• Users Online: 1315
  • Print this page
  • Email this page


 
 
Table of Contents
REVIEW ARTICLE
Year : 2019  |  Volume : 2  |  Issue : 2  |  Page : 37-42

The mala fides of BRAF in oncogenesis


1 Departments of Laboratory; Transfusion Services; Research; Rajiv Gandhi Cancer Institute and Research Centre, Delhi, India
2 Department of Research, Rajiv Gandhi Cancer Institute and Research Centre, Delhi, India
3 Department of Laboratory, Rajiv Gandhi Cancer Institute and Research Centre, Delhi, India

Date of Web Publication30-Dec-2019

Correspondence Address:
Dr. Anurag Mehta
Director Laboratory, Molecular Diagnostic Services & Research Rajiv Gandhi Cancer Institute & Research Centre, Sector-V, Rohini, Delhi- 110085.
India
Login to access the Email id

Source of Support: None, Conflict of Interest: None


DOI: 10.4103/jco.jco_25_19

Rights and Permissions
  Abstract 

BRAF is a proto-oncogene that encodes a serine threonine kinase belonging to intracellular RAS-RAF-MEK& ERK signaling cascade downstream of surface Receptor Tyrosine Kinase. The gain of function mutations in BRAF gene have been shown to have a powerful oncogenic effect especially a transversion at nucleotide 1,799 from thymidine to adenine (p.V600E), accounting for greater than 80% of the observed mutations in BRAF. This one mutation has been identified as an oncogenic driver in a diverse set of solid and hematologic cancers. Fortunately, introduction of BRAF & MEK inhibitors has modestly transformed the treatment outcomes in patients with BRAF mutations, especially those with non-small cell lung carcinoma, melanoma and thyroid cancers. Besides, the predictive nature of BRAF mutation, the mutational analysis also helps predict prognosis and secure diagnosis of a varied group of malignancies. This review comprehensively addresses to the various mala fides of BRAF in oncogenesis, as well as elucidates the nucleotide variants which have been reported in literature, the diagnostic and prognostic utility, and the testing methods which are available to test the same.

Keywords: BRAF, BRAF V600E, RAS–RAF–MEK–ERK, significantly mutated genes


How to cite this article:
Mehta A, Gupta G, Nathany S. The mala fides of BRAF in oncogenesis. J Curr Oncol 2019;2:37-42

How to cite this URL:
Mehta A, Gupta G, Nathany S. The mala fides of BRAF in oncogenesis. J Curr Oncol [serial online] 2019 [cited 2024 Mar 28];2:37-42. Available from: http://www.https://journalofcurrentoncology.org//text.asp?2019/2/2/37/274302




  Introduction Top


Gain-of-function mutations or rearrangements in proto-oncogenes confer survival advantage and endow transformative attributes on the host cell pushing it on the path of carcinogenesis. Of the 25,000 genes in human genome, approximately 400 genes have been identified as “Significantly Mutated Genes.”[1],[2],[3] Among these, RAS, PIK3CA, and BRAF have exceptional oncogenic potential, accounting for 15%–30%, 13%, and 7% of driver mutations in all cancer types, respectively.[4],[5],[6]

BRAF mutation and fusion rearrangements have been recognized as primary driver mutation in diverse solid and hematologic malignancies.[7] Their mechanistic intricacies, potential in diagnosis, therapy selection, and prognosis are being elucidated regularly and integrated into clinical practice. This review discusses these aspects of BRAF genetic alterations.


  BRAF Gene and Protein Top


The BRAF gene is located on the long arm of chromosome 7 (7q34) and has 18 exons.[8] It encodes a serine–threonine kinase named BRAF that belongs to the RAF family, which also includes ARAF and CRAF (RAF1).

BRAF protein is an intermediate signal transducer of RAS–RAF–MEK–ERK signaling pathway, which regulates:

  • cell division;


  • cellular differentiation;


  • cellular migration;


  • apoptosis; and


  • secretion.


  • Binding of “growth factor, hormone, or cytokine” to their receptor tyrosine kinase initiates conversion of membrane-bound inactive GDP-RAS to its active form GTP-RAS. The GTP RAS binds the BRAF protein. The high density KRAS molecules tethered to the inside of cell membrane bring BRAF protein molecules close enough to trigger BRAF protein dimerization or heterodimerization with CRAF. This activates BRAF especially the heterodimer and initiates a cascade that relays signal downstream activating ERK as the terminal effector [Figure 1].
    Figure 1: The MEK–ERK pathway. Receptor tyrosine kinase (RTK) binds with its ligand growth factor (GF) that leads to binding of docker protein GRB2 to its intracytoplasmic portion, which in turn binds to Son of Sevenless (SOS). SOS transfers GTP to RAS and activates it. The GTP RAS activates BRAF by dimerization. Resultant downstream signaling activates MEK and ERK proteins. The ERK protein internalizes to nucleus and initiates transcription of several proteins involved in cell division, differentiation, inhibition of apoptosis, and secretion. ER, FOS, GLUT1, CCND, cMYC, B3 integrin, and VEGF are positive regulators. DUSP and SPRY are autoregulatory proteins of the MEK–ERK pathway. BAD and BIM are inactivated. (Copyright Dr A. Mehta)

    Click here to view


    Activated ERK internalizes to nucleus and regulates cellular functions as listed below:

  • Promotes cellular proliferation through expression of cyclin D1, cyclin D2, cyclin D3, ER, FOS, and GLUT1 genes


  • Negatively regulates p53 by expression of MDM2


  • Inhibits apoptosis by inactivation of proapoptotic “bcl2-associated agonist of cell death”’ (BAD) and BIM


  • Induces insensitivity to growth inhibition signals through expression of MYC protein


  • Promotes angiogenesis by promoting expression of VEGF


  • Controls cellular migration through β3 integrin expression.


  • In normal physiological state, the process is switched off by negative feedback from activated ERK that undoes the BRAF heterodimerization by phosphorylation at S365 [Figure 2] and by conversion of GTP-RAS to GDP-RAS by RAS protein intrinsic GTPase activity further potentiated by cytosolic GTPase-activating protein (GAP). DUSP and SPRY are two autocrinal proteins blocking the relay of signaling from MEK to ERK. Once the signaling is switched off, the ERK protein returns to cytoplasm abrogating the transcription of aforementioned genes.
    Figure 2: Structure of the BRAF protein. The conserved regions superimposed on their exonic locations are shown as CR1, CR2, and CR3. S365 in CR2 is the negative regulatory phosphorylation site. The N terminal of kinase domain (KD) comprising SSDD amino acid sequence provides negative charge and phosphorylation site at S446. The S446 phosphorylation is constitutive and necessary for function of kinase domain. T599 and S602 in the activation segment TVKS are the phosphorylation site activated by binding of RAS GTP and are necessary for activation of downstream MEK. RBD = GTP-RAS binding domain, CRD = cysteine-rich domain. (Copyright Dr A. Mehta)

    Click here to view


    The BRAF protein is 766-amino acid (AA) long with 84kDa molecular weight. It has three conserved regions labeled as CR1, CR2, and CR3[9] [Figure 2]. The functionally significant domains in these conserved regions are as follows:

  • CR1 region has GTP-RAS binding domain (RBD) and autoinhibits BRAF kinase domain.


  • CR2 contains a negative regulatory phosphorylation site at S365.


  • CR3 contains the kinase domain with N terminal of SSDD AAs that provides negative charge to this site for its function. T599 and S602 are the activation-induced phosphorylation sites and are crucial for downstream relay of signal.



  •   Cancer and BRAF Gene Alterations Top


    BRAF-activating somatic mutations account for nearly 7% of all cancers.[5] Such mutations bestowed with manifold higher kinase activity amplify the downstream signaling causing unregulated cellular proliferation, genomic instability due to loss of p53 function, angiogenesis and migration along with suppression of apoptosis, and the classical makings of a transformed cell. Approximately 45 disease-producing somatic missense mutations have been identified in different types of cancer [Table 1].
    Table 1: Clinically significant variants of BRAF gene and their frequency as described in My Cancer Genome database[6]

    Click here to view


    A great majority (~90%) of these mutations reside at V600 and K601 of the TVKS activation segment.[10] T1799A that causes the AA substitution BRAFV600E accounts for 80% of driver mutations ascribed to BRAF gene.[11] The oncogenic potential of this mutation stems from its 500- to 700-fold increase of kinase activity.[12] This mutated protein is active in monomeric form and does not require activation by GTP–RAS. Another roughly 10% deleterious mutations reside in exon 15 either at AA K601 or in vicinity of V600 at AA sequence D594, G596, or L597. The other 10% exist in the glycine of the G-loop in the exon 11 at G464, G466, and G469.

    In addition to missense deleterious mutations, approximately 80 fusion rearrangements have also been identified in several cancers, predominantly in melanoma and non-small-cell lung carcinoma (NSCLC). Pilocytic astrocytoma, colorectal adenocarcinoma, and papillary thyroid carcinoma are other cancers with BRAF fusion rearrangement. The fusion rearrangements derive their oncogenic potential through loss of autoregulatory N terminal portion of the BRAF protein caused by fusion rearrangement.[13],[14] A large fraction of these are caused by inversion 7 causing rearrangement with several genes as shown in [Table 2].
    Table 2: Some of the examples of fusion rearrangements involving BRAF gene[13]

    Click here to view



      Diagnostic Methods Top


    A variety of molecular assays are used for detection of BRAF mutations. Sanger sequencing, Next-Generation Sequencing (NGS), and pyrosequencing are best suited to identify all the single nucleotide variants. Because of the dominant nature of BRAFV600E and its druggability, the current emphasis is on identifying BRAFV600E along with a few other mutations at codon 600 and 601. For this limited objective, strategies based on real-time polymerase chain reaction (PCR) using allele-specific PCR or amplification refractory mutation–specific system with approximate limit of detection of 5% are commonly used. A BRAFV600E mutation–specific antibody (VE1) for immunohistochemistry is also available and has the US Food and Drug Administration (FDA) approval for use in colorectal cancer for determination of MLH1-deficient tumors that should undergo germ line testing for Lynch syndrome. [Table 3] lists the various FDA-approved tests and the variant called by each of these assays.
    Table 3: FDA-approved testing methods for BRAFV600E and codon 600 and 601 testing[15],[16],[17]

    Click here to view



      Value of Identifying BRAF Mutations Top


    As stated before, BRAFV600E is by far the most common gain-of-function mutation in BRAF gene, and its detection is used for diagnostic, prognostic, and predictive purposes. Some important indications are listed as follows under each category implication:

    Diagnostic utility of BRAFV600E detection

  • The correct diagnosis of hairy cell leukemia (HCL) is essential because it can be effectively treated by purine analogues, whereas its mimics such as splenic marginal zone lymphoma or splenic lymphoma/leukemia unclassifiable including HCL variant (HCL-v) do not respond to these drugs. All cases of HCL carry BRAFV600E mutation and its presence is confirmatory of HCL in this context.[18],[19]


  • Indeterminate fine-needle aspirate (FNA) or aspirates suspicious for papillary thyroid carcinoma are molecularly tested for BRAFV600E and its presence is used to confirm the diagnosis of papillary thyroid carcinoma. A study has shown that the supplementing diagnostic effort by molecular detection of BRAFV600E significantly improves the sensitivity of FNA procedure, from 67.5% with FNAC alone to 89.6% with FNAC combined with molecular testing for BRAFV600E.[20]


  • MLH1-deficient colorectal carcinomas are negatively selected for the presence of BRAFV600E mutation to proceed further for germ line testing to diagnose a case of Lynch syndrome.[21]


  • Identification of BRAFV600E can help conclude the diagnosis of Pleomorphic Xanthoastrocytoma, ganglioglioma and pilocytic astrocytoma in appropriate clinical and pathological settings.[22]


  • Prognostic utility of BRAFV600E

  • BRAFV600E-mutated colorectal carcinomas (CRC) that are microsatellite stable (MSS) have inferior outcomes compared to those that are BRAFV600E wild type along with those that are BRAFV600E mutated but in addition show microsatellite instability (MSI-H) and carry CpG island methylator phenotype. It has been shown in a meta-analysis of 27 independent studies, comprising approximately 24,000 CRC patients, that compared MSS–BRAFwt with MSS–BRAFmut, that latter had a shorter overall survival (OS) with hazard ratio of 2.018. A trend toward inferior OS was also noted in MSI–BRAFmut with hazard ratio of 1.324; however, there was no association of MSI–BRAFwt with OS. Additionally, BRAFmut CRC have been reported to be associated with a high rate of peritoneal metastases.[23]


  • The prognostic value of two most common genetic alterations KIAA1549–BRAF fusion and BRAFV600E mutation in pediatric glial tumors is controversial with some studies showing better outcomes and others showing neutral effect of such genetic alterations.[24]


  • Predictive value of BRAF mutations

  • The predictive value of BRAF gain-of-function mutation has been established in melanoma and NSCLC and several BRAF inhibitors alone or in combinations with MEK inhibitors have been approved for treatment of advanced melanoma [Table 3]. Dual inhibition with dabrafenib + trametinib is approved for BRAFV600E-positive NSCLC. Newer strategies combining BRAF inhibitors with immunotherapy are focus of many ongoing trials and have shown superior survival nevertheless higher rates of adverse events have been observed.[25]


  • BRAFV600E mutation in metastatic CRC makes response to anti-EGFR antibodies highly unlikely unless given with a BRAF inhibitor. BRAFV600E has been included as a predictive biomarker for treatment of metastatic colorectal cancer National Comprehensive Cancer Network (NCCN) version 3.2019—September 26, combinations such as (a) irinotecan + anti EGFR antibody + vemurafenib (b) Dabrafenib + Trametinib+anti EGFR antibody (c) encorafenib + binimetinib + anti EGFR antibody.[26]


  • A phase 2, open-label VE-BASKET study with 26 enrolled subjects has shown gratifying results with vemurafenib in BRAFV600E-mutated Erdheim–Chester disease and Langerhans cell histiocytosis exhibiting objective response rate of 62% and the 2-year progression-free survival rate of 86%.[27]


  • Vemurafenib has recently been approved by US FDA for BRAFV600E-mutated advanced radioactive iodine refractory thyroid cancer. Further, dabrafenib (Tafinlar) has also been granted approval for BRAFV600E-mutated metastatic papillary thyroid carcinoma (PTC). A combination therapy with the BRAF inhibitor dabrafenib and the MEK inhibitor trametinib has also been approved for the treatment of patients with unresectable or metastatic BRAFV600E-positive anaplastic thyroid cancer. The study showed an impressive overall response rate of 69%.[28]


  • Early trials with BRAF–MEK and ERK inhibitors are exploring utility of genome-directed therapy in pediatric low-grade gliomas.



  •   Conclusion Top


    BRAF is an intermediate signal transducer of the RAS–RAF–MEK–ERK signaling pathway, which drives fundamental cell processes of proliferation, differentiation, and secretion. A gain-of-function genetic alteration in the BRAF gene has high oncogenic potentials. Of approximately 45 single-nucleotide variants and 80 fusion rearrangements, BRAFV600E is the archvillain. Fortunately, introduction of BRAF inhibitors used alone or in combination with MEK inhibitor has improved outcomes in BRAF-mutated advanced melanoma and NSCLC. Combination of immune checkpoint inhibitors and BRAF inhibitors are showing promise in further expanding the survival, though serious adverse events with such combinations is a challenge that needs to be addressed. Determining BRAF mutation status in a variety of cancer shall help explore therapeutic options beside helping in diagnosis and anticipating prognosis.

    Financial support and sponsorship

    Nil.

    Conflicts of interest

    There are no conflicts of interest.



     
      References Top

    1.
    International Cancer Genome Consortium; Hudson TJ, Anderson W, Artez A, Barker AD, Bell C, Bernabé RR, et al. International network of cancer genome projects. Nature 2010;464:993-8.  Back to cited text no. 1
        
    2.
    Tokheim CJ, Papadopoulos N, Kinzler KW, Vogelstein B, Karchin R. Evaluating the evaluation of cancer driver genes. Proc Natl Acad Sci U S A 2016;113:14330-5.  Back to cited text no. 2
        
    3.
    Wheeler DA, Wang L. From human genome to cancer genome: The first decade. Genome Res 2013;23:1054-62.  Back to cited text no. 3
        
    4.
    Govender D, Chetty R. Gene of the month: BRAF. J Clin Pathol 2012;65:986-8.  Back to cited text no. 4
        
    5.
    Cantwell-Dorris ER, O’Leary JJ, Sheils OM. BRAFV600E: Implications for carcinogenesis and molecular therapy. Mol Cancer Ther 2011;10:385-94.  Back to cited text no. 5
        
    6.
    My Cancer Genome. Mycancergenome.org. 2019. Available from: https://www.mycancergenome.org/. [Last accessed on 2019 Dec 9].  Back to cited text no. 6
        
    7.
    Galanina N, Bejar R, Choi M, Goodman A, Wieduwilt M, Mulroney C, et al. Comprehensive genomic profiling reveals diverse but actionable molecular portfolios across hematologic malignancies: Implications for next generation clinical trials. Cancers (Basel) 2019;11:E11.  Back to cited text no. 7
        
    8.
    Croce L, Coperchini F, Magri F, Chiovato L, Rotondi M. The multifaceted anti-cancer effects of BRAF-inhibitors. Oncotarget 2019;10:6623-40.  Back to cited text no. 8
        
    9.
    Ghorbani-Aghbolaghi A, Lechpammer M, Ali SF, Ku NK, Dwyre DM, Rashidi HH. An extremely rare case of concurrent BRAF V600E mutation driven hairy cell leukemia and melanoma: Case report and review of literature. Autops Case Rep 2017;7:13-9.  Back to cited text no. 9
        
    10.
    Ascierto PA, Kirkwood JM, Grob JJ, Simeone E, Grimaldi AM, Maio M, et al. The role of BRAF V600 mutation in melanoma. J Transl Med 2012;10:85.  Back to cited text no. 10
        
    11.
    Liu D, Liu Z, Condouris S, Xing M. BRAF V600E maintains proliferation, transformation, and tumorigenicity of BRAF-mutant papillary thyroid cancer cells. J Clin Endocrinol Metab 2007;92:2264-71.  Back to cited text no. 11
        
    12.
    Shin CH, Grossmann AH, Holmen SL, Robinson JP. The BRAF kinase domain promotes the development of gliomas in vivo. Genes Cancer 2015;6:9-18.  Back to cited text no. 12
        
    13.
    Domingo E, Schwartz S Jr. BRAF (v-raf murine sarcoma viral oncogene homolog B1). Atlas Genet Cytogenet Oncol Haematol 2004;8:294-8.  Back to cited text no. 13
        
    14.
    Cheng L, Lopez-Beltran A, Massari F, MacLennan GT, Montironi R. Molecular testing for BRAF mutations to inform melanoma treatment decisions: A move toward precision medicine. Mod Pathol 2018;31:24-38.  Back to cited text no. 14
        
    15.
    Summary of Safety and Effectiveness Data (SSED). 2019. Available from: https://www.accessdata.fda.gov/cdrh_docs/pdf17/P170019B.pdf. [Last accessed 2019 Dec 9].  Back to cited text no. 15
        
    16.
    Dvorak K, Aggeler B, Palting J, McKelvie P, Ruszkiewicz A, Waring P. Immunohistochemistry with the anti-BRAF V600E (VE1) antibody: Impact of pre-analytical conditions and concordance with DNA sequencing in colorectal and papillary thyroid carcinoma. Pathology 2014;46:509-17.  Back to cited text no. 16
        
    17.
    Brown NA, Betz BL, Weigelin HC, Elenitoba-Johnson KS, Lim MS, Bailey NG. Evaluation of allele-specific PCR and immunohistochemistry for the detection of BRAF V600E mutations in hairy cell leukemia. Am J Clin Pathol 2015;143:89-99.  Back to cited text no. 17
        
    18.
    Payandeh M, Sadeghi M, Sadeghi E, Iranshahi N. The prevalence of BRAF V600E mutation in hairy cell leukemia: A systematic review and meta-analysis study. Indian J Med Paediatr Oncol 2019;40:28.  Back to cited text no. 18
      [Full text]  
    19.
    Kim SW, Lee JI, Kim JW, Ki CS, Oh YL, Choi YL, et al. BRAFV600E mutation analysis in fine-needle aspiration cytology specimens for evaluation of thyroid nodule: A large series in a BRAFV600E-prevalent population. J Clin Endocrinol Metab 2010;95:3693-700.  Back to cited text no. 19
        
    20.
    Jin M, Hampel H, Zhou X, Schunemann L, Yearsley M, Frankel WL. BRAF V600E mutation analysis simplifies the testing algorithm for Lynch syndrome. Am J Clin Pathol 2013;140:177-83.  Back to cited text no. 20
        
    21.
    Schindler G, Capper D, Meyer J, Janzarik W, Omran H, Herold-Mende C, et al. Analysis of BRAF V600E mutation in 1,320 nervous system tumors reveals high mutation frequencies in pleomorphic xanthoastrocytoma, ganglioglioma and extra-cerebellar pilocytic astrocytoma. Acta Neuropathol 2011;121:397-405.  Back to cited text no. 21
        
    22.
    Yang Y, Wang D, Jin L, Wu G, Bai Z, Wang J, et al. Prognostic value of the combination of microsatellite instability and BRAF mutation in colorectal cancer. Cancer Manag Res 2018;10:3911-29.  Back to cited text no. 22
        
    23.
    Horbinski C. To BRAF or not to BRAF: Is that even a question anymore? J Neuropathol Exp Neurol 2013;72:2-7.  Back to cited text no. 23
        
    24.
    Khunger A, Khunger M, Velcheti V. Dabrafenib in combination with trametinib in the treatment of patients with BRAF V600-positive advanced or metastatic non-small cell lung cancer: Clinical evidence and experience. Ther Adv Respir Dis 2018;12:1753466618767611.  Back to cited text no. 24
        
    25.
    Ursem C, Atreya CE, Van Loon K. Emerging treatment options for BRAF-mutant colorectal cancer. Gastrointest Cancer 2018;8: 13-23.  Back to cited text no. 25
        
    26.
    Diamond EL, Subbiah V, Lockhart AC, Blay JY, Puzanov I, Chau I, et al. Vemurafenib for BRAF V600-mutant Erdheim–Chester disease and Langerhans cell histiocytosis: Analysis of data from the histology-independent, phase 2, open-label VE-BASKET study. JAMA Oncol 2018;4:384-8.  Back to cited text no. 26
        
    27.
    Crispo F, Notarangelo T, Pietrafesa M, Lettini G, Storto G, Sgambato A, et al. BRAF inhibitors in thyroid cancer: Clinical impact, mechanisms of resistance and future perspectives. Cancers (Basel) 2019;11:1388.  Back to cited text no. 27
        
    28.
    Subbiah V, Kreitman RJ, Wainberg ZA, Cho JY, Schellens JHM, Soria JC, et al. Dabrafenib and trametinib treatment in patients with locally advanced or metastatic BRAF V600-mutant anaplastic thyroid cancer. J Clin Oncol 2018;36:7-13.  Back to cited text no. 28
        


        Figures

      [Figure 1], [Figure 2]
     
     
        Tables

      [Table 1], [Table 2], [Table 3]



     

    Top
     
      Search
     
        Similar in PUBMED
       Search Pubmed for
       Search in Google Scholar for
     Related articles
        Access Statistics
        Email Alert *
        Add to My List *
    * Registration required (free)  

     
      In this article
    Abstract
    Introduction
    Cancer and BR...
    Diagnostic Methods
    Conclusion
    BRAF Gene...
    Value of Identif...
    References
    Article Figures
    Article Tables

     Article Access Statistics
        Viewed3498    
        Printed490    
        Emailed0    
        PDF Downloaded468    
        Comments [Add]    

    Recommend this journal