Open access

BRAF V600E Mutated Gene Variant as a Circulating Molecular Marker in Metastatic Melanoma Patients

Written By

Viviana Vallacchi, Licia Rivoltini and Monica Rodolfo

Submitted: 10 November 2010 Published: 12 September 2011

DOI: 10.5772/20779

From the Edited Volume

Research on Melanoma - A Glimpse into Current Directions and Future Trends

Edited by Mandi Murph

Chapter metrics overview

2,758 Chapter Downloads

View Full Metrics

1. Introduction

Cutaneous metastatic melanoma management has recently approached the age of individualized therapy (Romano et al., 2011). The discovery that the 1799T>A point mutation in the BRAF oncogene (BRAFV600E) occurs in ~50% of melanoma lesions and that melanoma cells bearing the mutation are oncogene addicted, i.e., strictly dependent upon BRAFV600E activity for growth and survival, have pointed to BRAFV600E as a promising target for therapy. Drugs targeting BRAF have been developed, and several clinical trials are currently ongoing. Phase I-II results recently reported remarkable tumor regression in the great majority of patients bearing disseminated BRAFV600E mutated melanoma disease after treatment with BRAFV600E-specific inhibitors.

In these trials, BRAF mutational status is determined to select patients who may benefit from therapy. However, melanoma specimens are not always available to perform this analysis; moreover, a negative result in a single tumor biopsy may cover the presence of mutation-positive tumor lesions. Because blood has been proven to represent a valuable alternative source of tumor-derived cells as well as of tumor-derived DNA, several technical approaches have been studied to detect BRAFV600E in RNA/DNA extracted from blood-derived circulating tumor cells and in circulating free DNA isolated from plasma or serum. For these reasons, circulating BRAFV600E has the potential as both a specific melanoma molecular marker and a monitoring factor to be used to evaluate clinical response.

In this chapter, we summarize the clinical and biological features of BRAF mutation in melanoma. Furthermore, we report a new BRAFV600E detection assay developed in our lab that shows high sensitivity and specificity.

Advertisement

2. BRAFV600E mutation in melanoma

Among the genetic lesions that frequently occur in melanoma, BRAF gene mutation is the most common and is detected in about 50% of melanoma (Davies et al., 2002). The BRAF gene encodes a serine-threonine kinase belonging to the MAPK kinase pathway, also known as the RAS/RAF/MEK/ERK pathway. This signaling pathway regulates important cellular processes, including cell growth, proliferation and migration; in physiological conditions, the signaling is triggered by activated growth factor receptors, which act as binding sites for adapter proteins that subsequently activate a cascade of kinases, including NRAS, BRAF, MEK and ERK, via phosphorylation. Activated ERK translocates to the cell nucleus, where it phosphorylates and activates many different substrates (Held et al., 2011; Poulikakos & Rosen, 2011; Young et al., 2009).

BRAF mutations identified in melanoma are in the kinase domain, which is encoded by exons 11 and 15, and are somatic. Somatic BRAF point mutations have been also detected in approximately 8% of other tumor types, including 30-70% of papillary thyroid cancers and 10% of colorectal cancers, and ovarian, breast and lung cancers (Cantwell-Dorris et al., 2011; Davies et al., 2002; Puzanov et al., 2011). No BRAF germline mutations have been found in familial or sporadic melanoma patients (Lang et al., 2003; Laud et al., 2003; Meyer et al., 2003, although germline mutations have been shown to occur in Noonan, LEOPARD and cardio-facio-cutaneous syndromes, which are developmental disorders with overlapping features including distinctive facial dysmorphia, reduced growth, cardiac defects, skeletal and ectodermal anomalies and variable cognitive deficits (Sarkozy et al., 2009; Tidyman & Raouen, 2009). The relationship between BRAF germline polymorphisms and melanoma risk has also been investigated, and data obtained suggest that BRAF can be considered a low-risk susceptibility gene for melanoma (James et al., 2005; Meyer et al., 2003.

It has been reported that melanocortin 1 receptor (MC1R) variants increase the risk of melanoma with BRAF mutations (Fargnoli et al., 2008; Landi et al., 2006; Scherer et al., 2010). The MC1R gene, which has been identified as a low-risk melanoma susceptibility gene (Williams et al., 2010), encodes a seven-pass transmembrane G-protein receptor that binds alpha-melanocyte stimulating hormone and plays a key role in the pigmentation process (Palmer et al., 2000; Rees, 2004; Valverde et al., 1995). The MC1R gene is highly polymorphic, and gene variants determine a partial or complete loss in the ability of the receptor to transduce signals, thus impairing the switch from pheomelanin to eumelanin production in response to UV radiation exposure (Healy et al., 2000). Further studies are needed to elucidate the mechanisms causing MC1R variants to select for BRAF somatic mutations (Hacker & Hayward, 2008).

BRAF mutations result in the constitutive activation of ERK, resulting in proliferation and growth advantage of melanoma cells. In 15-30% of melanoma, the RAS/RAF/MEK/ERK pathway is constitutively activated through NRAS mutation (Sekulic et al., 2008). As BRAF and NRAS mutations are mutually exclusive, hyperactivation of the MAPK pathway is very frequent in melanoma. Although the constitutive activation of the MAPK pathway is often required to promote the growth and proliferation of neoplastic cells, BRAF mutations are prevalent in melanoma, while mutations in tyrosine kinase receptors or in RAS genes are prevalent in other tumor types. Chromosomal rearrangements leading to the formation of BRAF fusion products, characterized by the lack of the BRAF auto-inhibitory domain and the aberrant activation of the MAPK pathway, have also been reported in pilocytic astrocytoma, thyroid, prostate and gastric cancer as well as melanoma (Ciampi et al., 2005; Cin et al., 2011; Dessars et al., 2007; Palanisamy et al., 2010).

The substitution of a valine (V) for glutamic acid (E) at position 600 (V600E) accounts for >90% of BRAF mutations identified in melanoma. BRAF mutations in melanoma are an early event as they can be detected in nevi and in primary melanoma (Figure 1) (Rodolfo et al., 2004; Thomas et al., 2006). Even if 60-70% of benign and dysplastic nevi show the BRAFV600E mutation, most of them do not progress to melanoma, suggesting that further alterations are necessary to promote malignant progression. In particular, it has been proposed that BRAF mutations may induce senescence and that abrogation of the mechanisms regulating this cellular process are required to induce tumor progression (Michaloglou et al., 2005). In primary melanoma, BRAFV600E mutation is rarely detected in lentigo maligna lesions which arise in chronically sun-exposed skin and show a high rate of RAS mutations, and in acral and mucosal melanoma that arise in non-exposed skin, which may show KIT gene mutation (Platz et al., 2008). Melanoma occurring in childhood and adolescence, as well as those occurring in patients with a family history of melanoma, show BRAFV600E mutation (Daniotti et al., 2009). Melanoma that arise in intermittently exposed body sites, in skin lacking signs of chronic sun-induced damage, and in younger people, show a >80% rate of BRAFV600E mutation (Curtin et al., 2005). BRAF mutation frequency appears higher in advanced lesions than primary tumors, and it does not seem to be related to the site of metastases. Moreover, different studies have reported that BRAF mutation is maintained during progression from primary tumor to metastatic lesions or is acquired during the development of metastases (Houben et al., 2004; Omholt et al., 2003; Shinozaki et al., 2004). These results suggested a possible direct link between mutated BRAF and the metastatic potential of melanoma cells.

Figure 1.

B: Benign nevi; B/S: Blue and Spitz nevi; D: Dysplastic nevi; SS: Superficial Spreading melanoma; N: Nodular melanoma; L: Lentigo maligna melanoma; A: Acral lentiginous melanoma; M: Mucosal melanoma; No: Nodal metastases; Cu: Cutaneous metastases; V: Visceral metastases; Ce: Cerebral metastases.Frequency of BRAFV600E mutation in nevi and melanoma lesions.

Several authors have studied the effects of BRAFV600E on global gene expression profiles of melanoma cells by microarray analysis and reported a BRAF mutation-associated gene expression signature (Pavey et al., 2004; Bloethner et al., 2005; Johansson et al., 2007). In particular, genes that encoded proteins involved in RAS/RAF/MEK/ERK signaling were identified among the genes differentially expressed between melanoma cell lines with or without BRAF mutation (Bloethner et al., 2005). In addition, a classifier able to discriminate between BRAF mutant and BRAF wild-type melanoma with high accuracy was built, including genes encoding phosphates and other genes biologically related to melanoma progression (Pavey et al., 2004). On the contrary, Hoek et al. failed to find a BRAF signature but identified three sample cohorts that represented melanoma groups characterized by different metastatic potential (Hoek et al., 2004). This discrepancy could be explained by considering the methods used to perform the analysis of array data; in fact, when the data by Hoek were re-analyzed with another statistical approach, a BRAF signature could be identified in these data (Johansson et al., 2007). Taken together, these results support the presence of a gene expression profile associated with BRAF mutation in melanoma and point to the genes that are potentially novel therapeutic targets.

Advertisement

3. BRAFV600E as a therapeutic target

The frequency and specificity of BRAFV600E mutation, together with the strict dependence of melanoma cell growth and survival on BRAFV600E activity (a phenomenon called oncogene addiction), have pointed to BRAFV600E as a promising therapeutic target.

Several BRAF inhibitors have been produced in the last years that have been or are currently being clinically tested (Sheperd et al., 2010). The first compound tested in clinical trials was Sorafenib (BAY43-9006), a multi-kinase inhibitor that targeted both wild-type and mutated BRAF, CRAF and other protein kinases, such as VEGFR2 and -3, PDGF, p38 MAPK, cKIT, FMS and RET (Wellbrock & Hurlstone, 2010). Sorafenib showed poor clinical activity when tested as a single agent, and in phase III trials in both front- and second-line therapies in combination with carboplatin and paclitaxol (Eisen et al., 2006; Hauschild et al., 2009). Other multi-kinase inhibitors that show a higher selectivity for BRAF than Sorafenib are currently under investigation in clinical trials (Dienstmann & Tabernero, 2011).

Several compounds that selectively inhibit BRAF have also been developed. Among them, GSK2118436 (SB-590885) has been tested in a phase I-II clinical trial and shows clinical responses in 60% of melanoma patients with BRAFV600 tumors, including patients having BRAFV600K and BRAFV600G mutations, with good tolerability (Kefford et al., 2010). Moreover, treatment with GSK2118436 induced a 20-100% reduction in the size of central nervous system lesions in patients with previously untreated brain metastases (Long et al., 2010).

Recently, the results of a phase I-II study that tested a specific BRAFV600E inhibitor, PLX4032 (RO5185426), were reported. Treatment with PLX4032 induced a complete or partial tumor regression in 81% of patients who had melanoma with BRAFV600E mutation, including progression-free survival for more than 7 months and manageable side effects, while patients with BRAF wild-type tumors showed no evidence of tumor regression (Flaherty et al., 2010). As a side effect, 31% of patients treated with PLX4032 developed low-grade squamous cell carcinomas, which were reported to occur also in patients treated with Sorafenib (Arnault et al., 2009). This side effect is possibly due to the selective mechanism of action of PLX4032 that shuts down only the activity of BRAFV600E while inducing the formation of BRAF-RAF1 heterodimers and RAF1-RAF1 homodimers, thus inducing hyper-activation of the MAPK pathway in both tumor cells and normal skin cells with wild-type BRAF (Hatzivassiliou et al., 2010; Heidorn et al., 2010; Poulikakos et al., 2010).

Advertisement

4. Mechanisms of resistance to BRAF inhibitors

Even if BRAFV600E tumors initially respond to PLX4032 treatment, the majority of patients relapsed within 2-18 months and developed resistance to the treatment. Furthermore, a subset of BRAFV600E tumors showed primary resistance as about 20% of patients did not respond to PLX4032 treatment (Flaherty et al., 2010). These findings indicate that the development of new therapeutic strategies using PLX4032 in combination with other targeted agents could be useful to prevent the acquisition of resistance. Several studies investigating the molecular mechanisms that promote resistance to RAF inhibitors have been recently reported. The restoration of MEK activity in BRAFV600E melanoma appears to be a crucial event in promoting the acquisition of resistance (Poulikakos & Rosen, 2011; Solit & Rosen, 2011; Solit & Sawyers, 2010; Tuma, 2011). In particular, MEK activity was restored by overexpressing other kinases such as RAF1 and COT/TPL2 (Johannessen et al., 2010) or by the de novo acquisition of a mutation in the NRAS gene (Nazarian et al., 2010). The COT gene was amplified in cell lines that showed intrinsic resistance to PLX4032 (Johannessen et al., 2010). Surprisingly, no secondary BRAF mutations were detected in tumors from patients with acquired resistance (Nazarian et al., 2010). Wagle et al. identified an activating mutation at codon 121 of MEK1 in the tumor from a patient who relapsed after developing resistance to PLX4032 treatment, thus demonstrating for the first time that resistance to PLX4032 is associated with the development of activating mutations in kinases downstream of BRAFV600E (Wagle et al., 2011). This discovery highlights the importance of establishing new combined therapies using MEK or ERK inhibitors with PLX4032 to overcome resistance. In fact, data obtained in preclinical studies demonstrated a synergism between BRAF and MEK inhibitors AZD6244 and GSK1120212 (Emery et al., 2009; Joseph et al., 2010; Paraiso et al., 2010). However, mechanisms that promote the acquisition of resistance independently of MEK activation have been described, including the increased activation of the receptor tyrosine kinases PDGFRβ (Nazarian et al., 2010) or IGF1R (Villaneuva et al., 2010), suggesting that the combination of receptor tyrosine kinase inhibitors with PLX4032 could be effective in the treatment of these patients. However in most patients, the mechanisms that promote the acquisition of resistance remain unclear (Nazarian et al., 2010).

Advertisement

5. BRAFV600E as a circulating disease biomarker

As blood has been proved to represent a valuable alternative source of tumor-derived cells and tumor-derived DNA/RNA, circulating BRAFV600E represents a potential circulating disease biomarker that could be useful when melanoma specimens are not available to test the BRAF mutational status for the selection of patients who will benefit from treatment with BRAF inhibitors. In addition, it could be used as a monitoring factor to evaluate clinical response.

Several studies reported that BRAFV600E is detectable in DNA/RNA extracted from circulating melanoma cells (CMC) (Kitago et al., 2009; Oldenburg et al., 2008). The assessment of CMC for monitoring the efficacy of therapeutic treatment and for predicting the disease outcome of melanoma patients has been proposed. Currently, RT-PCR and quantitative real-time RT-PCR are the methods most frequently used to detect CMC in melanoma. Both techniques are used to amplify genes expressed in melanoma cells, such as tyrosinase, MART-1, MAGE-3A and MITF (Koyanagi et al., 2010). Detection of the mutated BRAF variant in blood samples requires the efficient isolation of CMC (Kitago et al., 2009) or the development of extremely sensitive techniques to detect the mutant sequence in a large excess of wild-type BRAF forms (Oldenburg et al., 2008).

Circulating free DNA (cfDNA) isolated from plasma or serum samples represents an alternative source of melanoma-derived DNA. Several studies reported the feasibility of detecting BRAFV600E mutation in the cfDNA from patients with melanoma (Board et al., 2009; Daniotti et al., 2007; De Giorgi et al., 2010; Pinzani et al., 2010; Yancovitz et al., 2007). Interestingly, Shinozaki and coworkers reported that the detection of circulating BRAV600E in the serum of patients treated with biochemotherapy correlates with poorer outcomes due to absence of response to the treatment (Shinozaky et al., 2007).

Some important limitations should be overcome to consider BRAFV600E as a reliable circulating disease biomarker. In fact, the studies previously mentioned demonstrated that BRAFV600E is detectable at stage IV and only in a few stage III melanoma patients, suggesting that it does not represent a marker for the detection of the disease in early-stage patients. Moreover, when matched plasma/serum and tumor samples from melanoma patients were tested for BRAFV600E, the concordance between the BRAF mutation rates of cfDNA and tumors showed some discrepancies, which could be due to a low sensitivity of the techniques used to perform the mutational analysis or to the heterogeneity of the tumor for the BRAFV600E mutation. For these reasons, different methods were developed to detect BRAFV600E mutation in high levels of BRAF wild-type DNA by increasing the specificity and sensitivity of the assays, as shown in Table 1, mainly through enriching the sample for the mutant variant or by selectively inhibiting the amplification of the BRAF wild-type form (Kitago et al., 2009; Oldenburg et al., 2008; Pinzani et al., 2011; Shinozaki et al., 2007; Yancovitz et al., 2007)

Table 1.

PNA: Peptide Nucleic Acid; LNA: Locked Nucleic Acid; PBAS: Primer-Blocking Allele-Specific; ARMS: Amplification Refractory Mutation System; COLD: CO-amplification at Lower Denaturation temperature; mut: mutated; wt: wild-type; 1U: amount of target DNA contained in 1 µg/ml of genomic DNA; PBC: Peripheral Blood Cells; FFPE: Formalin Fixed Paraffin EmbeddedMethods developed to detect BRAFV600E.

5.1. Other melanoma circulating biomarkers

Melanoma serum markers that have significant potential both as prognostic indicators and for monitoring the treatment response include lactate dehydrogenases (LDH), S100 calcium binding protein B (S100B), and melanoma inhibitory activity (MIA) molecule.

LDH are cytochrome c- or NAD(P)-dependent enzymes that act on either D- or L-lactate. Serum LDH is the only circulating biomarker shown to have a prognostic relevance in melanoma. Several studies have shown that high levels of circulating LDH correlate with a poor prognosis in stage IV melanoma patients and in other neoplastic diseases (Balch et al., 2009; Bedikian et al., 2008; Keilholz et al., 2002). For this reason, LDH was included in the current AJCC staging system, and its level is currently determined in melanoma patients having distant metastasis because patients with elevated LDH are assigned directly to the M1C category without considering the site of distant metastasis (Dickson & Gershenwald, 2011).

S100B is a protein that belongs to the S100 protein family and is mainly expressed by astrocytes, where it acts as a neurotrophic factor to promote neuronal survival. S100B is a well-characterized melanoma marker, and it is used as a diagnostic marker of melanocytic skin lesions in immunohistochemical staining. Several studies pointed out S100B as a prognostic marker of disease progression (Gogas et al., 2009; Jury et al., 2000) as increased serum levels in melanoma patients were predictive of disease progression. Even if not included in the AJCC staging system, Swiss and German guidelines recommend the determination of S100B serum levels in patients with Breslow thickness >1 mm every 3-6 months (Dummer et al., 2005; Garbe et al., 2007; Garbe et al., 2008).

MIA is a small protein secreted by malignant melanoma cells that exhibits an inhibitory effect on cell growth in vitro (Blesch et al., 1994). Even if a correlation between high MIA serum levels and metastatic melanoma progression has been reported (Bosserhoff et al., 1997; Stahlecker et al., 2000), MIA was shown to have lower sensitivity and specificity as a melanoma marker than S100B and LDH (Krahn et al., 2001).

Recently, microRNAs (miRNAs) have been proposed as a new class of potential circulating biomarkers that are detectable in various body fluids. miRNAs are non-coding RNAs consisting of 18-24 nucleotides that regulate mRNA and protein levels mainly by inducing mRNA degradation or by inhibiting translation (Ambros, 2004; Bartel, 2004). Recently, deregulation of a group of miRNAs was found in melanoma lesions in association with BRAF mutational status (Caramuta et al., 2010). miRNAs are also released into the extracellular space, where they can be found free or contained within vesicles such as microvesicles, exosomes, apoptotic vesicles and senescent bodies. The functional role of extracellular miRNAs as an intercellular communication system is poorly characterized (Reid et al., 2010). Extracellular miRNAs have been identified as ideal tumor circulating biomarkers because of their stability and easy quantification (Etheridge et al., 2010). For these reasons, circulating miRNAs have been investigated in many tumor types, including lung, colorectal, ovarian and pancreatic cancers, to evaluate their prognostic and diagnostic value (Reid et al., 2010). Few studies have assessed circulating miRNAs in the context of melanoma. Kanemaru et al. demonstrated that miRNA-221 serum levels are higher in melanoma patients than in healthy controls; in addition, miRNA-221 levels directly correlate with tumor thickness, staging and disease course (Kanemaru et al., 2011). In another study, 16 miRNAs were identified deregulated in blood cells of melanoma patients by comparison of miRNA expression profiles in blood cells of healthy donors; moreover, they were sufficient to distinguish melanoma patients from healthy individuals with high accuracy (Leidinger et al., 2010). Taken together, these studies suggest that miRNAs potentially could be prognostic and diagnostic circulating markers in melanoma, although larger studies and the standardization of isolation and detection techniques are needed to confirm these results.

Advertisement

6. Allele-specific real-time PCR-based detection of circulating BRAFV600E

It is possible to selectively eliminate the BRAF wild-type sequence and thus improve the sensitivity of the PCR performed to detect mutated circulating BRAF by taking advantage of the presence of a TspRI enzyme restriction site located at codon 600 of the BRAF wild-type sequence (Rimoldi et al., 2003). This restriction site is abrogated by BRAFV600E, and therefore it is possible to enrich for the BRAV600E allele by selectively eliminating wild-type sequences by performing TspRI digestion. We modified the experimental conditions described by Rimoldi et al. to improve sensitivity and specificity of an allele-specific TaqMan-based real-time method to detect BRAFV600E in colorectal cancer tissues (Benlloch et al., 2006). Figure 2 summarizes the steps of the assay developed to screen for BRAV600E mutation plasma or melanoma tissue biopsies.

Figure 2.

Schematic representation of TaqMan-based Real-Time PCR method developed to detect few copies of BRAFV600E in a large amount of wild-type DNA.

Experimental conditions of each step are reported in paragraph 6.1.

6.1. Overview of the protocol

A 224-bp fragment that includes codon 600 of the BRAF gene is amplified from 5 ng DNA in a final reaction volume of 50 µl (Figure 2A). Amplification was performed with a pre-cycling hold at 95°C for 7 min followed by 37 cycles of PCR (95°C for 1 min, 55°C for 1 min and 72°C for 1 min) and a final extension at 72°C for 7 min using primers for exon 15 amplification reported by Davies (Davies et al., 2002). Twenty microliters of the PCR product were mixed with 1X NEB Buffer 4 supplemented with 100 µg/ml BSA (New England Biolabs) and then subjected to restriction digestion at 65°C overnight with 15 U TspRI (New England Biolabs) in a final digestion mix volume of 50 µl to enrich samples for the BRAFV600E mutant allelic variant (Figure 2B). Two microliters of the digestion product were used to perform an allele-specific TaqMan-based real-time PCR analysis (Figure 2C). The final reaction volume of 20 µl contained 10 µl 2X of TaqMan Genotyping Master Mix (Applied Biosystems), 18 pmol of each primer (BRAF-51F and BRAF-176R) and 5 pmol of each probe (BRAFmut and BRAFwt). The primer and probe sequences were reported previously by Benlloch (Benlloch et al., 2006). Amplification and detection were performed with an ABI PRISM 7900HT (Applied Biosystems) using the standard thermal profile conditions of the Absolute Quantification protocol. Data analysis was performed using the SDS (Sequence Detection System) version 2.2.2 software. Each experiment was performed in duplicate.

6.2. Results

Specificity of the technique was tested by assaying dilutions of BRAF mutated DNA (5 ng/µl) in wild-type DNA (5 ng/µl). The mutated DNA was obtained from a heterozygous melanoma cell line showing 2 copies of BRAF gene. Therefore, the allelic ratio was calculated considering one mutated allele out of 4 total alleles. Results obtained show that 1 copy of V600E allele can be detected when diluted in 8X105 copies of wild-type alleles. Sensitivity of the technique was assayed by testing progressive dilutions in water of the BRAF mutated DNA. Results obtained show that BRAFV600E mutation can be detected starting from 6.25X10-5 ng of DNA.

This method showed an increase in both sensitivity and specificity when compared to the assays previously used in our lab to detect BRAFV600E (Daniotti et al., 2007). As shown in Figures 3 and 4, the selective elimination of the BRAF wild-type allele is a critical step required to increase both parameters. In fact, the BRAF mutated allele became detectable only when the wild-type allele was greatly reduced (Figure 3) or eliminated (Figure 4) after digestion with TsprRI.

Taken together, these results indicate that the new assay has an improved sensitivity and specificity for detecting BRAFV600E when tested on genomic DNA from melanoma when diluted in an excess of wild-type DNA or when present in a few copies as in water dilutions. Preliminary results obtained by testing matched plasma and tissues samples indicate that more samples test positive for BRAFV600E compared to the previously described technique (Daniotti et al., 2007) and suggest a potential clinical application of this technique.

Figure 3.

TspRI digestion increases the specificity of BRAFV600E allele-specific real-time PCR.

TspRI digestion reduces the excess of the wild-type allele that is detected 9 cycles later compared to the undigested sample and allows the detection of the BRAFV600E allele. The red line represents the threshold line. wt: wild-type. mut: mutated.

Figure 4.

TspRI digestion increases the sensitivity of the BRAFV600E allele-specific real-time PCR.

The amplification plot shows that the complete elimination of the BRAF wild-type template by TspRI digestion improves the sensitivity of mutated allele-specific PCR, anticipating its detection of about 14 cycles compared to undigested samples. The red line represents the threshold line. wt: wild-type. mut: mutated.

Advertisement

7. Conclusion

BRAFV600E currently represents the most specific circulating tumor marker available for cutaneous melanoma, although it will only detect about 50% of melanoma. Circulating BRAFV600E can be used to select patients to be treated with BRAF inhibitors when the tissue samples are not available for the analysis. In addition, detection methods for circulating BRAFV600E can be used to monitor the treatment response and evaluate disease relapse during follow-up. However, to use BRAFV600E as a blood marker, more sensitive technologies must be designed and validated to improve the sensitivity and specificity of the assays used to detect this mutation.

Advertisement

Acknowledgments

VV is recipient of a fellowship by the Italian Health Ministry, Grant RFPS 2006-4-341458.

References

  1. 1. AmbrosV.2004The functions of animal microRNAs. Nature, 4317006350355
  2. 2. ArnaultJ. P.WechslerJ.RobertC.2009Keratoacanthomas and squamous cell carcinomas in patients receiving sorafenib. J Clin Oncol, 27235961
  3. 3. BalchC. M.GershenwaldJ. E.SondakV. K.2009Final version of 2009 AJCC melanoma staging and classification. J Clin Oncol, 273661996206
  4. 4. BartelD. P.2004MicroRNAs: genomics, biogenesis, mechanism, and function. Cell, 1162281297
  5. 5. BedikianA. Y.MMJohnsonHwuP.2008Prognostic factors that determine the long-term survival of patients with unresecTable metastatic melanoma. Cancer Invest, 266624633
  6. 6. BenllochS.PayáA.MassutíB.2006Detection of BRAF V600E mutation in colorectal cancer: comparison of automatic sequencing and real-time chemistry methodology. J Mol Diagn, 85540543
  7. 7. BleschA.BosserhoffA. K.BogdahnU.1994Cloning of a novel malignant melanoma-derived growth-regulatory protein, MIA. Cancer Res, 542156955701
  8. 8. BloethnerS.SnellmanE.KumarR.2007Differential gene expression in melanocytic nevi with the V600E BRAF mutation. Genes Chromosomes Cancer, 461110191027
  9. 9. BoardR. E.EllisonG.HughesA.2009Detection of BRAF mutations in the tumour and serum of patients enrolled in the AZD6244 (ARRY-142886) advanced melanoma phase II study. Br J Cancer, 1011017241730
  10. 10. BosserhoffA. K.KaufmannM.BuettnerR.1997Melanoma-inhibiting activity, a novel serum marker for progression of malignant melanoma. Cancer Res, 571531493153
  11. 11. Cantwell-DorrisE. R.O’LearyJ. J.SheilsO. M.2011BRAFV600E: Implications for Carcinogenesis and Molecular Therapy. Mol Cancer Ther, 103385394
  12. 12. CaramutaS.EgyháziS.LuiW. O.2010MicroRNA expression profiles associated with mutational status and survival in malignant melanoma. J Invest Dermatol, 130820622070
  13. 13. CiampiR.KnaufJ. A.NikiforovY. E.2005Oncogenic AKAP9-BRAF fusion is a novel mechanism of MAPK pathway activation in thyroid cancer. J Clin Invest, 115994101
  14. 14. CinH.MeyerC.PfisterS. M.2011Oncogenic FAM131B-BRAF fusion resulting from 7q34 deletion comprises an alternative mechanism of MAPK pathway activation in pilocytic astrocytomaActa Neuropathol, (2011)
  15. 15. CurtinJ. A.FridlyandJ.BastianB. C.2005Distinct sets of genetic alterations in melanoma. N Engl J Med, 3532021352147
  16. 16. DaniottiM.VallacchiV.RodolfoM.2007Detection of mutated BRAFV600E variant in circulating DNA of stage III-IV melanoma patients. Int J Cancer, 120111243912444
  17. 17. DaniottiM.FerrariA.RodolfoM.2009Cutaneous melanoma in childhood and adolescence shows frequent loss of INK4A and gain of KIT. J Invest Dermatol, 129717591768
  18. 18. DaviesH.BignellG. R.FutrealP. A.2002Mutations of the BRAF gene in human cancer. Nature, 4176892949954
  19. 19. De GiorgiV.PinzaniP.MassiD.2010Circulating benign nevus cells detected by ISET technique: Warning for melanoma molecular diagnosis. Arch Dermatol, 1461011201124
  20. 20. DessarsB.De RaeveL. E.HeimannP.2007Chromosomal translocations as a mechanism of BRAF activation in two cases of large congenital melanocytic nevi. J Invest Dermatol, 127614681470
  21. 21. DicksonP. V.GershenwaldJ. E.2011Staging and prognosis of cutaneous melanoma. Surg Oncol Clin N Am, 201117
  22. 22. DienstmannR.TaberneroJ.2011BRAF as a Target for Cancer Therapy.Anticancer Agents Med Chem, 2011
  23. 23. DummerR.PanizzonR.BurgG.2005Updated Swiss guidelines for the treatment and follow-up of cutaneous melanoma. Dermatology, 21013944
  24. 24. EisenT.AhmadT.MJRatain2006Sorafenib in advanced melanoma: a Phase II randomised discontinuation trial analysis. Br J Cancer, 955581586
  25. 25. EmeryC. M.VijayendranK. G.GarrawayL. A.2009MEK1 mutations confer resistance to MEK and B-RAF inhibition. Proc Natl Acad Sci U S A, 106482041120416
  26. 26. EtheridgeA.LeeI.WangK.2011Extracellular microRNA: a new source of biomarkers. Mutat Res, 2011
  27. 27. FargnoliM. C.PikeK.LandiM. T.2008MC1R variants increase risk of melanoma harboring BRAF mutations. J Invest Dermatol, 1281024852490
  28. 28. FlahertyK. T.PuzanovI.ChapmanP. B.2010Inhibition of mutated, activated BRAF in metastatic melanoma. N Engl J Med, 3639809819
  29. 29. GarbeC.HauschildA.KaufmannR.2007Evidence and interdisciplinary consense-based German guidelines: diagnosis and surveillance of melanoma. Melanoma Res, 176393399
  30. 30. GarbeC.SchadendorfD.HauschildA.2008Short German guidelines: malignant melanoma. J Dtsch Dermatol Ges, 6Suppl 1, S9S14
  31. 31. GogasH.EggermontA. M.DummerR.2009Biomarkers in melanoma. Ann Oncol, 206vi8v13
  32. 32. HackerE.HaywardN. K.2008Germline MC1R variants and BRAF mutant melanoma. J Invest Dermatol, 1281023542356
  33. 33. HatzivassiliouG.SongK.MalekS.2010RAF inhibitors prime wild-type RAF to activate the MAPK pathway and enhance growth. Nature, 4647287431435
  34. 34. HauschildA.AgarwalaS. S.KeilholzU.2009Results of a phase III, randomized, placebo-controlled study of sorafenib in combination with carboplatin and paclitaxel as second-line treatment in patients with unresecTable stage III or stage IV melanoma. J Clin Oncol, 271728232830
  35. 35. HealyE.FlannaganN.ReesJ. L.2000Melanocortin-1-receptor gene and sun sensitivity in individuals without red hair. Lancet, 355920910721073
  36. 36. HeidornS. J.MilagreC.MaraisR.2010Kinase-dead BRAF and oncogenic RAS cooperate to drive tumor progression through CRAF. Cell, 1402209221
  37. 37. HeldL.EigentlerT. K.BauerJ.2011Oncogenetics of melanoma: basis for molecular diagnostics and therapyJ Dtsch Dermatol Ges, (2011)
  38. 38. HoekK. S.SchlegelN. C.DummerR.2006Metastatic potential of melanoma defined by specific gene expression profiles with no BRAF signature. Pigment Cell Res, 194290302
  39. 39. HoubenR.BeckerJ. C.RappU. R.2004Constitutive activation of the Ras-Raf signaling pathway in metastatic melanoma is associated with poor prognosis. J Carcinog, 316
  40. 40. JamesM. R.RothR. B.DuffyD. L.2005BRAF polymorphisms and risk of melanocytic neoplasia. J Invest Dermatol, 125612521258
  41. 41. JohannessenC. M.BoehmJ. S.GarrawayL. A.2010COT drives resistance to RAF inhibition through MAP kinase pathway reactivation. Nature, 4687326968972
  42. 42. JohanssonP.PaveyS.HaywardN.2007Confirmation of a BRAF mutation-associated gene expression signature in melanoma. Pigment Cell Res, 203216221
  43. 43. JosephE. W.CAPratilasRosenN.2010The RAF inhibitor PLX4032 inhibits ERK signaling and tumor cell proliferation in a V600E BRAF-selective manner. Proc Natl Acad Sci U S A, 107331490314908
  44. 44. JuryC. S.Mc AllisterE. J.MacKie. R. M.2000Rising levels of serum S100 protein precede other evidence of disease progression in patients with malignant melanoma. Br J Dermatol, 1432269274
  45. 45. KanemaruH.FukushimaS.IhnH.2011The circulating microRNA-221 level in patients with malignant melanoma as a new tumor marker. J Dermatol Sci, 613187193
  46. 46. Kefford, R., Arkenau, H., & Lebowitz, PF. Phase I/II study of GSK2118436, a selective inhibitor of oncogenic mutant BRAF kinase, in patients with metastatic melanoma and other solid tumors.J Clin Oncol, 28(15s), abstract 8503
  47. 47. KeilholzU.MartusP.EggermontA. M.2002Prognostic factors for survival and factors associated with long-term remission in patients with advanced melanoma receiving cytokine-based treatments: second analysis of a randomised EORTC Melanoma Group trial comparing interferon-alpha2a (IFNalpha) and interleukin 2 (IL-2) with or without cisplatin. Eur J Cancer, 381115011511
  48. 48. KitagoM.KoyanagiK.DSHoon2009mRNA expression and BRAF mutation in circulating melanoma cells isolated from peripheral blood with high molecular weight melanoma-associated antigen-specific monoclonal antibody beads. Clin Chem, 554757764
  49. 49. KoyanagiK.O’DayS. J.DSHoon2010Serial monitoring of circulating tumor cells predicts outcome of induction biochemotherapy plus maintenance biotherapy for metastatic melanoma. Clin Cancer Res, 16824022408
  50. 50. KrähnG.KaskelP.PeterR. U.2001S100 beta is a more reliable tumor marker in peripheral blood for patients with newly occurred melanoma metastases compared with MIA, albumin and lactate-dehydrogenase. Anticancer Res, 212B13111316
  51. 51. LandiM. T.BauerJ.BastianB. C.2006MC1R germline variants confer risk for BRAF-mutant melanoma. Science, 3135786521522
  52. 52. LangJ.BoxerM.MacKie. R.2003Absence of exon 15 BRAF germline mutations in familial melanoma. Hum Mutat, 213327330
  53. 53. LaudK.KannengiesserC.Bressac-dePaillerets. B.2003BRAF as a melanoma susceptibility candidate gene? Cancer Res, 631230613065
  54. 54. LeidingerP.KellerA.MeeseE.2010High-throughput miRNA profiling of human melanoma blood samples. BMC Cancer, 10262
  55. 55. LongG. V.KeffordR. F.FalchookG.2010Phase ½ study of GSK2118436, a selective inhibitor of 600mutant BRAf kinase: evidence of activity in melanoma brain metastases. Annals Oncol, 21 (Suppl. 8), abstract LBA 27
  56. 56. MeyerP.KlaesR.GarbeC.2003Exclusion of BRAFV599E as a melanoma susceptibility mutation. Int J Cancer, 10617880a)
  57. 57. MeyerP.SergiC.GarbeC.2003Polymorphisms of the BRAF gene predispose males to malignant melanoma.J Carcinog, 217b)
  58. 58. MichaloglouC.VredeveldL. C.DSPeeper2005BRAFE600-associated senescence-like cell cycle arrest of human naevi. Nature, 4367051720724
  59. 59. NazarianR.ShiH.LoR. S.2010Melanoma acquire resistance to B-RAF(V600E) inhibition by RTK or NRAS upregulation. Nature, 4687326973977
  60. 60. OldenburgR. P.MSLiuMSKolodney2008Selective amplification of rare mutations using locked nucleic acid oligonucleotides that competitively inhibit primer binding to wild-type DNA. J Invest Dermatol, 1282398402
  61. 61. OmholtK.PlatzA.HanssonJ.2003NRAS and BRAF mutations arise early during melanoma pathogenesis and are preserved throughout tumor progression. Clin Cancer Res, 91764836488
  62. 62. PalanisamyN.AteeqB.ChinnaiyanA. M.2010Rearrangements of the RAF kinase pathway in prostate cancer, gastric cancer and melanoma. Nat Med., 167793798
  63. 63. PalmerJ. S.DuffyD. L.SturmR. A.2000Melanocortin-1 receptor polymorphisms and risk of melanoma: is the association explained solely by pigmentation phenotype? Am J Hum Genet, 661176186
  64. 64. ParaisoK. H.FedorenkoI. V.SmalleyK. S.2010Recovery of phospho-ERK activity allows melanoma cells to escape from BRAF inhibitor therapy. Br J Cancer, 1021217241730
  65. 65. PaveyS.JohanssonP.HaywardN. K.2004Microarray expression profiling in melanoma reveals a BRAF mutation signature. Oncogene, 232340604067
  66. 66. PinzaniP.SalviantiF.OrlandoC.2010Allele specific taqman-based real-time PCR assay to quantify circulating BRAFV600E mutated DNA in plasma of melanoma patients. Clin Chim Acta, 41117-1813191324
  67. 67. PinzaniP.SantucciC.OrlandoC.2011BRAF(600Edetection in melanoma is highly improved by COLD-PCR. Clin Chim Acta, 2011
  68. 68. PlatzA.EgyhaziS.HanssonJ.2008Human cutaneous melanoma; a review of NRAS and BRAF mutation frequencies in relation to histogenetic subclass and body site. Mol Oncol, 14395405
  69. 69. PoulikakosP. I.ZhangC.RosenN.2010RAF inhibitors transactivate RAF dimers and ERK signalling in cells with wild-type BRAF. Nature, 4647287427430
  70. 70. PoulikakosP. I.RosenN.2011Mutant BRAF melanoma--dependence and resistance. Cancer Cell, 1911115
  71. 71. PuzanovI.BurnettP.FlahertyK. T.2011The cardiofaciocutaneous syndrome. Mol Oncol, (2011)
  72. 72. Rees JL.2004The genetics of sun sensitivity in humans. Am J Hum Genet, 755739751
  73. 73. ReidG.KirschnerM. B.van ZandwijkN.2010Circulating microRNAs: Association with disease and potential use as biomarkersCrit Rev Oncol Hematol, (2010)
  74. 74. RimoldiD.SalviS.CerottiniJ. C.2003Lack of BRAF mutations in uveal melanoma.Cancer Res, 631857125715
  75. 75. RodolfoM.DaniottiM.VallacchiV.2004Genetic progression of metastatic melanoma. Cancer Lett, 2142133147
  76. 76. RomanoE.SchwartzG. K.CarvajalR. D.2011Treatment implications of the emerging molecular classification system for melanomaLancet Oncol
  77. 77. SarkozyA.CartaC.TartagliaM.2009Germline BRAF mutations in Noonan, LEOPARD, and cardiofaciocutaneous syndromes: molecular diversity and associated phenotypic spectrum. Hum Mutat, 304695702
  78. 78. SchererD.RachakondaP. S.KumarR.2010Association between the germline MC1R variants and somatic BRAF/NRAS mutations in melanoma tumors. J Invest Dermatol, 1301228442848
  79. 79. SekulicA.HaluskaP.Jr MarkovicS. N.MelanomaStudy.Groupof.MayoClinic.CancerCenter.2008Malignant melanoma in the 21st century: the emerging molecular landscape. Mayo Clin Proc, 837825846
  80. 80. ShepherdC.PuzanovI.SosmanJ. A.2010B-RAF inhibitors: an evolving role in the therapy of malignant melanoma. Curr Oncol Rep, 12331463152
  81. 81. ShinozakiM.FujimotoA.DSHoon2004Incidence of BRAF oncogene mutation and clinical relevance for primary cutaneous melanoma. Clin Cancer Res, 10517531757
  82. 82. ShinozakiM.O’DayS. J.DSHoon2007Utility of circulating B-RAF DNA mutation in serum for monitoring melanoma patients receiving biochemotherapy. Clin Cancer Res, 13720682074
  83. 83. SolitD.SawyersC. L.2010Drug discovery: How melanoma bypass new therapy. Nature, 4687326902903
  84. 84. SolitD. B.RosenN.2011Resistance to BRAF inhibition in melanoma. N Engl J Med, 3648772774
  85. 85. StahleckerJ.GaugerA.HeinR.2000MIA as a reliable tumor marker in the serum of patients with malignant melanoma. Anticancer Res, 206D50415044
  86. 86. ThomasN. E.2006BRAF somatic mutations in malignant melanoma and melanocytic naevi. Melanoma Res, 16297103
  87. 87. TidymanW. E.RauenK. A.2009The RASopathies: developmental syndromes of Ras/MAPK pathway dysregulation. Curr Opin Genet Dev, 193230236
  88. 88. TumaR. S.2011Getting around PLX4032: studies turn up unusual mechanisms of resistance to melanoma drug. J Natl Cancer Inst, 1033170171
  89. 89. ValverdeP.HealyE.ThodyA. J.1995Variants of the melanocyte-stimulating hormone receptor gene are associated with red hair and fair skin in humans.Nat Genet, 113328330
  90. 90. VillanuevaJ.VulturA.HerlynM.2010Acquired resistance to BRAF inhibitors mediated by a RAF kinase switch in melanoma can be overcome by cotargeting MEK and IGF-1R/PI3K. Cancer Cell, 186683695
  91. 91. WagleN.EmeryC.GarrawayL. A.2011Dissecting Therapeutic Resistance to RAF Inhibition in Melanoma by Tumor Genomic ProfilingJ Clin Oncol
  92. 92. WellbrockC.HurlstoneA.2010BRAF as therapeutic target in melanoma. Biochem Pharmacol, 805561567
  93. 93. WilliamsP. F.OlsenC. M.WhitemanD. C.2010Melanocortin- 1 -receptor and risk of cutaneous melanoma: A meta-analysis and estimates of population burdenInt J Cancer
  94. 94. YancovitzM.YoonJ.PolskyD.2007Detection of mutant BRAF alleles in the plasma of patients with metastatic melanoma. J Mol Diagn, 92178183
  95. 95. YoungA.LyonsJ.Mc CormickF.2009Ras signaling and therapies. Adv Cancer Res, 102117

Written By

Viviana Vallacchi, Licia Rivoltini and Monica Rodolfo

Submitted: 10 November 2010 Published: 12 September 2011