Rare Variant, Gene-Based Association Study of Hereditary Melanoma Using Whole-Exome Sequencing

Extraordinary progress has been made in our understanding of common variants in many diseases, including melanoma. Because the contribution of rare coding variants is not as well characterized, we performed an exome-wide, gene-based association study of familial cutaneous melanoma (CM) and ocular me...

Full description

Saved in:
Bibliographic Details
Published inJNCI : Journal of the National Cancer Institute Vol. 109; no. 12
Main Authors Artomov, Mykyta, Stratigos, Alexander J, Kim, Ivana, Kumar, Raj, Lauss, Martin, Reddy, Bobby Y, Miao, Benchun, Daniela Robles-Espinoza, Carla, Sankar, Aravind, Njauw, Ching-Ni, Shannon, Kristen, Gragoudas, Evangelos S, Marie Lane, Anne, Iyer, Vivek, Newton-Bishop, Julia A, Timothy Bishop, D, Holland, Elizabeth A, Mann, Graham J, Singh, Tarjinder, Daly, Mark J, Tsao, Hensin
Format Journal Article
LanguageEnglish
Published United States Oxford University Press 01.12.2017
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:Extraordinary progress has been made in our understanding of common variants in many diseases, including melanoma. Because the contribution of rare coding variants is not as well characterized, we performed an exome-wide, gene-based association study of familial cutaneous melanoma (CM) and ocular melanoma (OM). Using 11 990 jointly processed individual DNA samples, whole-exome sequencing was performed, followed by large-scale joint variant calling using GATK (Genome Analysis ToolKit). PLINK/SEQ was used for statistical analysis of genetic variation. Four models were used to estimate the association among different types of variants. In vitro functional validation was performed using three human melanoma cell lines in 2D and 3D proliferation assays. In vivo tumor growth was assessed using xenografts of human melanoma A375 melanoma cells in nude mice (eight mice per group). All statistical tests were two-sided. Strong signals were detected for CDKN2A (Pmin = 6.16 × 10-8) in the CM cohort (n = 273) and BAP1 (Pmin = 3.83 × 10-6) in the OM (n = 99) cohort. Eleven genes that exhibited borderline association (P < 10-4) were independently validated using The Cancer Genome Atlas melanoma cohort (379 CM, 47 OM) and a matched set of 3563 European controls with CDKN2A (P = .009), BAP1 (P = .03), and EBF3 (P = 4.75 × 10-4), a candidate risk locus, all showing evidence of replication. EBF3 was then evaluated using germline data from a set of 132 familial melanoma cases and 4769 controls of UK origin (joint P = 1.37 × 10-5). Somatically, loss of EBF3 expression correlated with progression, poorer outcome, and high MITF tumors. Functionally, induction of EBF3 in melanoma cells reduced cell growth in vitro, retarded tumor formation in vivo, and reduced MITF levels. The results of this large rare variant germline association study further define the mutational landscape of hereditary melanoma and implicate EBF3 as a possible CM predisposition gene.
Bibliography:ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
Authors contributed equally to this work.
For the full list of authors and affiliations, please see the Notes section.
ISSN:0027-8874
1460-2105
1460-2105
DOI:10.1093/jnci/djx083