Skip to main content
UKPMC Funders Author Manuscripts logoLink to UKPMC Funders Author Manuscripts
. Author manuscript; available in PMC: 2016 Aug 5.
Published in final edited form as: Histopathology. 2013 Feb 12;62(4):543–550. doi: 10.1111/his.12050

Mutation profiling of adenoid cystic carcinomas from multiple anatomical sites identifies mutations in the RAS pathway, but no KIT mutations

Daniel Wetterskog 1, Paul M Wilkerson 1, Daniel N Rodrigues 1, Maryou B Lambros 1, Karen Fritchie 1, Mattias K Andersson 2, Rachael Natrajan 1, Arnaud Gauthier 1, Silvana Di Palma 3, Sami Shousha 4, Zoran Gatalica 5, Chantal Töpfer 1, Vesna Vukovic 1, Roger A’Hern 6, Britta Weigelt 7, Anne Vincent-Salomon 8, Göran Stenman 2, Brian P Rubin 1, Jorge S Reis-Filho 1,*
PMCID: PMC4975515  EMSID: EMS69400  PMID: 23398044

Abstract

Aims

The majority of adenoid cystic carcinomas (AdCCs), regardless of anatomical site, harbour the MYBNFIB fusion gene. The aim of this study was to characterize the repertoire of somatic genetic events affecting known cancer genes in AdCCs.

Methods and results

DNA was extracted from 13 microdissected breast AdCCs, and subjected to a mutation survey using the Sequenom OncoCarta Panel v1.0. Genes found to be mutated in any of the breast AdCCs and genes related to the same canonical molecular pathways, as well as KIT, a proto-oncogene whose protein product is expressed in AdCCs, were sequenced in an additional 68 AdCCs from various anatomical sites by Sanger sequencing. Using the Sequenom MassARRAY platform and Sanger sequencing, mutations in BRAF and HRAS were identified in three and one cases, respectively (breast, and head and neck). KIT, which has previously been reported to be mutated in AdCCs, was also investigated, but no mutations were identified.

Conclusions

Our results demonstrate that mutations in genes pertaining to the canonical RAS pathway are found in a minority of AdCCs, and that activating KIT mutations are either absent or remarkably rare in these cancers, and unlikely to constitute a driver and therapeutic target for patients with AdCC.

Keywords: adenoid cystic carcinoma, BRAF, HRAS, KIT, sequencing

Introduction

Adenoid cystic carcinomas (AdCCs) are malignant tumours that often affect the salivary glands, but can also occur in other anatomical sites, such as the lung and breast.1 Salivary gland AdCC is the most prevalent type of salivary gland carcinoma,2 and presents as a slow-growing but aggressive cancer, frequently resulting in multiple local recurrences and the development of distant metastases. Patients with salivary gland AdCC have a poor long-term outcome.3 In contrast, breast AdCCs show an indolent clinical behaviour, with a 90–100% 10-year survival rate and a low incidence of distant metastases.1,4,5

AdCCs provide a clear example of genotypic–phenotypic correlation, as they have similar morphological characteristics and genetic aberrations, independently of the site of origin. Recent studies have demonstrated that a subgroup of AdCCs are characterized by the presence of a recurrent t(6:9)(q22–23;p23–24) chromosomal translocation.6 This translocation generates a fusion transcript involving the oncogene MYB and the transcription factor gene NFIB. In the resulting fusion gene, the MYB 3′-untranslated region (UTR) is replaced by the NFIB 3′-UTR, leading to loss of microRNA-mediated down-regulation of MYB.6 The prevalence of the fusion gene is reported to range from 30–100% in salivary gland AdCCs69 to 90–100% in breast AdCCs.6,10 Even in AdCCs lacking the MYB–NFIB fusion gene, MYB has been shown to be consistently overexpressed in AdCCs,10 indicating that other mechanisms resulting in MYB overexpression are operational in these tumours. These observations provide circumstantial evidence that MYB is likely to constitute a driver of AdCCs, regardless of the anatomical site.

The genetic features of salivary gland and breast AdCCs have been investigated, and these tumours have been shown to harbour low levels of genetic instability, frequent loss of 6q and 9p, and a low frequency of high-level amplification of cancer-related genes.1013 In salivary glands, amplification of CCND1, MDM2 and CTTN has been reported, but only in up to 5% of cases.11,12 No recurrent amplifications have been reported in breast AdCCs.10,14 On the basis of these observations, it is plausible that mechanisms other than gene copy number aberrations drive the diverse clinical behaviour of AdCCs. In fact, other oncogenic events have been described in AdCCs,15 including mutations of KIT,15,16 PIK3CA, and PTEN.17 Currently, there is no additional information on the prevalence of mutations affecting the components of the phosphoinositide-3-kinase (PI3K) canonical pathway in AdCCs or on the mutational repertoire of breast AdCCs.

The tyrosine kinase receptor c-KIT is expressed in up to 100% of AdCCs of the breast15,18 and salivary glands,19 and has been proposed as a driver and potential therapeutic target in AdCCs.15 The efficacy of therapies targeting c-KIT, in particular of imatinib mesylate, appears to be restricted to patients whose tumours harbour a mutant KIT gene.20 The prevalence of KIT mutations in AdCC has been investigated in several studies,15,16,19,2124 and has proven to be controversial. Although independent investigators have failed to identify KIT mutations in AdCCs,19,2124 two studies reported KIT mutations in 12% and 88% of formalin-fixed paraffin-embedded (FFPE) salivary gland AdCCs.15,16 Although case reports of AdCCs have suggested a benefit from imatinib,25,26 clinical trials assessing imatinib as targeted therapy in patients with AdCC have yielded negative results.22,27 Thus, currently, no specific targeted therapy exists for the treatment of patients with breast or salivary gland AdCCs.

The aims of this study were to investigate whether breast AdCCs harbour point mutations in 19 known cancer-related genes,2832 and to investigate the prevalence of KIT mutations in a series of breast, lung and salivary gland AdCCs.

Materials and methods

Case Selection

A series of 26 fresh-frozen and 65 FFPE primary and metastatic AdCCs of the breast (n = 25), lung (n = 7), head and neck (n = 56), pancreas (n = 1), kidney (n = 1) and vaginal wall (n = 1) were retrieved from the tissue banks of the Royal Marsden Hospital, London, UK, the Cleveland Clinic, Ohio, USA, the Curie Institute, Paris, France, the Royal Surrey County Hospital, Guildford, UK, and the Department of Pathology, Sahlgrenska Cancer Centre, Gothenburg, Sweden (Tables 1 and S1). Cases were anonymized prior to inclusion in this study, and approval by the local ethics committees was obtained. Cases were locally reviewed by the contributing pathologists, and centrally reviewed by two of the authors (D.N.R. and J.S.R.-F.). Only bona fide AdCCs were included in this study.

Table 1.

Summary of tumour samples analysed in this study

Anatomical site Fixation Analysis
Fresh frozen FFPE Sequenom Sanger Not analysed
Breast (n = 25) 10 15 13* 14 0
Head and neck (n = 56) 16 40 0 48 8*
Lung (n = 7) 0 7 0 7 0
Pancreas 0 1 0 0 1
Kidney 0 1 0 0 1
Vaginal wall 0 1 0 1 0

FFPE, formalin-fixed paraffin-embedded.

*

All FFPE samples.

Microdissection and DNA Extraction

To ensure >90% purity of neoplastic cells, microdissection was performed with a sterile needle under a stereomicroscope (Olympus SZ61, Tokyo, Japan) from four to 10 consecutive 8-µm-thick sections stained with nuclear fast red, as previously described.33 DNA was extracted using the DNeasy Blood and Tissue Kit (Qiagen, Crawley, UK), as previously described.33 The double-stranded DNA concentration was measured using the Picogreen assay, according to the manufacturer’s instructions (Invitrogen, Paisley, UK). Ten cases did not yield DNA of sufficient quantity and/or quality, and were excluded from further analyses.

Genotyping With Sequenom Oncocarta

Thirteen AdCCs of the breast were subjected to mutation screening using the OncoCarta Panel v1.0 (Sequenom, San Diego, CA, USA), consisting of 24 pools of polymerase chain reaction (PCR) primer pairs and extension primers with the capacity to interrogate 238 known mutations in 19 cancer-related genes (ABL1, AKT1, AKT2, BRAF, CDK4, EGFR, ERBB2, FGFR1, FGFR3, FLT3, JAK2, KIT, MET, HRAS, KRAS, NRAS, PDGFA, PIK3CA, and RET). In brief, 20 ng of DNA was amplified using the 24 sets of OncoCarta PCR primer pools. Subsequently, a single base extension reaction using the OncoCarta extension primers was performed. After using a cation exchange resin to remove salts, the products were spotted, using the nanodispensing system, on a 384 SpectroChipII, and analysed on a matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometer (Sequenom). Data were interrogated using MassARRAY TYPER analyser software 4. Mutations were manually reviewed by use of the visual and the raw spectrum patterns. The prevalence of mutant alleles was estimated by calculating the ratio of the height of the raw spectrum of the mutant allele to that of its wild-type allelle.

Sanger Sequencing

Mutations with an allelic ratio ≥25%, as defined by the Sequenom MassARRAY assay, were validated in the index cases by Sanger sequencing, where primers were designed using PRIMER3 (http://frodo.wi.mit.edu/primer3/), as previously described.34 Mutations validated in the index cases were tested in an additional 68 AdCC cases (48 head and neck, seven lung, 12 breast, and one vaginal wall) from which DNA of sufficient quantity and quality could be extracted. In addition, the presence of activating mutations affecting codons 12, 13 and 61 of KRAS, HRAS, and NRAS, respectively, and exons 8, 9, 11, 13 and 17 of KIT, were investigated in all 68 cases (primer sequences are given in Table S2). Sequences were analysed with MUTATION SURVEYOR software (Softgenetics LLC., State College, PA, 16803 USA). Sequencing was performed twice for each sample harbouring a potential mutation, to rule out the possibility of PCR fidelity artefacts, and was carried out in both directions.

Results

To identify potential secondary driver events in AdCCs, 13 previously characterized AdCCs of the breast10 were subjected to Sequenom analysis. No mutations in cancer genes previously implicated in breast cancer (Cosmic database, http://www.sanger.ac.uk/genetics/CGP/cosmic/), including AKT1, ERBB2, and PIK3CA, were detected. Interestingly, however, BRAF, a gene reported to be mutated in <1% of invasive breast cancers, was found to be mutated in two breast AdCCs (15%; Figure 1; Table 2). Although both mutations identified, V600E and G464E, are reported to be activating,35,36 the former has been shown to be a high-activity mutant, whereas the latter is an intermediate-activity mutant.35,36 Both mutations were successfully validated by means of Sanger sequencing. In addition, one KIT mutation and one KRAS mutation were identified by Sequenom analysis; however, these mutations could not be validated by Sanger sequencing (data not shown). This may be because of the relatively low allelic frequency of the mutations, making confident verification with Sanger sequencing challenging. Importantly, even after repeated microdissections and alternative Sanger sequencing strategies (e.g. cDNA sequencing; data not shown), these mutations were not validated, and should therefore be interpreted as false-positive results.

Figure 1.

Figure 1

Mutational profiling of adenoid cystic carcinoma (AdCC) of the breast using the Sequenom MassARRAY platform. Sequenom call cluster plots and mass spectrometry profiles for the detection of the (a) BRAF G464E (BRAF_G1391A) and (b) V600E (BRAF_T1799A) mutations are shown, together with the Sanger sequencing validation for each affected case. Call cluster plots show the low and high mass allele ratios on the x-axis and y-axis; wild-type calls are in orange (G464E) or blue (V600E), and mutant calls are in red (circled red). Mass spectrometry profiles show the allelic mass on the x-axis and the intensity on the y-axis. The corresponding sequencing chromatogram for case 7, which harbours a wild-type BRAF gene (i.e. G464 and V600), is shown below the mutant case for each mutation (in black). UEP, unextended primer. The red arrow marks a mutant allele.

Table 2.

Characteristics of tumour samples in which validated mutations were identified in this study

Case Anatomical site Histological pattern Gene Mutation (amino acid) Allelic frequency*
1 Breast Cribriform–solid BRAF G464E 0.25
2 Breast Cribriform–solid BRAF V600E 0.8
46 Parotid NK HRAS Q61K Sanger
91 Breast NK BRAF D594G Sanger

NK, not known; Sanger, mutations identified and validated by Sanger sequencing; hence, their allelic frequency could not be estimated.

*

For mutations identified by Sequenom MassARRAY.

The identified BRAF mutations suggested potential alteration of the RAS pathway in two AdCCs. As other members of the pathway also represent targets of oncogenic mutations, we sequenced the mutational hotspots of codons 12, 13 and 61 of KRAS, HRAS and NRAS in addition to BRAF hotspot mutations (i.e. exons 8, 9, 11, 13 and 17) in an independent series of 68 AdCCs from various sites. Sanger sequencing identified an HRAS Q61K hotspot mutation in a head and neck AdCC, and a BRAF D594G kinase-dead37 hotspot mutation in another breast AdCC (Figure 2). All mutations were confirmed in independent PCRs and sequencing reactions. None of the additional AdCCs investigated harboured mutations in KRAS. The above mutations were all identified in distinct samples (i.e. no tumour sample harboured more than one mutation in the genes tested in this study). Therefore, out of the components of the RAS pathway investigated in this study, ~4% and 1% harboured validated BRAF and HRAS mutations, respectively.

Figure 2.

Figure 2

Mutational analysis of adenoid cystic carcinoma (AdCC) from various anatomical sites by Sanger sequencing. A, The top panel shows the Sanger sequencing chromatogram of the HRAS Q61K (C181A) mutation identified in case 46, a salivary gland AdCC. The bottom panel shows case 51, a HRAS wild-type salivary gland AdCC. B, The top panel shows the Sanger sequencing chromatogram of the BRAF D594G (A1781G) mutation identified in case 91, a breast AdCC. The bottom panel shows case 90, a BRAF wild-type breast AdCC. Red arrows mark the position of the base change.

Because of the controversy surrounding the existence of activating KIT mutations in AdCCs, and their potential clinical implications, we sequenced exons 8, 9, 11, 13 and 17 of KIT. None of the AdCCs analysed here harboured a mutation in KIT. Power calculations revealed that, if the true prevalence of KIT mutations was ≥4% (relative to an expected prevalence of 15%), the probability of a negative result (i.e. no mutations found in 81 samples) by chance would be <10−4, which equates to a statistical power of >95%. Therefore, a negative result is unlikely to be attributable to a type II or β error.

Discussion

AdCCs from different sites show similar patterns of genomic alterations. Here, we characterized the mutation status of known cancer genes in a large series of breast AdCCs, and selected cancer genes in a large series of AdCCs from various anatomical sites. Our results demonstrate that mutations affecting the RAS pathway are found in a subset of AdCCs. In addition, our results show that KIT mutations are either absent or vanishingly rare in AdCCs, suggesting that they are unlikely to constitute clinically useful therapeutic targets for this disease.

Dysregulated signalling of the RAS pathway has a major role in the development of human cancers, and activating hotspot mutations have been identified in several members of the pathway.38,39 The hotspot mutations of KRAS, HRAS or NRAS occur at codon 12, 13 or 61, and cause them to become constitutively active. Mutations of KRAS have been previously described in AdCCs of the salivary gland, with a prevalence of 4%.40 Our study confirms this observation, and provides the novel observation that other components of the RAS pathway are also mutated in a small subset of AdCCs. Likewise, two of the BRAF mutations identified in AdCCs in this study (i.e. V600E and G464E) also result in an increase in kinase activity;35,36 however, the BRAF D594G mutation found is a kinase-dead mutation37,41 that results in reduction/abrogation of the kinase activity of BRAF. Activating BRAF mutations are found in wide range of cancers, including melanomas, and ovarian, lung and colorectal cancer.36 Targeted therapy against mutated BRAF V600E exists in the form of PLX403242 (vemurafenib; Plexxikon/Roche, Berkeley, CA, USA), and high PLX4032 response rates in patients with BRAF V600E mutant melanoma have been observed.43 Kinase-dead BRAF mutations, on the other hand, are rarer. The BRAF D594G mutation does not lead to phosphorylation of MEK, activation of CRAF, or stimulation of cell signalling.35,37 Importantly, however, in in-vitro and animal model studies,44 kinase-dead BRAF mutations such as D594G may result in paradoxical and strong activation of the MEK pathway if the pathway is activated by an event upstream of BRAF.44 In the case of AdCCs, one could hypothesize that c-KIT activation could result in strong activation of the extracellular signal-related kinase pathway in the presence of the kinase-dead BRAF D594G mutation. It should be noted that, in the context of kinase-dead BRAF mutations, BRAF-specific inhibitors are unlikely to be effective; however, on the basis of preclinical models of melanoma, it is plausible that MEK inhibitors may potentially be of clinical benefit.44

Although we demonstrated here that BRAF was recurrently mutated in breast AdCCs (~4%), and that HRAS was mutated in another head and neck AdCC, the mutations identified in this study account for only 5% of the AdCCs assessed. Recently, potential novel driver mutations in the mitogen-activated protein kinase (MAPK) family other than RAS and BRAF have been described, including MAP2K4 and MAP3K1.45 Further studies to determine the mutation status of MAP2K4 and MAP3K1 in AdCCs are warranted. Functional studies are also required to determine whether mutations of genes in the RAS pathway act as oncogenic drivers in AdCCs. Critically, however, clinical trials testing MEK-specific or BRAF-specific inhibitors in patients with AdCC will require careful patient selection and the development of robust biomarkers.

Mutations in additional oncogenes have been described in AdCCs. A previous study suggested that AdCCs may harbour mutations in the components of the PI3K pathway, and a PTEN mutation and a PIK3CA mutation (in exon 1, 169A>G, I31M) were described in one case of AdCC from the breast and the renal metastasis of the same patient;17 however, no PIK3CA mutations were identified in any of the breast AdCCs analysed by Sequenom analysis, and the functional effects of the PIK3CA I31M mutation have not been reported. The presence of activating KIT mutations in AdCCs has proven controversial. Although multiple studies19,2124 have suggested that AdCCs do not harbour activating KIT mutations, Vila et al.15 and Tetsu et al.16 reported mutations affecting this oncogene in 12–88% of AdCCs. Our results do not confirm the observations of Vila et al. and Tetsu et al. In fact, none of the 81 AdCCs tested harboured KIT mutations, suggesting that mutations affecting this proto-oncogene are either nonexistent or vanishingly rare in AdCCs, given that the likelihood of our negative findings in 81 AdCCs being attributable to a type II or β error is remarkably low (P < 10−4). Importantly, it should be noted that, in the studies reporting on the presence of KIT mutations in AdCCs,15,16 FFPE material was analysed, and that 19 of the 21 mutations reported by Vila et al.15 were C>T or A>G transitions. These patterns of single-nucleotide changes have been previously reported as potential artefacts of sequencing DNA extracted from FFPE tumours.46 Taken together, our results and those of previous studies19,2124 suggest that KIT mutations are either non-existent or vanishingly rare in AdCCs, and that imatinib mesylate is unlikely to be a potential targeted therapy for patients with this malignancy, given that this small-molecule inhibitor has proven to be efficacious in patients whose tumours harbour activating KIT mutations.

This study has a number of limitations. First, as samples were accrued from a number of centres across the world, it was not possible to retrieve sufficient clinical annotations to determine whether the mutations identified were from tumours that followed an aggressive clinical course. Second, despite our efforts in retrieving AdCCs from multiple institutions, the statistical power of the study was still limited. Third, given the retrospective nature of our study and the potential biases in sample selection, it should be perceived as hypothesis-generating. Fourth, given the multi-institutional origin of samples, the retrospective nature of the study, the various therapies that the patients received, and the incomplete follow-up information available for the patients whose tumours were included, formal survival analyses could not be performed.

Taken together, our results demonstrate that a minority of AdCCs harbour activating mutations in RAS pathway genes. Further studies investigating whether these mutations act as drivers in AdCCs and could be exploited as potential therapeutic targets for patients with this disease are warranted. Finally, our results and those from previous studies that also failed to identify activating KIT mutations19,2124 in AdCCs from various anatomical sites call into question their very existence in AdCCs, demonstrating that c-KIT small-molecule inhibitors are unlikely to constitute clinically useful targeted therapies for patients with AdCCs.

Supplementary Material

Table S1
Table S2

Acknowledgements

This study was funded, in part, by Breakthrough Breast Cancer, the Swedish Cancer Society, and BioCARE—a National Strategic Research Programme at the University of Gothenburg. P. M. Wilkerson is funded by a Wellcome Trust clinical fellowship grant, and B. Weigelt by a Cancer Research UK postdoctoral fellowship. We acknowledge NHS funding to the NIHR Biomedical Research Centre. The study sponsors had no involvement in the design of this study, the literature review, data interpretation, writing of the manuscript, or the decision to submit it for publication.

Footnotes

Conflict of interest

The authors declare no conflict of interest.

References

  • 1.Marchio C, Weigelt B, Reis-Filho JS. Adenoid cystic carcinomas of the breast and salivary glands (or ‘the strange case of Dr jekyll and Mr hyde’ of exocrine gland carcinomas) J Clin Pathol. 2010;63:220–228. doi: 10.1136/jcp.2009.073908. [DOI] [PubMed] [Google Scholar]
  • 2.Fletcher C, Cheuk W, Chan J. Salivary gland tumors. In: Fletcher C, editor. Diagnostic histopathology of tumours. Churchill Livingstone Elsevier; New York: 2007. pp. 280–284. [Google Scholar]
  • 3.Wahlberg P, Anderson H, Biorklund A, Moller T, Perfekt R. Carcinoma of the parotid and submandibular glands—a study of survival in 2465 patients. Oral Oncol. 2002;38:706–713. doi: 10.1016/s1368-8375(02)00007-6. [DOI] [PubMed] [Google Scholar]
  • 4.Arpino G, Clark GM, Mohsin S, Bardou VJ, Elledge RM. Adenoid cystic carcinoma of the breast: molecular markers, treatment, and clinical outcome. Cancer. 2002;94:2119–2127. doi: 10.1002/cncr.10455. [DOI] [PubMed] [Google Scholar]
  • 5.Bennett AK, Mills SE, Wick MR. Salivary-type neoplasms of the breast and lung. Semin Diagn Pathol. 2003;20:279–304. doi: 10.1053/j.semdp.2003.09.001. [DOI] [PubMed] [Google Scholar]
  • 6.Persson M, Andren Y, Mark J, Horlings HM, Persson F, Stenman G. Recurrent fusion of MYB and NFIB transcription factor genes in carcinomas of the breast and head and neck. Proc Natl Acad Sci USA. 2009;106:18740–18744. doi: 10.1073/pnas.0909114106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Mitani Y, Li J, Rao PH, et al. Comprehensive analysis of the MYB–NFIB gene fusion in salivary adenoid cystic carcinoma: incidence, variability, and clinicopathologic significance. Clin Cancer Res. 2010;16:4722–4731. doi: 10.1158/1078-0432.CCR-10-0463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Brill LB, 2nd, Kanner WA, Fehr A, et al. Analysis of MYB expression and MYB–NFIB gene fusions in adenoid cystic carcinoma and other salivary neoplasms. Mod Pathol. 2011;24:1169–1176. doi: 10.1038/modpathol.2011.86. [DOI] [PubMed] [Google Scholar]
  • 9.West RB, Kong C, Clarke N, et al. MYB expression and translocation in adenoid cystic carcinomas and other salivary gland tumors with clinicopathologic correlation. Am J Surg Pathol. 2011;35:92–99. doi: 10.1097/PAS.0b013e3182002777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Wetterskog D, Lopez-Garcia MA, Lambros MB, et al. Adenoid cystic carcinomas constitute a genomically distinct subgroup of triple-negative and basal-like breast cancers. 2012;226:84–96. doi: 10.1002/path.2974. [DOI] [PubMed] [Google Scholar]
  • 11.Greer RO, Jr, Said S, Shroyer KR, Marileila VG, Weed SA. Overexpression of cyclin D1 and cortactin is primarily independent of gene amplification in salivary gland adenoid cystic carcinoma. Oral Oncol. 2007;43:735–741. doi: 10.1016/j.oraloncology.2006.09.007. [DOI] [PubMed] [Google Scholar]
  • 12.Bernheim A, Toujani S, Saulnier P, et al. High-resolution array comparative genomic hybridization analysis of human bronchial and salivary adenoid cystic carcinoma. Lab Invest. 2008;88:464–473. doi: 10.1038/labinvest.2008.18. [DOI] [PubMed] [Google Scholar]
  • 13.Persson M, Andren Y, Moskaluk CA, et al. Clinically significant copy number alterations and complex rearrangements of MYB and NFIB in head and neck adenoid cystic carcinoma. Genes Chromosomes Cancer. 2012;51:805–817. doi: 10.1002/gcc.21965. [DOI] [PubMed] [Google Scholar]
  • 14.Sequeiros-Santiago G, Garcia-Carracedo D, Fresno MF, Suarez C, Rodrigo JP, Gonzalez MV. Oncogene amplification pattern in adenoid cystic carcinoma of the salivary glands. Oncol Rep. 2009;21:1215–1222. doi: 10.3892/or_00000344. [DOI] [PubMed] [Google Scholar]
  • 15.Vila L, Liu H, Al-Quran SZ, Coco DP, Dong HJ, Liu C. Identification of c-kit gene mutations in primary adenoid cystic carcinoma of the salivary gland. Mod Pathol. 2009;22:1296–1302. doi: 10.1038/modpathol.2009.95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Tetsu O, Phuchareon J, Chou A, Cox DP, Eisele DW, Jordan RC. Mutations in the c-kit gene disrupt mitogen-activated protein kinase signaling during tumor development in adenoid cystic carcinoma of the salivary glands. Neoplasia. 2010;12:708–717. doi: 10.1593/neo.10356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Vranic S, Bilalovic N, Lee LM, Kruslin B, Lilleberg SL, Gatalica Z. Pik3ca and PTEN mutations in adenoid cystic carcinoma of the breast metastatic to kidney. Hum Pathol. 2007;38:1425–1431. doi: 10.1016/j.humpath.2007.03.021. [DOI] [PubMed] [Google Scholar]
  • 18.Weigelt B, Horlings HM, Kreike B, et al. Refinement of breast cancer classification by molecular characterization of histological special types. J Pathol. 2008;216:141–150. doi: 10.1002/path.2407. [DOI] [PubMed] [Google Scholar]
  • 19.Holst VA, Marshall CE, Moskaluk CA, Frierson HF., Jr KIT protein expression and analysis of c-kit gene mutation in adenoid cystic carcinoma. Mod Pathol. 1999;12:956–960. [PubMed] [Google Scholar]
  • 20.Heinrich MC, Griffith DJ, Druker BJ, Wait CL, Ott KA, Zigler AJ. Inhibition of c-kit receptor tyrosine kinase activity by STI 571, a selective tyrosine kinase inhibitor. Blood. 2000;96:925–932. [PubMed] [Google Scholar]
  • 21.Jeng YM, Lin CY, Hsu HC. Expression of the c-kit protein is associated with certain subtypes of salivary gland carcinoma. Cancer Lett. 2000;154:107–111. doi: 10.1016/s0304-3835(00)00387-6. [DOI] [PubMed] [Google Scholar]
  • 22.Lin CH, Yen RF, Jeng YM, Tzen CY, Hsu C, Hong RL. Unexpected rapid progression of metastatic adenoid cystic carcinoma during treatment with imatinib mesylate. Head Neck. 2005;27:1022–1027. doi: 10.1002/hed.20274. [DOI] [PubMed] [Google Scholar]
  • 23.Pan CC, Chen PC, Chiang H. Kit (CD117) is frequently overexpressed in thymic carcinomas but is absent in thymomas. J Pathol. 2004;202:375–381. doi: 10.1002/path.1514. [DOI] [PubMed] [Google Scholar]
  • 24.Aubry MC, Heinrich MC, Molina J, et al. Primary adenoid cystic carcinoma of the lung: absence of KIT mutations. Cancer. 2007;110:2507–2510. doi: 10.1002/cncr.23075. [DOI] [PubMed] [Google Scholar]
  • 25.Faivre S, Raymond E, Casiraghi O, Temam S, Berthaud P. Imatinib mesylate can induce objective response in progressing, highly expressing kit adenoid cystic carcinoma of the salivary glands. J Clin Oncol. 2005;23:6271–6273. doi: 10.1200/JCO.2005.01.6055. author reply 6273–6274. [DOI] [PubMed] [Google Scholar]
  • 26.Alcedo JC, Fabrega JM, Arosemena JR, Urrutia A. Imatinib mesylate as treatment for adenoid cystic carcinoma of the salivary glands: report of two successfully treated cases. Head Neck. 2004;26:829–831. doi: 10.1002/hed.20094. [DOI] [PubMed] [Google Scholar]
  • 27.Hotte SJ, Winquist EW, Lamont E, et al. Imatinib mesylate in patients with adenoid cystic cancers of the salivary glands expressing c-kit: a Princess Margaret Hospital phase II consortium study. J Clin Oncol. 2005;23:585–590. doi: 10.1200/JCO.2005.06.125. [DOI] [PubMed] [Google Scholar]
  • 28.Falzoi M, Mossa A, Congeddu E, Saba L, Pani L. Multiplex genotyping of CYP3a4, CYP3a5, CYP2c9 and CYP2c19 SNPs using MALDI-TOF mass spectrometry. Pharmacogenomics. 2010;11:559–571. doi: 10.2217/pgs.09.172. [DOI] [PubMed] [Google Scholar]
  • 29.Humeny A, Rodel F, Rodel C, et al. Mdr1 single nucleotide polymorphism c3435t in normal colorectal tissue and colorectal carcinomas detected by MALDI-TOF mass spectrometry. Anticancer Res. 2003;23:2735–2740. [PubMed] [Google Scholar]
  • 30.Thomas RK, Nickerson E, Simons JF, et al. Sensitive mutation detection in heterogeneous cancer specimens by massively parallel picoliter reactor sequencing. Nat Med. 2006;12:852–855. doi: 10.1038/nm1437. [DOI] [PubMed] [Google Scholar]
  • 31.Fumagalli D, Gavin PG, Taniyama Y, et al. A rapid, sensitive, reproducible and cost-effective method for mutation profiling of colon cancer and metastatic lymph nodes. BMC Cancer. 2010;10:101. doi: 10.1186/1471-2407-10-101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.MacConaill LE, Campbell CD, Kehoe SM, et al. Profiling critical cancer gene mutations in clinical tumor samples. PLoS One. 2009;4:e7887. doi: 10.1371/journal.pone.0007887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Lacroix-Triki M, Suarez PH, MacKay A, et al. Mucinous carcinoma of the breast is genomically distinct from invasive ductal carcinomas of no special type. J Pathol. 2010;222:282–298. doi: 10.1002/path.2763. [DOI] [PubMed] [Google Scholar]
  • 34.Geyer FC, Lacroix-Triki M, Savage K, et al. Beta-catenin pathway activation in breast cancer is associated with triple-negative phenotype but not with CTNNB1 mutation. Mod Pathol. 2011;24:209–231. doi: 10.1038/modpathol.2010.205. [DOI] [PubMed] [Google Scholar]
  • 35.Wan PT, Garnett MJ, Roe SM, et al. Mechanism of activation of the RAF–ERK signaling pathway by oncogenic mutations of B-RAF. Cell. 2004;116:855–867. doi: 10.1016/s0092-8674(04)00215-6. [DOI] [PubMed] [Google Scholar]
  • 36.Davies H, Bignell GR, Cox C, et al. Mutations of the BRAF gene in human cancer. Nature. 2002;417:949–954. doi: 10.1038/nature00766. [DOI] [PubMed] [Google Scholar]
  • 37.Ikenoue T, Hikiba Y, Kanai F, et al. Functional analysis of mutations within the kinase activation segment of B-RAF in human colorectal tumors. Cancer Res. 2003;63:8132–8137. [PubMed] [Google Scholar]
  • 38.Downward J. Targeting RAS signalling pathways in cancer therapy. Nat Rev Cancer. 2003;3:11–22. doi: 10.1038/nrc969. [DOI] [PubMed] [Google Scholar]
  • 39.Maurer G, Tarkowski B, Baccarini M. RAF kinases in cancer—roles and therapeutic opportunities. Oncogene. 2011;30:3477–3488. doi: 10.1038/onc.2011.160. [DOI] [PubMed] [Google Scholar]
  • 40.Dahse R, Driemel O, Schwarz S, Kromeyer-Hauschild K, Berndt A, Kosmehl H. KRAS status and epidermal growth factor receptor expression as determinants for anti-EGFR therapies in salivary gland carcinomas. Oral Oncol. 2009;45:826–829. doi: 10.1016/j.oraloncology.2009.01.013. [DOI] [PubMed] [Google Scholar]
  • 41.Ritt DA, Monson DM, Specht SI, Morrison DK. Impact of feedback phosphorylation and RAF heterodimerization on normal and mutant B-RAF signaling. Mol Cell Biol. 2010;30:806–819. doi: 10.1128/MCB.00569-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Joseph EW, Pratilas CA, Poulikakos PI, et al. The RAF inhibitor PLX4032 inhibits ERK signaling and tumor cell proliferation in a V600E BRAF-selective manner. Proc Natl Acad Sci USA. 2010;107:14903–14908. doi: 10.1073/pnas.1008990107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Bollag G, Hirth P, Tsai J, et al. Clinical efficacy of a RAF inhibitor needs broad target blockade in BRAF-mutant melanoma. Nature. 2010;467:596–599. doi: 10.1038/nature09454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Heidorn SJ, Milagre C, Whittaker S, et al. Kinase-dead BRAF and oncogenic RAS cooperate to drive tumor progression through CRAF. Cell. 2010;140:209–221. doi: 10.1016/j.cell.2009.12.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Stephens PJ, Tarpey PS, Davies H, et al. The landscape of cancer genes and mutational processes in breast cancer. Nature. 2012;486:400–404. doi: 10.1038/nature11017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Do H, Dobrovic A. Limited copy number-high resolution melting (LCN-HRM) enables the detection and identification by sequencing of low level mutations in cancer biopsies. Mol Cancer. 2009;8:82. doi: 10.1186/1476-4598-8-82. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Table S1
Table S2

RESOURCES