Molecular mechanism and clinical outcome of non-small cell lung cancer patients with high BRAF expression
Editorial

Molecular mechanism and clinical outcome of non-small cell lung cancer patients with high BRAF expression

Yuji Uehara1,2,3^

1Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan; 2Department of Precision Cancer Medicine, Center for Innovative Cancer Treatment, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; 3Department of Thoracic Oncology, National Cancer Center Hospital East, Tokyo, Japan

^ORCID: 0000-0001-8047-8730.

Correspondence to: Yuji Uehara, MD. Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, 3-18-22 Honkomagoame, Bunkyo, Tokyo, Japan. Email: yuuji.csa@gmail.com.

Comment on: Dora D, Vörös I, Varga ZV, et al. BRAF RNA is prognostic and widely expressed in lung adenocarcinoma. Transl Lung Cancer Res 2023;12:27-41.


Keywords: Lung adenocarcinoma (LUAD); non-small cell lung cancer (NSCLC); prognosis; amplification; overexpression


Submitted Feb 12, 2023. Accepted for publication Mar 16, 2023. Published online Mar 24, 2023.

doi: 10.21037/tlcr-23-103


Non-small cell lung cancer (NSCLC) is a genetically heterogeneous disease, with several distinct oncogenic alterations having been characterized. One such subpopulation of lung cancers is BRAF-mutated lung cancer, which constitutes approximately 4% of advanced NSCLC (1,2). The most prevalent BRAF mutations, BRAF V600E mutations, are present in approximately 2% of NSCLC (2,3). These mutations engender activation of the RAF-MEK-ERK (MAPK) pathway, leading to cell proliferation and growth. NSCLC with BRAF V600E mutations and selected non-V600 mutations respond to the treatment of dabrafenib (BRAF inhibitor) and trametinib (MEK inhibitor), while immune checkpoint inhibitors (ICI)-based regimens or chemotherapy, including platinum-based doublet chemotherapy, demonstrate modest efficacy against these BRAF alterations (4-10). Despite the lack of comprehensive studies on the clinical implications of BRAF amplification in tumor development and progression, de novo BRAF amplification has been reported to be 0.1% of NSCLC in American Association for Cancer Research (AACR) Project Genomics, Evidence, Neoplasia, Information, Exchange (GENIE) cohort, and it has been linked to resistance to RTK/RAS/RAF/MEK inhibitors (11-16).

There are three functional classes of BRAF mutations (17-19). Class I BRAF mutations, which operate as monomers independently of RAS activation, result in elevated ERK activation and subsequent negative feedback inhibition of upstream signaling. This class includes BRAF V600E mutations. Class II BRAF mutations, which are RAS-independent and form mutant-mutant BRAF dimers, elicit robust ERK activation and negative feedback inhibition of upstream signaling. Class III BRAF mutations exhibit augmented binding to RAS and CRAF, signaling as mutant BRAF-wild-type (WT) CRAF dimers. They amplify downstream signaling from RAS and thus necessitate upstream activation to enhance ERK signaling, either through genomic alterations (such as RAS mutations or NF1 loss) or through receptor tyrosine kinase (RTK) signaling.

In the accompanying article, Dora et al. demonstrate that a substantial proportion of patients with lung adenocarcinoma (LUAD), namely 73% (47/64), exhibited high levels of WT BRAF RNA across all tumor stages (20). This high expression was associated with never-smoker status, tumor necrosis, low programmed death ligand 1 (PD-L1) expression in immune cells, and poor prognosis to chemotherapy (platinum-based doublet). The gene’s protein product was detected in 80% (8/10) samples from the Human Protein Atlas database using immunohistochemistry (IHC), with moderate or strong staining intensity. The negative prognostic role of high BRAF RNA expression was also confirmed in The Cancer Genome Atlas (TCGA) cohort of LUAD patients (n=318). This study aligns with previous TCGA analyses, which showed that metastatic lesions of LUAD exhibited significantly higher BRAF expression compared to corresponding adjacent normal tissues (14). Furthermore, in the context of papillary thyroid carcinoma, previous TCGA analysis indicated that higher BRAF RNA expression was associated with more aggressive tumor features, including higher tumor stage and the presence of extrathyroidal extension, regardless of BRAF mutational status (21).

The authors noted that the prevalence of LUAD patients with high BRAF expression (73%, 47/64) could not be attributed to BRAF mutations (20), given that only approximately 4% of NSCLC harbor BRAF mutations (1). BRAF amplification could be ruled out as a potential explanation because it is a rare population, 0.1% of NSCLC (12,14,16). Oncogenic alterations in RTK or RAS proteins (such as KRAS, NRAS, and HRAS) are known to activate the MAPK signaling pathway, and we hypothesized these alterations might drive elevated BRAF RNA expression as an aberrant downstream pathway (Figure 1). The proportion of the high BRAF expression group (73%) in this cohort was comparable to that of patients with oncogenic RTK or RAS alterations (75%) in LUAD from MSK-IMPACT Clinical Sequencing Cohort (KRAS, 30%; EGFR, 30%; HER2, 4%; ALK, 4%; MET, 3%; RET, 2%; ROS1, 2%; total, 75%, data from cBio cancer genomics portal) (22). Additionally, never smokers demonstrated significantly higher BRAF expression than current smokers and ex-smokers, suggesting the association between higher BRAF expression and frequent oncogenic RTK or RAS alterations in never smokers. Despite the lack of genomic analysis in this study, the poor prognosis associated with the high BRAF expression group in this cohort and the TCGA cohort may be due to the absence of potential targeted therapies for patients with various driver alterations in RTK or RAS. Most patients in this cohort received platinum-based doublet therapy, thus missing the opportunity for targeted therapies such as osimertinib for EGFR-mutated lung cancer and alectinib for ALK-rearranged lung cancer. If our hypothesis is correct, targeting the overexpressed BRAF pathway would involve inhibiting the upstream activation pathway with RAS inhibitors or RTK inhibitors. A more extensive examination of BRAF RNA expression, RTK-RAS-RAF-MEK-ERK pathway alterations, and clinical prognostic implications is necessary to discern the underlying mechanism behind the prevalent proportion of patients with high BRAF expression and their associated inferior prognosis.

Figure 1 Hypothesis of association between high BRAF expression and RTK/RAS driver alterations in lung adenocarcinoma. LUAD, lung adenocarcinoma.

The relationship between genetic heterogeneity and prognostication has been analyzed in the field of lung cancer; however, the relationship between genotypic and phenotypic heterogeneity and prognostication remains inadequately understood. Most of the consequences of genetic and cytogenetic modifications will influence gene expression through aberrant transcription, epigenetic regulation, and cellular signaling. The vehicle by which driver mutations give rise to carcinogenesis is transcription, facilitated through a labyrinthine cellular signaling circuitry that links genomic alterations to the clinical phenotype of cancers and their prognosis (23). A more comprehensive understanding of driver mutations and differential RNA expression with clinical outcomes beyond BRAF expression is imperative.


Acknowledgments

Funding: None.


Footnote

Provenance and Peer Review: This article was commissioned by the editorial office, Translational Lung Cancer Research. The article did not undergo external peer review.

Conflicts of Interest: The author has completed the ICMJE uniform disclosure form (available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-23-103/coif). The author has no conflicts of interest to declare.

Ethical Statement: The author is 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. Skoulidis F, Heymach JV. Co-occurring genomic alterations in non-small-cell lung cancer biology and therapy. Nat Rev Cancer 2019;19:495-509. [Crossref] [PubMed]
  2. Tissot C, Couraud S, Tanguy R, et al. Clinical characteristics and outcome of patients with lung cancer harboring BRAF mutations. Lung Cancer 2016;91:23-8. [Crossref] [PubMed]
  3. Villaruz LC, Socinski MA, Abberbock S, et al. Clinicopathologic features and outcomes of patients with lung adenocarcinomas harboring BRAF mutations in the Lung Cancer Mutation Consortium. Cancer 2015;121:448-56. [Crossref] [PubMed]
  4. Gibson AJW, Pabani A, Dean ML, et al. Real-World Treatment Patterns and Effectiveness of Targeted and Immune Checkpoint Inhibitor-Based Systemic Therapy in BRAF Mutation-Positive NSCLC. JTO Clin Res Rep 2023;4:100460. [Crossref] [PubMed]
  5. Negrao MV, Raymond VM, Lanman RB, et al. Molecular Landscape of BRAF-Mutant NSCLC Reveals an Association Between Clonality and Driver Mutations and Identifies Targetable Non-V600 Driver Mutations. J Thorac Oncol 2020;15:1611-23. [Crossref] [PubMed]
  6. Uehara Y, Watanabe K, Hakozaki T, et al. Efficacy of first-line immune checkpoint inhibitors in patients with advanced NSCLC with KRAS, MET, FGFR, RET, BRAF, and HER2 alterations. Thorac Cancer 2022;13:1703-11. [Crossref] [PubMed]
  7. Li H, Zhang Y, Xu Y, et al. Tumor immune microenvironment and immunotherapy efficacy in BRAF mutation non-small-cell lung cancer. Cell Death Dis 2022;13:1064. [Crossref] [PubMed]
  8. Mazieres J, Drilon A, Lusque A, et al. Immune checkpoint inhibitors for patients with advanced lung cancer and oncogenic driver alterations: results from the IMMUNOTARGET registry. Ann Oncol 2019;30:1321-8. [Crossref] [PubMed]
  9. Couraud S, Barlesi F, Fontaine-Deraluelle C, et al. Clinical outcomes of non-small-cell lung cancer patients with BRAF mutations: results from the French Cooperative Thoracic Intergroup biomarkers France study. Eur J Cancer 2019;116:86-97. [Crossref] [PubMed]
  10. Planchard D, Smit EF, Groen HJM, et al. Dabrafenib plus trametinib in patients with previously untreated BRAF(V600E)-mutant metastatic non-small-cell lung cancer: an open-label, phase 2 trial. Lancet Oncol 2017;18:1307-16. [Crossref] [PubMed]
  11. Corcoran RB, Dias-Santagata D, Bergethon K, et al. BRAF gene amplification can promote acquired resistance to MEK inhibitors in cancer cells harboring the BRAF V600E mutation. Sci Signal 2010;3:ra84. [Crossref] [PubMed]
  12. Tan I, Stinchcombe TE, Ready NE, et al. Therapeutic outcomes in non-small cell lung cancer with BRAF mutations: a single institution, retrospective cohort study. Transl Lung Cancer Res 2019;8:258-67. [Crossref] [PubMed]
  13. Zhang L, Zheng L, Yang Q, et al. The Evolution of BRAF Activation in Non-Small-Cell Lung Cancer. Front Oncol 2022;12:882940. [Crossref] [PubMed]
  14. Yi Q, Peng J, Xu Z, et al. Spectrum of BRAF Aberrations and Its Potential Clinical Implications: Insights From Integrative Pan-Cancer Analysis. Front Bioeng Biotechnol 2022;10:806851. [Crossref] [PubMed]
  15. Xu T, Wang X, Wang Z, et al. Molecular mechanisms underlying the resistance of BRAF V600E-mutant metastatic colorectal cancer to EGFR/BRAF inhibitors. Ther Adv Med Oncol 2022;14:17588359221105022. [Crossref] [PubMed]
  16. AACR Project GENIE: Powering Precision Medicine through an International Consortium. Cancer Discov 2017;7:818-31. [Crossref] [PubMed]
  17. Yaeger R, Corcoran RB. Targeting Alterations in the RAF-MEK Pathway. Cancer Discov 2019;9:329-41. [Crossref] [PubMed]
  18. Yao Z, Yaeger R, Rodrik-Outmezguine VS, et al. Tumours with class 3 BRAF mutants are sensitive to the inhibition of activated RAS. Nature 2017;548:234-8. [Crossref] [PubMed]
  19. Yao Z, Torres NM, Tao A, et al. BRAF Mutants Evade ERK-Dependent Feedback by Different Mechanisms that Determine Their Sensitivity to Pharmacologic Inhibition. Cancer Cell 2015;28:370-83. [Crossref] [PubMed]
  20. Dora D, Vörös I, Varga ZV, et al. BRAF RNA is prognostic and widely expressed in lung adenocarcinoma. Transl Lung Cancer Res 2023;12:27-41. [Crossref] [PubMed]
  21. Chai YJ, Yi JW, Jee HG, et al. Significance of the BRAF mRNA Expression Level in Papillary Thyroid Carcinoma: An Analysis of The Cancer Genome Atlas Data. PLoS One 2016;11:e0159235. [Crossref] [PubMed]
  22. Zehir A, Benayed R, Shah RH, et al. Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat Med 2017;23:703-13. [Crossref] [PubMed]
  23. Gerstung M, Pellagatti A, Malcovati L, et al. Combining gene mutation with gene expression data improves outcome prediction in myelodysplastic syndromes. Nat Commun 2015;6:5901. [Crossref] [PubMed]
Cite this article as: Uehara Y. Molecular mechanism and clinical outcome of non-small cell lung cancer patients with high BRAF expression. Transl Lung Cancer Res 2023;12(3):401-404. doi: 10.21037/tlcr-23-103

Download Citation