Abstract
Purpose:
The activation of the mitogen-activated protein kinase (MAPK) pathway has been suggested as the major downstream target when GNAQ and GNA11 (GNAQ/11) are mutated in uveal melanoma (UM). However, clinical trials with single agent MEK inhibitor showed no clinical significance in altering the overall outcome of the disease in UM; therefore, we investigated the correlation between naturally occurring mutations in GNAQ/11 and activation of MAPK pathway in vivo in primary UM.
Methods:
Screening for activating mutations in codons 183 and 209 of GNAQ/11 was carried out by sequencing and restriction fragment length polymorphism (RFLP) in a cohort of 42 primary UM. Activation of the MAPK pathway and other potential downstream signals was assessed by immunohistochemistry and/or Western blot analysis. Potential downstream signaling of mutant and wild type GNAQ/11 was studied by transient transfection assay in nonmutant cell lines.
Results:
Somatic mutations in GNAQ/11 were observed in 35/42 (83.3%) of primary UM. Tumors with GNAQ/11 mutations showed variations in the activation of ERK1/2 with significant tumor heterogeneity. Weak and undetectable ERK1/2 activation was observed in 4/35 (11.4%) and 8/35 (22.9%) of the GNAQ/11 mutant UM, respectively. Tumor heterogeneity of GNAQ/11 mutations was also observed in a subset of tumors.
Conclusions:
Our results indicate that there is marked variation in MAPK activation in UM with GNAQ/11 mutations. Thus, GNAQ/11 mutational status is not a sufficient biomarker to adequately predict UM patient responses to single-agent selective MEK inhibitor therapy.
Uveal melanoma (UM) is the most common primary intraocular tumor in adults. Currently, there is no effective therapy for metastatic UM and research is ongoing to identify promising targeted therapies. Activation of mitogen-activated protein kinase (MAPK) has been observed in UM.
1 It has been suggested that such activation is mostly due to somatic mutations in
GNAQ and
GNA11 (
GNAQ/11).
2–4 Somatic mutations in
GNAQ/11 are reported in more than 80% of the UM tumors.
2–5 With the exception of blue nevi and melanomas of the central nervous system, somatic mutations in
GNAQ/
11 are unique to UM and have not been reported in other cancers.
6 The mutations in both genes occur mostly in two loci one in exon 4 (codon 183) and the other in exon 5 (codon 209). Codon 209 mutations in both genes are much more common than codon 183 mutations.
4 Mutations in another two genes
PLCB4 and
CYSLTR2 leading to constitutively activated G-protein signaling have also been reported in UM, although at much lower frequencies than
GNAQ/11.
7,8 Mutations in
GNAQ,
GNA11,
PLCB4, and
CYSLTR2 are mostly mutually exclusive.
7,9 The contribution of
PLCB4 and
CYSLTR2 mutations to MAPK activation is still not clear.
It has been suggested that selective inhibitors of MAPK pathway could be useful targets for therapy in patients with metastatic disease.
4 This has been supported by in vitro studies and a phase II clinical trial of the selective MEK inhibitor selumetinib (AZD6244) for metastatic uveal melanoma
10 where a modest improvement of progression-free but not overall survival was observed. However, such outcomes were not replicated in a phase III clinical trial of combined selumetinib and chemotherapeutic agent temozolomide.
11 Interestingly, in both trials the response to therapy did not correlate with the
GNAQ/11 mutation status. It has been suggested that resistance genes such as the RNA helixase
DDX21 and the cyclin-dependent kinase regulator
CDK5R1 could play an important role in lack of response to selumetinib in
GNAQ/11 mutant UM.
12 Intratumoral variation in the degree of MAPK activation could be additional explanation for the lack of response to selective MEK inhibition in a subset of tumors.
13,14 A study on primary UM with
GNAQQ209L/P mutation suggested that a subset of tumors with the mutation showed weak or no activation in MAPK.
15 However, that report was limited by studying only
GNAQ codon 209 mutations and the small number of samples included.
15 Our current study was conceived prior to the two clinical trials and was carried out to validate our preliminary findings that there was a lack of MAPK activation in a significant number of UM primary tumors with somatic
GNAQ/11 mutations.
16 In addition to confirming our earlier findings, we identified significant heterogeneity of MAPK activation within individual tumors. These findings provide an additional explanation for the lack of response to selective MEK inhibition in a subset of tumors and reveal the need to develop additional biomarkers to predict UM tumor responses to selective MEK inhibitors.
Plasmids containing the GNAQ (wild type), GNA11 (wild type), GNAQQ209L, and GNA11Q209L cDNAs were obtained from the Missouri S&T cDNA Resource Center. All the constructs contained an internal epitope tag (Glu-Glu; Sigma-Aldrich Corp., St. Louis, MO, USA) and were verified by sequencing. The primary uveal melanoma culture (UM7007) and the ARPE-19 cell line, both with no GNAQ/11 mutations, were used for transfection. Cells were grown in duplicates in six well plates to 75% to 80% confluence in RPMI medium containing 10% FBS, and 1% Penicillin/Streptomycin. Then they were transiently transfected with 4 μg of plasmid pcDNA3.1+ constructed with complete coding regions of wild type GNAQ, and GNA11, and mutant GNAQQ209L, and GNA11Q209L genes using a transfection reagent (Lipofectamine 2000; Invitrogen, Carlsbad, CA, USA). Cells transfected with the empty vector or treated with only lipofectamine 2000 were used as controls. Cells were grown in medium (Opti-MEM I Reduced Serum Medium, Cat. No. 31985-062; Invitrogen) and lysed 24 hours posttransfection.
The activation of MAPK and AKT pathways was assessed by Western blot for the levels of phosphorylated ERK1/2, MEK1/2, and AKT proteins. Activation of MAPK in tumors was also assessed by immunohistochemistry using antibody for pERK1/2. Immunohistochemistry was carried-out on 39 formalin-fixed paraffin embedded primary UMs and Western blot was carried-out on 17 fresh frozen tumor specimens. For 14 tumors, both Western blot and immunohistochemistry were performed (see
Supplementary Table S3). Antibodies for phospho p44/42 MAPK (pERK1/2-Thr202/Tyr204), phospho MEK1/2 (pMEK1/2-Ser217/221), and phospho AKT (pAKT-Ser473) were obtained from Cell Signaling Technology (Danvers, MA, USA) and β-Actin antibody was obtained from Sigma-Aldrich Corp.
For Western blot analysis, total proteins were extracted from tumor tissues and cell lines by incubation for 10 minutes with ice cold 1X cell lysis buffer (Cell Signaling Technology) spiked with 1 mM PMSF and 1X phosphatase inhibitor cocktail 2 (Sigma-Aldrich Corp.) immediately before use. The protein concentration was determined using a BCA protein assay kit (Pierce Biotechnology, Rockford, IL, USA). For Western blot analysis, 10 to 30 μg of protein extracts/sample were loaded on 12% Tris-HCl gel and transferred to Trans-Blot nitrocellulose filters (Bio-Rad Laboratories, Hercules, CA, USA). The primary and secondary antibodies were used at 1:1000 and 1:2000 dilutions respectively. Signals were developed using a chemiluminescent substrate (Super Signal West Pico Chemiluminescent Substrate; Pierce Biotechnology, Rockford, IL, USA). Densitometry was carried out on the scanned Western blot images using image analysis (Alphaease FC, software version 6.0.0; Alpha Innotech Corp., San Leandro, CA, USA) Data are representative of two experiments.
For immunohistochemistry, deparaffinized tissue sections were heat pretreated with citrate buffer, pH of 6, and incubated overnight at 4°C with pERK1/2 antibody at a 1:200 dilution. Immunostaining omitting the primary antibody was used as negative control. For detection of the immunostaining, chromogen (Vector NovaRED; Vector Laboratories, Burlingame, CA, USA) was utilized to produce a dark red staining to minimize the interference from melanin pigment. After counterstaining with hematoxylin and mounting, the slides were evaluated under a light microscope by two independent investigators (MHA, CMC) masked in regard to the mutation status of each tumor. Based on the correlation between immunostaining and Western blot analysis in the 14 tumors, staining limited to ≤1% of the tumor was considered negative (pERK1/2 score = 0), staining >1% to <10% was considered weak (score = 1), and staining ≥10% of the tumor was considered moderate to strong (score = 2). Staining of retina was utilized as an internal positive control for assessment of the adequacy of the immunostaining assay. Tumor areas with intense melanin pigmentation were excluded from our analysis.
Out of the 80 tumors sequenced through the NIH TCGA (The Cancer Genome Atlas) project, 72 had a single mutation in either GNAQ or GNA11 and two tumors had two mutations, one in each gene. The average allele frequency of mutations in GNAQ was 0.42 (range, 0.07–0.65) and of GNA11 was 0.41 (range, 0.05–0.56). Four tumors had somatic mutation allele frequency less than 0.25 (0.24, 0.23, 0.07, 0.05). The two tumors with very low mutation allele frequencies (0.05 and 0.07) were those with mutations in both genes. This suggests that in a small subset of tumors the variation in MAPK activation could be due to allele heterogeneity.
Supported by the Patti Blow Research Fund in Ophthalmology, and funds from the Ohio Lions Eye Research Foundation, Ocular Melanoma Foundation, Melanoma Know More Foundation, the R21CA191943 Grant from the National Cancer Institute, the National Eye Institute grant K08EY022672, a cancer center core grant 2P30CA016058-40 and award number 8UL1TR000090-05 from the National Center For Advancing Translational Sciences. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.
Disclosure: G. Boru, None; C.M. Cebulla, None; K.M. Sample, None; J.B. Massengill, None; F.H. Davidorf, None; M.H. Abdel-Rahman, None