Routine multiplex mutational profiling of melanomas enables enrollment in genotype-driven therapeutic trials

Christine M Lovly, Kimberly Brown Dahlman, Laurel E Fohn, Zengliu Su, Dora Dias-Santagata, Donna J Hicks, Donald Hucks, Elizabeth Berry, Charles Terry, MarKeesa Duke, Yingjun Su, Tammy Sobolik-Delmaire, Ann Richmond, Mark C Kelley, Cindy L Vnencak-Jones, A John Iafrate, Jeffrey Sosman, William Pao, Christine M Lovly, Kimberly Brown Dahlman, Laurel E Fohn, Zengliu Su, Dora Dias-Santagata, Donna J Hicks, Donald Hucks, Elizabeth Berry, Charles Terry, MarKeesa Duke, Yingjun Su, Tammy Sobolik-Delmaire, Ann Richmond, Mark C Kelley, Cindy L Vnencak-Jones, A John Iafrate, Jeffrey Sosman, William Pao

Abstract

Purpose: Knowledge of tumor mutation status is becoming increasingly important for the treatment of cancer, as mutation-specific inhibitors are being developed for clinical use that target only sub-populations of patients with particular tumor genotypes. Melanoma provides a recent example of this paradigm. We report here development, validation, and implementation of an assay designed to simultaneously detect 43 common somatic point mutations in 6 genes (BRAF, NRAS, KIT, GNAQ, GNA11, and CTNNB1) potentially relevant to existing and emerging targeted therapies specifically in melanoma.

Methods: The test utilizes the SNaPshot method (multiplex PCR, multiplex primer extension, and capillary electrophoresis) and can be performed rapidly with high sensitivity (requiring 5-10% mutant allele frequency) and minimal amounts of DNA (10-20 nanograms). The assay was validated using cell lines, fresh-frozen tissue, and formalin-fixed paraffin embedded tissue. Clinical characteristics and the impact on clinical trial enrollment were then assessed for the first 150 melanoma patients whose tumors were genotyped in the Vanderbilt molecular diagnostics lab.

Results: Directing this test to a single disease, 90 of 150 (60%) melanomas from sites throughout the body harbored a mutation tested, including 57, 23, 6, 3, and 2 mutations in BRAF, NRAS, GNAQ, KIT, and CTNNB1, respectively. Among BRAF V600 mutations, 79%, 12%, 5%, and 4% were V600E, V600K, V600R, and V600M, respectively. 23 of 54 (43%) patients with mutation harboring metastatic disease were subsequently enrolled in genotype-driven trials.

Conclusion: We present development of a simple mutational profiling screen for clinically relevant mutations in melanoma. Adoption of this genetically-informed approach to the treatment of melanoma has already had an impact on clinical trial enrollment and prioritization of therapy for patients with the disease.

Conflict of interest statement

Competing Interests: WP: Consulting for Bristol-Myers Squibb; funding from AstraZeneca. DDS and AJI submitted a patent application for the SNaPshot genotyping methods described, which are the subject of licensing discussions. This does not alter the authors' adherence to all the PLoS ONE policies on sharing data and materials.

Figures

Figure 1. Melanoma SNaPshot screen (v1.0).
Figure 1. Melanoma SNaPshot screen (v1.0).
A, five multiplexed panels can detect the mutational status of twenty gene loci. Each peak color represents a particular nucleotide at that locus. The gene name, amino acid, and nucleotide are labeled above each peak. An “(R)” after the nucleotide denotes a reverse extension primer. B, pan-positive control for melanoma SNaPshot screen. Peaks are labeled as described in A. C, SNaPshot sensitivity measurement using cell line DNA carrying known mutations. Numbers indicate the arbitrary fluorescence units of WT (panel 1: green, panels 2, 3: blue) and mutant (panel 1: blue, panels 2, 3: green) peaks. Solid arrows indicate mutant peaks and dotted arrows show background peaks. Background peaks in the negative controls (far right panel) are indicated by their peak height and a star (*).
Figure 2. Distribution of mutations in the…
Figure 2. Distribution of mutations in the first 150 tumors genotyped in the molecular diagnostic lab.
Left: distribution of all mutations. Right: distribution of V600 mutations. See Table S10 for more details.

References

    1. Siegel R, Ward E, Brawley O, Jemal A. Cancer statistics, 2011: the impact of eliminating socioeconomic and racial disparities on premature cancer deaths. CA Cancer J Clin. 2011;61:212–236.
    1. Davies H, Bignell GR, Cox C, Stephens P, Edkins S, et al. Mutations of the BRAF gene in human cancer. Nature. 2002;417:949–954.
    1. Curtin JA, Busam K, Pinkel D, Bastian BC. Somatic activation of KIT in distinct subtypes of melanoma. J Clin Oncol. 2006;24:4340–4346.
    1. Curtin JA, Fridlyand J, Kageshita T, Patel HN, Busam KJ, et al. Distinct sets of genetic alterations in melanoma. N Engl J Med. 2005;353:2135–2147.
    1. Fisher DE, Barnhill R, Hodi FS, Herlyn M, Merlino G, et al. Melanoma from bench to bedside: meeting report from the 6th international melanoma congress. Pigment Cell Melanoma Res. 2010;23:14–26.
    1. Van Raamsdonk CD, Bezrookove V, Green G, Bauer J, Gaugler L, et al. Frequent somatic mutations of GNAQ in uveal melanoma and blue naevi. Nature. 2009;457:599–602.
    1. Van Raamsdonk CD, Griewank KG, Crosby MB, Garrido MC, Vemula S, et al. Mutations in GNA11 in uveal melanoma. N Engl J Med. 2010;363:2191–2199.
    1. Delmas V, Beermann F, Martinozzi S, Carreira S, Ackermann J, et al. Beta-catenin induces immortalization of melanocytes by suppressing p16INK4a expression and cooperates with N-Ras in melanoma development. Genes Dev. 2007;21:2923–2935.
    1. Flaherty KT, Puzanov I, Kim KB, Ribas A, McArthur GA, et al. Inhibition of mutated, activated BRAF in metastatic melanoma. N Engl J Med. 2010;363:809–819.
    1. Sosman J, Kim K, Schuchter L, Gonzalez R, Pavlick A, et al. An open-label, multicenter Phase II study of continuous oral dosing of RG7204 (PLX4032) in previously treated patients with BRAF V600E mutation-positive metastatic melanoma. Pigment Cell Melanoma Res. 2010;23:Abs. 30.
    1. Kefford R, Arkenau H, Brown MP, Millward M, Infante JR, et al. 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. 2010;28:abstr 8503.
    1. Terheyden P, Houben R, Pajouh P, Thorns C, Zillikens D, et al. Response to imatinib mesylate depends on the presence of the V559A-mutated KIT oncogene. J Invest Dermatol. 2010;130:314–316.
    1. Handolias D, Hamilton AL, Salemi R, Tan A, Moodie K, et al. Clinical responses observed with imatinib or sorafenib in melanoma patients expressing mutations in KIT. Br J Cancer. 2010;102:1219–1223.
    1. Antonescu CR, Busam KJ, Francone TD, Wong GC, Guo T, et al. L576P KIT mutation in anal melanomas correlates with KIT protein expression and is sensitive to specific kinase inhibition. Int J Cancer. 2007;121:257–264.
    1. Lutzky J, Bauer J, Bastian BC. Dose-dependent, complete response to imatinib of a metastatic mucosal melanoma with a K642E KIT mutation. Pigment Cell Melanoma Res. 2008;21:492–493.
    1. Hodi FS, Friedlander P, Corless CL, Heinrich MC, Mac Rae S, et al. Major response to imatinib mesylate in KIT-mutated melanoma. J Clin Oncol. 2008;26:2046–2051.
    1. Jiang X, Zhou J, Yuen NK, Corless CL, Heinrich MC, et al. Imatinib targeting of KIT-mutant oncoprotein in melanoma. Clin Cancer Res. 2008;14:7726–7732.
    1. Carvajal RD, Chapman PB, Wolchok JD, Cane L, Teitcher JB, et al. A phase II study of imatinib mesylate (IM) for patients with advanced melanma harboring somatic alterations of KIT. J Clin Oncol. 2009;27:abstr 9001.
    1. Guo J, Si L, Kong Y, Flaherty KT, Xu X, et al. Phase II, Open-Label, Single-Arm Trial of Imatinib Mesylate in Patients With Metastatic Melanoma Harboring c-Kit Mutation or Amplification. J Clin Oncol 2011
    1. Carvajal RD, Antonescu CR, Wolchok JD, Chapman PB, Roman RA, et al. KIT as a therapeutic target in metastatic melanoma. JAMA. 2011;305:2327–2334.
    1. Jaiswal BS, Janakiraman V, Kljavin NM, Eastham-Anderson J, Cupp JE, et al. Combined targeting of BRAF and CRAF or BRAF and PI3K effector pathways is required for efficacy in NRAS mutant tumors. PLoS One. 2009;4:e5717.
    1. Chattopadhyay C, Ellerhorst JA, Ekmekcioglu S, Greene VR, Davies MA, et al. Association of activated c-Met with NRAS-mutated human melanomas: A possible avenue for targeting. Int J Cancer 2011
    1. Dias-Santagata D, Akhavanfard S, David SS, Vernovsky K, Kuhlmann G, et al. Rapid targeted mutational analysis of human tumours: a clinical platform to guide personalized cancer medicine. EMBO Mol Med. 2010;2:146–158.
    1. Su Z, Dias-Santagata D, Duke M, Hutchinson K, Lin YL, et al. A platform for rapid detection of multiple oncogenic mutations with relevance to targeted therapy in non-small-cell lung cancer. J Mol Diagn. 2011;13:74–84.
    1. Sequist LV, Heist RS, Shaw AT, Fidias P, Rosovsky R, et al. Implementing multiplexed genotyping of non-small-cell lung cancers into routine clinical practice. Ann Oncol. 2011;22:2616–2624.
    1. Spittle C, Ward MR, Nathanson KL, Gimotty PA, Rappaport E, et al. Application of a BRAF pyrosequencing assay for mutation detection and copy number analysis in malignant melanoma. J Mol Diagn. 2007;9:464–471.
    1. Xing F, Persaud Y, Pratilas CA, Taylor BS, Janakiraman M, et al. Concurrent loss of the PTEN and RB1 tumor suppressors attenuates RAF dependence in melanomas harboring (V600E)BRAF. Oncogene 2011
    1. McGuinness C, Wesley UV. Dipeptidyl peptidase IV (DPPIV), a candidate tumor suppressor gene in melanomas is silenced by promoter methylation. Front Biosci. 2008;13:2435–2443.
    1. Zuidervaart W, van der Velden PA, Hurks MH, van Nieuwpoort FA, Out-Luiting CJ, et al. Gene expression profiling identifies tumour markers potentially playing a role in uveal melanoma development. Br J Cancer. 2003;89:1914–1919.
    1. Levy C, Khaled M, Iliopoulos D, Janas MM, Schubert S, et al. Intronic miR-211 assumes the tumor suppressive function of its host gene in melanoma. Mol Cell. 2010;40:841–849.
    1. Tsao H, Goel V, Wu H, Yang G, Haluska FG. Genetic interaction between NRAS and BRAF mutations and PTEN/MMAC1 inactivation in melanoma. J Invest Dermatol. 2004;122:337–341.
    1. Woodman SE, Trent JC, Stemke-Hale K, Lazar AJ, Pricl S, et al. Activity of dasatinib against L576P KIT mutant melanoma: molecular, cellular, and clinical correlates. Mol Cancer Ther. 2009;8:2079–2085.
    1. Yang H, Higgins B, Kolinsky K, Packman K, Go Z, et al. RG7204 (PLX4032), a selective BRAFV600E inhibitor, displays potent antitumor activity in preclinical melanoma models. Cancer Res. 2010;70:5518–5527.
    1. Maldonado JL, Fridlyand J, Patel H, Jain AN, Busam K, et al. Determinants of BRAF mutations in primary melanomas. J Natl Cancer Inst. 2003;95:1878–1890.
    1. Chapman PB, Einhorn LH, Meyers ML, Saxman S, Destro AN, et al. Phase III multicenter randomized trial of the Dartmouth regimen versus dacarbazine in patients with metastatic melanoma. J Clin Oncol. 1999;17:2745–2751.
    1. Atkins MB, Hsu J, Lee S, Cohen GI, Flaherty LE, et al. Phase III trial comparing concurrent biochemotherapy with cisplatin, vinblastine, dacarbazine, interleukin-2, and interferon alfa-2b with cisplatin, vinblastine, and dacarbazine alone in patients with metastatic malignant melanoma (E3695): a trial coordinated by the Eastern Cooperative Oncology Group. J Clin Oncol. 2008;26:5748–5754.
    1. Middleton MR, Grob JJ, Aaronson N, Fierlbeck G, Tilgen W, et al. Randomized phase III study of temozolomide versus dacarbazine in the treatment of patients with advanced metastatic malignant melanoma. J Clin Oncol. 2000;18:158–166.
    1. Falkson CI, Ibrahim J, Kirkwood JM, Coates AS, Atkins MB, et al. Phase III trial of dacarbazine versus dacarbazine with interferon alpha-2b versus dacarbazine with tamoxifen versus dacarbazine with interferon alpha-2b and tamoxifen in patients with metastatic malignant melanoma: an Eastern Cooperative Oncology Group study. J Clin Oncol. 1998;16:1743–1751.
    1. Chapman PB, Hauschild A, Robert C, Haanen JB, Ascierto P, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med. 2011;364:2507–2516.
    1. Sosman J, Kim KB, Schuchter L, Gonzalez R, Pavlick AC, et al. Long-term Survival in Vemurafenib-Treated BRAFV600-mutant Advanced Melanoma. New England Journal of Medicine. 2012 In press.
    1. Long GV, Menzies AM, Nagrial AM, Haydu LE, Hamilton AL, et al. Prognostic and Clinicopathologic Associations of Oncogenic BRAF in Metastatic Melanoma. J Clin Oncol. 2011;29:1239–1246.
    1. Nazarian R, Shi H, Wang Q, Kong X, Koya RC, et al. Melanomas acquire resistance to B-RAF(V600E) inhibition by RTK or N-RAS upregulation. Nature. 2010;468:973–977.
    1. Andre F, Delaloge S, Soria JC. Biology-Driven Phase II Trials: What Is the Optimal Model for Molecular Selection? J Clin Oncol. 2011:1236–1238.
    1. King AJ, Patrick DR, Batorsky RS, Ho ML, Do HT, et al. Demonstration of a genetic therapeutic index for tumors expressing oncogenic BRAF by the kinase inhibitor SB-590885. Cancer Res. 2006;66:11100–11105.
    1. Tsai J, Lee JT, Wang W, Zhang J, Cho H, et al. Discovery of a selective inhibitor of oncogenic B-Raf kinase with potent antimelanoma activity. Proc Natl Acad Sci U S A. 2008;105:3041–3046.
    1. Rubinstein JC, Sznol M, Pavlick AC, Ariyan S, Cheng E, et al. Incidence of the V600K mutation among melanoma patients with BRAF mutations, and potential therapeutic response to the specific BRAF inhibitor PLX4032. J Transl Med. 2010;8:67.
    1. Infante JR, Fecher LA, Nallapareddy S, Gordon MS, Flaherty K, et al. Safety and efficacy results from the first-in-human study of the oral MEK 1/2 inhibitor GSK1120212. J Clin Oncol. 2010;28:abstr 2503.
    1. Beadling C, Jacobson-Dunlop E, Hodi FS, Le C, Warrick A, et al. KIT gene mutations and copy number in melanoma subtypes. Clin Cancer Res. 2008;14:6821–6828.
    1. Poulikakos PI, Zhang C, Bollag G, Shokat KM, Rosen N. RAF inhibitors transactivate RAF dimers and ERK signalling in cells with wild-type BRAF. Nature. 2010;464:427–430.
    1. Hatzivassiliou G, Song K, Yen I, Brandhuber BJ, Anderson DJ, et al. RAF inhibitors prime wild-type RAF to activate the MAPK pathway and enhance growth. Nature. 2010;464:431–435.
    1. Heidorn SJ, Milagre C, Whittaker S, Nourry A, Niculescu-Duvas I, et al. Kinase-dead BRAF and oncogenic RAS cooperate to drive tumor progression through CRAF. Cell. 2010;140:209–221.
    1. Omholt K, Platz A, Ringborg U, Hansson J. Cytoplasmic and nuclear accumulation of beta-catenin is rarely caused by CTNNB1 exon 3 mutations in cutaneous malignant melanoma. Int J Cancer. 2001;92:839–842.
    1. Reifenberger J, Knobbe CB, Wolter M, Blaschke B, Schulte KW, et al. Molecular genetic analysis of malignant melanomas for aberrations of the WNT signaling pathway genes CTNNB1, APC, ICAT and BTRC. Int J Cancer. 2002;100:549–556.
    1. Demunter A, Libbrecht L, Degreef H, De Wolf-Peeters C, van den Oord JJ. Loss of membranous expression of beta-catenin is associated with tumor progression in cutaneous melanoma and rarely caused by exon 3 mutations. Mod Pathol. 2002;15:454–461.
    1. Bauer J, Kilic E, Vaarwater J, Bastian BC, Garbe C, et al. Oncogenic GNAQ mutations are not correlated with disease-free survival in uveal melanoma. Br J Cancer. 2009;101:813–815.
    1. Dunn EF, Iida M, Myers RA, Campbell DA, Hintz KA, et al. Dasatinib sensitizes KRAS mutant colorectal tumors to cetuximab. Oncogene. 2010;30:561–574.

Source: PubMed

3
Subscribe