological Oncology Laboratory, KU Leuven, Leuven Cancer Institute, Leuven, Belgium; 2Department of Gynaecology and Obstetrics, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium

Correspondence to: Thaïs Baert, MD, PhD. Gynaecological Oncology Laboratory, KU Leuven, Leuven Cancer Institute, Herestraat 49, 3000 Leuven, Belgium; Department of Gynaecology and Obstetrics, University Hospitals Leuven, Leuven Cancer Institute, Herestraat 49, 3000 Leuven, Belgium. Email: thais.baert@uzleuven.be.

Comment on: Grisham RN, Vergote I, Banerjee S, et al. Molecular Results and Potential Biomarkers Identified from the Phase 3 MILO/ENGOTov11 Study of Binimetinib versus Physician Choice of Chemotherapy in Recurrent Low-Grade Serous Ovarian Cancer. Clin Cancer Res 2023;29:4068-75.


Keywords: Mitogen-activated protein kinase (MAPK); MEK1/2 inhibition; binimetinib


Received: 05 December 2023; Accepted: 26 February 2024; Published online: 20 March 2024.

doi: 10.21037/gpm-23-50


Low-grade serous ovarian carcinoma (LGSOC) is a distinct subtype of ovarian cancer that presents unique challenges in terms of treatment due to intrinsic chemoresistance. LGSOC, in comparison to high-grade serous ovarian carcinoma, has a high prevalence of mutations in KRAS (20–40%), NRAS (26%) and BRAF (5–10%) (1,2). Patients with activating mutations in canonical mitogen-activated protein kinase (MAPK) pathway components are reported to respond more frequently to targeted agents, such as MEK inhibitors, compared to patients without a mutation in the MAPK pathway (3,4). MEK inhibitors are a class of targeted therapies designed to block the activity of MEK1 and MEK2. These are proteins that play a crucial role in the MAPK signaling cascade and can contribute to uncontrolled cell growth, resistance to apoptosis and increased angiogenesis (5).

The post-hoc tumor tissue analysis from the phase 3 ENGOT-ov11/MILO study evaluated the efficacy of binimetinib, a potent MEK1/2 inhibitor, in treating recurrent LGSOC (6). The study enrolled 341 patients between June 2013 and April 2016, who were randomized 2:1 to receive either binimetinib or physician’s choice of chemotherapy (PCC). The most commonly altered gene was KRAS (33%), which is in accordance with literature (7-10).

The study found that patients with a KRAS mutation (KRASmut) treated with binimetinib were 3.4 times more likely to respond [complete remission (CR) or partial remission (PR)] compared to patients without a KRAS mutation (KRASwt) [95% confidence interval (CI): 1.57–7.67]. Among patients treated with binimetinib, 44% of KRASmut patients achieved a CR/PR compared to 19% of KRASwt patients.

The study also found that patients who harbor MAPK pathway alterations (NRAS, BRAF, NF1, RAF1 mutations) and are treated with binimetinib have longer progression-free survival (PFS) compared to those without MAPK pathway alterations [hazard ratio (HR) 0.50; 95% CI: 0.31–0.79]. A similar trend towards improved PFS was observed among patients harboring a MAPK pathway alteration treated with PCC (HR 0.82; 95% CI: 0.43–1.59). This is in line with other evidence suggesting that women with low-grade serous ovarian tumors containing a KRAS or BRAF mutation experience a more favorable outcome compared to those with tumors that lack these activating MAPK pathway mutations (11).

In vitro and in vivo data indicate that patients harboring the KRAS G12V mutation may be more sensitive to MEK inhibition compared to other KRAS variants. Despite this, the study found no difference in PFS in patients with KRAS G12V mutation (n=19) versus patients with other KRAS variants (n=26).

In the GOG-281/LOGS phase 2/3 trial, patients with recurrent LGSOC were treated with trametinib, a MEK1/2 inhibitor, versus PCC. PFS and overall response rate (ORR) were both markedly higher in patients with a KRAS, BRAF or NRAS mutation compared to patients with wild-type KRAS, BRAF and NRAS (3). This confirms the findings of the post-hoc tumor analysis of the MILO/ENGOT-ov11 trial. Additionally, the GOG-281/LOGS trial found that this mutation profile might be predictive for ORR, but not for PFS.

Similar impact of a KRAS mutation on ORR were described in the ENGOT-ov60/GOG-3052/RAMP201 study including patients with recurrent LGSOC (12). The initial results of this study showed that avutometinib, a dual RAF/MEK inhibitor, in monotherapy or in combination with defactinib, a FAK inhibitor, had a higher ORR in KRASmut patients (monotherapy 10% vs. combination 60%) compared to KRASwt patients (monotherapy 6% vs. combination 29%). Based on these results, the study will continue as a randomized phase 3 clinical trial of the combination of avutometinib and defactinib versus PCC.

In conclusion, this post-hoc analysis identifies alterations in the MAPK pathway as possible biomarkers to select patients for MEK1/2 inhibitor treatment in the future. MEK inhibitors could be an effective treatment option for LGSOC patients with alterations in the MAPK pathway and not limited to KRAS. Biomarker research is crucial in identifying the right patient population to select for targeted agents, making this type of translational research important in LGSOC.


Acknowledgments

Funding: None.


Footnote

Provenance and Peer Review: This article was commissioned by the editorial office, Gynecology and Pelvic Medicine. The article has undergone external peer review.

Peer Review File: Available at https://gpm.amegroups.com/article/view/10.21037/gpm-23-50/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://gpm.amegroups.com/article/view/10.21037/gpm-23-50/coif). T.B. received a grant from Roche, honoraria for lectures or consultancy from AstraZeneca, BioNTech, Eisai Co, GSK, ImmunoGen, Incyte, MSD/Merck, OncXerna, Seagen, Tubulis and Zentalis support for attending meetings from GSK, MSD and Astrazeneca. All payments were made to the author’s institution. T.V.G. received research funding from Amgen, AstraZeneca and Roche, honoraria for lectures from Lilly, Novartis and Astrazeneca, support for attending meetings from AstraZeneca, GSK, ImmunoGen, MSD/Merck and PharmaMar. All payments were made to the author’s institution. The other authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Thomson JP, Hollis RL, van Baal J, et al. Whole exome sequencing of low grade serous ovarian carcinoma identifies genomic events associated with clinical outcome. Gynecol Oncol 2023;174:157-66. [Crossref] [PubMed]
  2. Hunter SM, Anglesio MS, Ryland GL, et al. Molecular profiling of low grade serous ovarian tumours identifies novel candidate driver genes. Oncotarget 2015;6:37663-77. [Crossref] [PubMed]
  3. Gershenson DM, Miller A, Brady WE, et al. Trametinib versus standard of care in patients with recurrent low-grade serous ovarian cancer (GOG 281/LOGS): an international, randomised, open-label, multicentre, phase 2/3 trial. Lancet 2022;399:541-53. [Crossref] [PubMed]
  4. Monk BJ, Grisham RN, Banerjee S, et al. MILO/ENGOT-ov11: Binimetinib Versus Physician's Choice Chemotherapy in Recurrent or Persistent Low-Grade Serous Carcinomas of the Ovary, Fallopian Tube, or Primary Peritoneum. J Clin Oncol 2020;38:3753-62. [Crossref] [PubMed]
  5. Caunt CJ, Sale MJ, Smith PD, et al. MEK1 and MEK2 inhibitors and cancer therapy: the long and winding road. Nat Rev Cancer 2015;15:577-92. [Crossref] [PubMed]
  6. Grisham RN, Vergote I, Banerjee S, et al. Molecular Results and Potential Biomarkers Identified from the Phase 3 MILO/ENGOT-ov11 Study of Binimetinib versus Physician Choice of Chemotherapy in Recurrent Low-Grade Serous Ovarian Cancer. Clin Cancer Res 2023;29:4068-75. [Crossref] [PubMed]
  7. Wong KK, Tsang YT, Deavers MT, et al. BRAF mutation is rare in advanced-stage low-grade ovarian serous carcinomas. Am J Pathol 2010;177:1611-7. [Crossref] [PubMed]
  8. Grisham RN, Iyer G, Garg K, et al. BRAF mutation is associated with early stage disease and improved outcome in patients with low-grade serous ovarian cancer. Cancer 2013;119:548-54. [Crossref] [PubMed]
  9. Hunter SM, Anglesio MS, Ryland GL, et al. Molecular profiling of low grade serous ovarian tumours identifies novel candidate driver genes. Oncotarget 2015;6:37663-77. [Crossref] [PubMed]
  10. Van Nieuwenhuysen E, Busschaert P, Laenen A, et al. Loss of 1p36.33 Frequent in Low-Grade Serous Ovarian Cancer. Neoplasia 2019;21:582-90. [Crossref] [PubMed]
  11. Gershenson DM, Sun CC, Wong KK. Impact of mutational status on survival in low-grade serous carcinoma of the ovary or peritoneum. Br J Cancer 2015;113:1254-8. [Crossref] [PubMed]
  12. Banerjee SN, Ring KL, Van Nieuwenhuysen E, et al. Initial efficacy and safety results from ENGOT-ov60/GOG-3052/RAMP 201: A phase 2 study of avutometinib (VS-6766) ± defactinib in recurrent low-grade serous ovarian cancer (LGSOC). J Clin Oncol 2023;41:5515. [Crossref]
doi: 10.21037/gpm-23-50
Cite this article as: Baert T, Ottenbourgs T, Van Gorp T, Van Nieuwenhuysen E. Alterations in the MAPK pathway: promising biomarkers for MEK1/2 inhibition. Gynecol Pelvic Med 2024;7:7.

Download Citation