Abstract
The classification of melanoma subtypes into prognostically relevant and therapeutically insightful categories has been a challenge since the first descriptions of melanoma in the 1800s. One limitation has been the assumption that the two most common histological subtypes of melanoma, superficial spreading and nodular, evolve according to a linear model of progression, as malignant melanocytes spread radially and then invade vertically. However, recent clinical, pathological, and molecular data indicate that these two histological subtypes might evolve as distinct entities. We here review the published data that support distinct molecular characterization of superficial spreading and nodular melanoma, the clinical significance of this distinction including prognostic relevance, and the therapeutic implications.
Keywords: melanoma, histological subtype, nodular melanoma, superficial spreading melanoma, linear progression
Background
Since the first descriptions of melanoma in the 1800s, pathologists and clinicians have been trying to categorize it in meaningful ways. In 1912, Dubreuilh, a French dermatologist, provided the first classification system by dividing melanomas according to their precursor lesion such as those arising in a nevus, those arising in pigmented skin lesions that appeared in adult life and those arising in previously blemish-free skin [1]. Fifty-five years later, Australian pathologists published their own recommendations for categorization based on both histological and clinical features [2]. However, Wallace Clark and colleagues’ seminal publication in 1969 on the histopathological classification of melanoma has retained its importance and remains the basis of the World Health Organization (WHO) classification for melanoma [3, 4]. The current four major clinical-histopathological subtypes are superficial spreading melanoma (SSM), nodular melanoma (NM), acral-lentiginous melanoma (ALM) and lentigo maligna melanoma (LMM) [3, 5].
To date, the impact of histologic subtypes on melanoma prognosis and treatment has been relatively limited. Staging is determined by the American Joint Committee on Cancer (AJCC) system that incorporates tumor thickness, ulceration, mitotic index and the lymph node status but not histological subtype in the recurrence/metastases risk assessment of patients with primary, localized melanoma [6]. Yet, recent discoveries in other solid tumor types have highlighted the potential of histology driven molecular characterization to impact clinical decision making [7–10]. For example, in lung adenocarcinomas mutational analysis identified correlations between major histopathological subtypes and genetic aberrations that were responsive to targeted therapies [11–13].
The clinical utility of histological classification in melanoma has been exemplified by acral-lentiginous melanoma (ALM). ALM, which makes up roughly 10% of primary melanomas, has been redefined as a distinct and useful sub-classification of melanoma based on the prevalence of the c-kit mutation. The identification of distinct molecular alterations characterizing ALM and melanoma arising in sun protected areas [14–17] has prompted the initiation of phase II trials evaluating the efficacy of the c-KIT inhibitor imatinib as part of a subtype-specific melanoma treatment strategy for metastatic melanoma [8, 9]. Preliminary results from one such trial reported that the patient whose metastatic tumor had the most robust expression of KIT had an almost complete response, emphasizing the importance of delineating and targeting molecular pathways that define specific melanoma histologic subtypes [8].
Though over 80% of melanomas are histologically classified as either superficial spreading (SSM) or nodular melanoma (NM) (or alternatively referred to as radial growth phase (RGP) and vertical growth phase (VGP), respectively), the clinical relevance of these subtypes have been largely overlooked. This is in part due to the current understanding of the linear model of melanoma progression, which entails that melanoma begins with the transformation of epidermal melanocytes that initially undergo radial growth (SSM) and subsequently transition to vertical growth (NM) [18]. It is currently accepted that the speed of dermal invasion is the only aspect that separates the NM and SSM subtypes [19, 20]. Nevertheless, evidence has emerged challenging this linear model of progression and suggesting that these two subtypes are distinct entities [16, 21–28].
Here we review current data on the clinical and biologic heterogeneity of SSM and NM, and discuss its potential implications for treatment options and melanoma outcome.
Clinical Differences of Superficial Spreading and Nodular Melanoma
The incidence of melanoma has increased 3–7% per year, faster than any other cancer, and is predicted to double every 10–20 years [29]. This increase is in part due to widespread awareness of melanoma and the “ABCD” rule for self-monitoring of moles. This criteria is used to raise consciousness about mole Asymmetry, Border irregularity, Color (uneven), and Diameter (>6mm) as warnings signs for melanoma.
Although the incidence of melanoma is increasing, SSM and NM make up steady rates of about 65% and 20%, respectively [20], of all primary melanomas diagnosed. However, different clinical presentations suggest divergent characteristics of SSM and NM. Studies comparing melanoma cohorts over time have supported that there have been changes in the clinical presentation of SSM without corresponding differences in NM. Specifically, our work at New York University found a 25% increase in the proportion of Stage I SSM when comparing a cohort form 1972–82 to a cohort from 2002–07 [22]. This was consistent with our finding that the median SSM thickness had decreased over time by nearly 50% from 1.10 mm to 0.6 mm. However, there was not a significant shift in NM tumor thickness at diagnosis and stage at presentation remained stable with a trend towards an increase in Stage I [22]. Population based studies in Queensland, Australia, and Southern Germany found a similar trend of thick NM compared to SSM [24, 30]. More recently, a population based study in Italy that examined melanoma patients diagnosed between 1984 and 2004 also found no change in median thickness for NM over time, while SSM thickness significantly decreased [23]. The data from the 4 studies are summarized in Table 1. These findings are consistent with non-hospital-based prospective databases such as the Surveillance, Epidemiology, and End Results (SEER) cancer registry [31–37], and illustrate stage migration of SSM, likely due to increased surveillance, while there is a lack of progress with NM.
Table 1.
Changes in clinical presentation of superficial spreading and nodular melanoma over time
| Location | Sample Source | Time Period |
Histological Subtype Proportions |
Changes in Thickness |
Trends Noted |
Limitations | References |
|---|---|---|---|---|---|---|---|
| New York, USA | Distinct cohorts from the New York University Medical Center |
1971–1982 & 2002–2007 |
1119 SSMs 253 NMs 283 Others |
SSM: median of 1.1 mm to 0.6 mm (p<0.001) NM: median of 3.2 mm to 2.4 mm (p=0.10) |
SSM thickness decreased while NM stayed constant | Academic referral centers tend to see more aggressive melanoma cases | Warycha et al 2008 |
| Queensland, Australia | Population based cancer registry in Queensland, Australia | 2000–2003 | 369 NM | NM: Median: 1.90 mm Range: 0.40 mm– 9.30 mm |
Only examined nodular melanoma over 4 years | Geller et al 2009 | |
| Munich, Germany | Cohort from Ludwig-Maximilian-University of Munich | 1977–2000 | 3660 SSMs 1703 NMs 1112 Others |
Female: SSM: −1.5 AAPC (p<0.001) NM: −0.04 AAPC (p=0.379) Male: SSM: −1.7 AAPC (p<0.001) NM: −0.7 AAPC (p=0.090) |
No thickness values reported. Values reported based on gender. | Baumert et al 2009 | |
| Tuscany, Italy | Population based cancer registry in providences of Florence and Prato, Italy | 1985– 2004 (1985–1989 & 2000–2004) |
1655 SSMs 238 NMs 1009 Others |
SSM: median of 1.20 mm to 0.68 mm (p<0.001) NM: median of 3.33 mm to 4.00 mm (p=0.517) |
Included in-situ cases. | Crocetti et al 2010 |
Key: SSM: superficial spreading melanoma; NM: nodular melanoma; AAPC: average annual percentage change
The lack of improvement toward a more prognostically favorable stage at diagnosis in NM may be explained by its unique phenotype that often fails the ABCD categorization of suspicious moles, as they are more likely to exhibit border regularity and homogeneous color [22]. However, the majority of nodular melanoma patients do report a history of change in their mole, typically shorter in duration than for SSM [38]. Notably, our group has previously reported that the median duration of change in a lesion before treatment, as assessed by patient recall, was only 5 months for NM compared to 9 months for SSM [22, 38, 39]. This suggests that it is not late presentation and delayed diagnosis that accounts for advanced thickness of NM at presentation, but rather indicates a more clinically aggressive biology of NM and supports the importance of adding ‘E’ for ‘evolution’ to the ABCD criteria [22, 30, 40, 41].
There are consistent pathological differences between SSM and NM, lending further support for inherent differences in the tumor types. For example, NMs have been more invasive, ulcerated, and have more mitoses than SSMs [22]. Taken together, these differences lend support to the postulate that SSM and NM are distinct entities and suggest that biological heterogeneity might play a role in driving different phenotypic and clinical presentation of SSM and NM.
Biologic Heterogeneity of Superficial Spreading and Nodular Melanoma
Different Alterations in the MAPK Pathway in Superficial Spreading and Nodular Melanoma
Discoveries related to the disruption of normal functioning of the mitogen activated protein kinase (MAPK) pathway have driven breakthroughs in understanding the biology of melanoma. Constitutive activation of the pathway by NRAS mutations on chronically sun-exposed skin and BRAF on intermittently sun-exposed skin has launched the development of a variety of targeted therapies [42]. It is crucial to consider and incorporate known the molecular aberrations involved in the MAPK pathway in order to make histological subtypes more meaningful distinctions, but there is conflicting evidence regarding the association of melanoma subtypes and these mutations [43, 44].
As the first oncogenic mutation defined in melanoma, NRAS mutations make up almost virtually all RAS mutations in melanoma and occur in about 15% of primary melanomas [45–49]. These mutations occur at codons 12, 13, and 61 and transform the GTPase into a constitutively active state [50]. Several groups found no difference in NRAS mutation rates among SSM and NM [51, 52], although other studies report higher rates in NM [43, 53, 54]. These mixed associations with histopatholoigcal subtype need to be further explored in larger cohorts that take the degree of sun damage into account in a standardized way [17, 44, 55, 56].
BRAF somatic missense mutations occur in about 66% of melanomas, and tumors almost never harbor both NRAS and BRAF mutations simultaneously [17, 57–59]. Studies have shown that the BRAF mutation is not sufficient on its own for malignant transformation [60] although the recent FDA approval of a BRAF specific inhibitor has made the testing for BRAF mutations part of the standard of care both to identify patients who might benefit from targeted therapy and to utilize in clinical management.
Several studies have demonstrated that BRAF mutation is less common in NM than SSM [17, 44, 53, 55, 56, 61–63] and is more frequently mutated in AML melanomas [64]. Of note, this study was conducted in a Japanese population, which has been shown to have higher frequencies of ALM (41.9% vs. 10.0% in US), and lower frequency of SSM (11.8% vs. 69.6% in US), bringing the general applicability of these results in other populations into discussion. Other studies have found similar BRAF mutation rates of SSM and NM, but all the tumors included were relatively thick [51, 65]. The lack of consensus on histologic-genetic correlations in known genetic alterations in melanoma underlies the importance of utilizing unbiased approached and new technologies to explore other alterations.
Differences in Superficial Spreading and Nodular Melanoma Identified using Integrative Genomics
Uncovering biologic distinctions between SSM and NM is integral to understanding the phenotypic differences between these histological subtypes. High throughput techniques such as comparative genomic hybridization (CGH), single nucleotide polymorphism (SNP) arrays and microarrays have potential as unbiased approaches to identifying subtype specific alterations. Although both CGH and SNP array can detect chromosomal amplification and loss as well as cytogenic abnormalities, SNP array can also identify regions of loss of heterozygosity [66]. CGH has been extensively utilized in melanoma in the last 10 years, but it has mainly been used for three purposes: 1. to distinguish nevi (usually Spitz nevi) from melanoma [67, 68]; 2. to identify the genomic alterations that characterize uveal melanoma (subtypes) and correlate with prognosis [69–72]; and 3. to identify the differences between primary cutaneous melanoma (irrespectively of the histologic subtype) and metastasis [73, 74]. While CGH has also been used to identify the genomic alterations that distinguish melanomas with different degrees of sun damage as measured by the presence or absence of solar elastosis, it has not been applied to identify the differences between SSM and NM. As such, the CGH characterization of SSM and NM is still missing.
Microarray technology has enabled gene expression profiling to provide better understanding of tumorigenesis via simultaneous evaluation of the expression levels of thousands of genes in a single experiment [75–77]. Our group recently examined genomic and gene expression alternations using integrative genomics to reveal molecular differences in SSM and NM. In a comparison of normal melanocytes, primary NM, primary SSM, metastatic NM and other metastatic melanoma cell lines, single nucleotide polymorphism (SNP) array data indentified 408 SSM-specific regions with significant genomic alteration and 543 that were NM-specific [21]. Moreover, 114 differentially expressed genes were identified using gene expression array, over half of which, including MTAP and ALDH7A1, fell into the category of metabolic processes. MTAP, ALDH7A1 and erythrocyte membrane protein EPB41L3 displayed the highest fold changes in gene expression, and at the same time showed lower expression in SSM compared to NM and nevi, with results verified by genomic PCR and qRT-PCR [21]. The linear progression model predicts that all the genomic differences would follow step-wise transformations from SSM to NM. However, the dip in gene expression levels in SSM compared to nevi and NM cannot be explained by a step-wise transformation. These alterations suggest underlying genetic differences and support the hypothesis of separate biological entities. When the SNP array data and the gene expression array data were analyzed for interaction, five genes showed significant copy number loss that correlated with lower gene expression in SSM compared to NM. The presence of recurrent SSM-specific deletions that are present or even amplified NM cannot be reconciled with the linear progression model, even taking epigenetic modifications into account. To further support the fidelity of these findings, external validation was performed using two publically available gene expression data sets [16, 26]. Jaeger et al. identified 67 genes differentially expressed in SSM and NM, including SSM-specific down regulation of the five genes validated by using quantitative PCR and qRT-PCR in our study. Whole genome microarrays performed by Scatoloini and colleagues only included 3 of the genes (MTAP, SEC23IP and G3BP2) but all showed the same downregulation in SSM-like RGP cell lines compares to VGP cell lines for the three genes [26]. These results represent a discordance with the current paradigm of linear progression and introduce the existence of differentially expressed genetic alterations that can potentially be exploited in the search for targeted, subtype specific therapies.
Not only has genetic evidence emerged supporting discrete pathways of development from the transformed melanocyte to invasive melanoma for SSM and NM that is in contrast to the theory of sequential progression, but gene expression patterns also suggest that there are subtype-specific alterations. The studies reporting differentially expressed genes are summarized in Table 2.
Table 2.
Summary of studies reporting differential gene and microRNA expression found between superficial spreading and nodular melanoma
| Sample Size | Identified Alterations between SSM and NM |
Technologies Used | References | ||
|---|---|---|---|---|---|
| Array Based | Validated Targets | Discovery | Validation | ||
|
Cell Lines: 1 NHM 2 Melanocytes 3 SSM 2 NM 2 VGPM 2 Metastatic NM 12 Met UP Tissues: 20 NM 20SSM |
114 differentially expressed genes | DIS3, FGFR1OP, G3BP2, GALNT7, MTAP, SEC23IP, USO1, and ZNF668 with NM/SSM copy number gain/loss correlated to altered gene expression Deleted in SSM: USO1, G3BP2, DIS3, and SEC23IP |
|
|
Rose et al 2011 |
|
Tissues: 13 SSM 5 NM 1 ALM |
60 differentially expressed genes | Lower in SSM: MMP16, BCL2A1, ICAM1 and CEACAM1 |
|
|
Jaeger et al 2007 |
|
Screening: 18 CN 11 DN 8 RGPM 15 VGPM 5 MTS Validation: 5 CN 5 RGPM 5 VGPM 5 MTS |
540 differentially expressed sequences | Lower in RGPM: AKT3, CTNNB1, NRAS, API5, BCL2A1, HSPA5, BIRC2, ANXA5, NPM1, TNS3, HSP90AB1, PAIP1 and DAPL1 |
|
|
Scatolini et al 2010 |
|
Tissues: 26 SSM 56 NM 9 CN Cell Lines: 4 RGPM 6 VGPM |
134 microRNAs differentially expressed | Genomic loss in SSM: let-7g, miR-15a, miR-16, miR-138, miR-181a, miR-191 and miR-933 |
|
|
Poliseno et al 2011 |
Key: SSM: superficial spreading melanoma; NM: nodular melanoma, ALM: acral-lentiginous melanoma; CN: common nevi; DN: dysplastic nevi; RGPM: radial growth phase melanomas; VGPM: vertical growth phase melanomas; MTS: melanoma metastases; qRT-PCR: quantitative Reverse Transcriptase Polymerase Chain Reaction; NHM: normal melanocytes cultured from infant foreskin; Met UP: metastatic melanoma of unknown primary histologic subtype
microRNA Differential Expression in Superficial Spreading and Nodular Melanoma
Improvements in our understanding of the functional relevance of microRNAs and advances in array based technology have allowed for high-throughput profiling of these genetic regulators to further uncover tumor biology. microRNAs have consequently emerged as a key regulations in melanomagenesis. As short, endogenous, non-coding RNA molecules, they negatively regulate gene expression at the post-transcriptional level. microRNAs can also accurately distinguish different cell types [78, 79]. Moreover, since each microRNA can potentially modulate several hundred genes, changes in expression of a single microRNA can have deep pathological consequences [80–82]. Their ability to successfully discriminate between cancer tissues and normal tissues, subtype, age at diagnosis and mutational status in other cancers suggests that microRNA expression can also yield insight into distinctions among melanoma histopathological subtypes [83–86]. ALM, already distinguished as a unique histologic subtype, has been shown to exhibit an ALM-specific microRNA pattern [87].
A study by our group found that among primary melanoma samples and congenital nevi (CN), the majority of microRNAs (n=31) were downregulated in SSM compared to both CN and NM, and real time PCR on genomic DNA revealed that the genomic loci corresponding 7 microRNAs selectively lost in SSM, supporting the results obtained by microRNA array [25]. Only about half of the microRNAs showed stepwise differential expression from CN to SSM to NM. This data also suggests that genomic differences cannot be reconciled with the linear model of progression.
Additional Studies Supporting Differences between Superficial Spreading and Nodular Melanoma
Several recent studies have aimed to undercover relevant molecular trends among the current histologic subtypes. Kanoh and colleagues examined 15 NM and 12 SSM formalin-fixed, paraffin-embedded (FFPE) tissues for levels of nestin, a marker or neural stem cells that has recently been suggested to be a marker of melanoma cells of origin [88]. They found nestin immunoreactivity in all amelanotic and 80% of the melanotic NMs while expression was only seen in 17% of the SSM cases [28]. However, this small sample size was not matched for thickness and previous reports have shown that nestin expression was significantly correlated with the stage of melanomas [89].
CDKN2A is a gene known to be important in both sporadic and hereditary melanoma. A mutation in this gene, which codes for p16INK4A (p16), in melanoma prone families is more common in SSM compared to NM and other histological subtypes [90]. In examining p16 protein levels, one group found decreased expression among NM compared to SSM [91] although previous studies report mixed expression levels [92–95]. However, Pavey and colleagues do not control for mitosis and thickness in their analysis, both of which have been shown to vary with p16 expression [91, 96].
In another analysis of subtype differences, our group investigated the clinicopathological relevance of cryptic epitope HU177 in serum from 291 melanoma patients. Extracellular matrix remodeling has been shown to generate protein fragments, such as Hu177 and other cryptic collagen peptides, which may function in tumor migration and angiogenesis [97–100]. After controlling for time of blood draw and tumor thickness, multivariate analysis showed a significant relationship between higher HU177 levels and NM. Elevated levels were trending towards association with ulcerated melanomas and recurrence but remained insignificant [101]. HU177 has also been shown to correlative with tumor response in a phase II trial of sorafenib (BAY 43-9006) in melanoma, suggesting future development of histotype specific biomarkers might have clinical utility [102].
Future Steps
New technologies using high throughput sequencing have provided an opportunity to gain a more complex understanding of the biology underlying melanomagenesis. Present evidence suggests that NM and SSM cannot arise sequentially via the linear model of progression and that they are two distinct biologic entities. This shift in the understanding of how melanoma develops dictates a revision of clinical care procedures based on histologic-genetic correlations. Building on the expanding foundation of knowledge regarding the biology of melanoma, alterations defining these two subtypes can be utilized moving forward to improve prognosis and develop novel therapeutic approaches. Histological subtypes can be integrated into prognostic decision trees or added to the AJCC staging as a complement to existing schemes.
While there have been new classifications proposed based solely on molecular alterations and sun exposure and ignoring histopathological subtype [103], the existing subtypes have a wealth of molecularly relevant information. The ALM subtype in melanoma as well histological subtypes in other cancers have effectively transformed their molecularly enhanced classification into individualized targeted treatments. In addition to ALM, mucosal and uveal melanoma, which are less common melanoma histologic subtypes, have also utilized specific molecular alterations for treatment [104–106]. The experimental evidence that we have summarized offers clear rationale for extending this approach to SSM and NM through the identification of new, and hopefully more effective, subtype-specific therapeutic strategies.
Acknowledgments
Grant support: This work was supported by the NYU Cancer Institute Cancer Center Support Grant (5 P30 CA016087-27 to I.O.).
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Conflicts of interest: None to disclose.
References
- 1.Scolyer RA, Long GV, Thompson JF. Evolving concepts in melanoma classification and their relevance to multidisciplinary melanoma patient care. Molecular oncology. 2011;5(2):124–136. doi: 10.1016/j.molonc.2011.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Moles and malignant melanoma: terminology and CLASSIFICATION. The Medical journal of Australia. 1967;1(3):123–125. doi: 10.5694/j.1326-5377.1967.tb21063.x. [DOI] [PubMed] [Google Scholar]
- 3.Clark WH, Jr., et al. The histogenesis and biologic behavior of primary human malignant melanomas of the skin. Cancer research. 1969;29(3):705–727. [PubMed] [Google Scholar]
- 4.LeBoit PE, et al. World Health Organization classification of tumours. Lyon: IARC Press; 2006. Pathology and genetics of skin tumours; p. 355. [Google Scholar]
- 5.McGovern VJ, et al. The classification of malignant melanoma and its histologic reporting. Cancer. 1973;32(6):1446–1457. doi: 10.1002/1097-0142(197312)32:6<1446::aid-cncr2820320623>3.0.co;2-8. [DOI] [PubMed] [Google Scholar]
- 6.Balch CM, et al. Final version of 2009 AJCC melanoma staging and classification. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2009;27(36):6199–6206. doi: 10.1200/JCO.2009.23.4799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Motoi N, et al. Lung adenocarcinoma: modification of the 2004 WHO mixed subtype to include the major histologic subtype suggests correlations between papillary and micropapillary adenocarcinoma subtypes, EGFR mutations and gene expression analysis. The American journal of surgical pathology. 2008;32(6):810–827. doi: 10.1097/PAS.0b013e31815cb162. [DOI] [PubMed] [Google Scholar]
- 8.O E, et al. Phase II trial of imatinib mesylate (STI-571) in metastatic melanoma (MM) Journal of Clinical Oncology. 2004;22(14S):7528. [Google Scholar]
- 9.Wyman K, et al. Multicenter Phase II trial of high-dose imatinib mesylate in metastatic melanoma: significant toxicity with no clinical efficacy. Cancer. 2006;106(9):2005–2011. doi: 10.1002/cncr.21834. [DOI] [PubMed] [Google Scholar]
- 10.Weigelt B, Reis-Filho JS. Histological and molecular types of breast cancer: is there a unifying taxonomy? Nature reviews. Clinical oncology. 2009;6(12):718–730. doi: 10.1038/nrclinonc.2009.166. [DOI] [PubMed] [Google Scholar]
- 11.Riely GJ, et al. Clinical course of patients with non-small cell lung cancer and epidermal growth factor receptor exon 19 and exon 21 mutations treated with gefitinib or erlotinib. Clinical cancer research : an official journal of the American Association for Cancer Research. 2006;12(3 Pt 1):839–844. doi: 10.1158/1078-0432.CCR-05-1846. [DOI] [PubMed] [Google Scholar]
- 12.Mitsudomi T, et al. Mutations of the epidermal growth factor receptor gene predict prolonged survival after gefitinib treatment in patients with non-small-cell lung cancer with postoperative recurrence. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2005;23(11):2513–2520. doi: 10.1200/JCO.2005.00.992. [DOI] [PubMed] [Google Scholar]
- 13.Jackman DM, et al. Exon 19 deletion mutations of epidermal growth factor receptor are associated with prolonged survival in non-small cell lung cancer patients treated with gefitinib or erlotinib. Clinical cancer research : an official journal of the American Association for Cancer Research. 2006;12(13):3908–3914. doi: 10.1158/1078-0432.CCR-06-0462. [DOI] [PubMed] [Google Scholar]
- 14.Chernoff KA, et al. GAB2 amplifications refine molecular classification of melanoma. Clinical cancer research : an official journal of the American Association for Cancer Research. 2009;15(13):4288–4291. doi: 10.1158/1078-0432.CCR-09-0280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Smalley KS, et al. Identification of a novel subgroup of melanomas with KIT/cyclin-dependent kinase-4 overexpression. Cancer research. 2008;68(14):5743–5752. doi: 10.1158/0008-5472.CAN-08-0235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Jaeger J, et al. Gene expression signatures for tumor progression, tumor subtype, and tumor thickness in laser-microdissected melanoma tissues. Clinical cancer research : an official journal of the American Association for Cancer Research. 2007;13(3):806–815. doi: 10.1158/1078-0432.CCR-06-1820. [DOI] [PubMed] [Google Scholar]
- 17.Curtin JA, et al. Distinct sets of genetic alterations in melanoma. The New England journal of medicine. 2005;353(20):2135–2147. doi: 10.1056/NEJMoa050092. [DOI] [PubMed] [Google Scholar]
- 18.Kwong L, Chin L, Wagner SN. Growth factors and oncogenes as targets in melanoma: lost in translation? Advances in dermatology. 2007;23:99–129. doi: 10.1016/j.yadr.2007.07.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Ackerman AB. Malignant melanoma. A unifying concept. The American Journal of dermatopathology. 1980;2(4):309–313. doi: 10.1097/00000372-198000240-00004. [DOI] [PubMed] [Google Scholar]
- 20.Barnhill RL, Mihm MC., Jr. The histopathology of cutaneous malignant melanoma. Seminars in diagnostic pathology. 1993;10(1):47–75. [PubMed] [Google Scholar]
- 21.Rose AE, et al. Integrative Genomics Identifies Molecular Alterations that Challenge the Linear Model of Melanoma Progression. Cancer Res. 2011 doi: 10.1158/0008-5472.CAN-10-2958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Warycha MA, et al. Changes in the presentation of nodular and superficial spreading melanomas over 35 years. Cancer. 2008;113(12):3341–3348. doi: 10.1002/cncr.23955. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Crocetti E, et al. The thickness of melanomas has decreased in central Italy, but only for thin melanomas, while thick melanomas are as thick as in the past. Melanoma research. 2010;20(5):422–426. doi: 10.1097/CMR.0b013e32833d9d36. [DOI] [PubMed] [Google Scholar]
- 24.Geller AC, et al. Factors related to the presentation of thin and thick nodular melanoma from a population-based cancer registry in Queensland Australia. Cancer. 2009;115(6):1318–1327. doi: 10.1002/cncr.24162. [DOI] [PubMed] [Google Scholar]
- 25.Poliseno L, et al. Distinguishing between nodular and superficial spreading melanoma using specific microRNA alterations. Journal of Clinical Oncology. 2011;29 p. (suppl; abstr 8540) [Google Scholar]
- 26.Scatolini M, et al. Altered molecular pathways in melanocytic lesions. Int J Cancer. 2010;126(8):1869–1881. doi: 10.1002/ijc.24899. [DOI] [PubMed] [Google Scholar]
- 27.Gaziel-Sovran A, et al. miR-30b/30d regulation of GalNAc transferases enhances invasion and immunosuppression during metastasis. Cancer Cell. 2011;20(1):104–118. doi: 10.1016/j.ccr.2011.05.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Kanoh M, et al. Nestin is expressed in HMB-45 negative melanoma cells in dermal parts of nodular melanoma. The Journal of dermatology. 2010;37(6):505–511. doi: 10.1111/j.1346-8138.2010.00791.x. [DOI] [PubMed] [Google Scholar]
- 29.Garbe C, Leiter U. Melanoma epidemiology and trends. Clin Dermatol. 2009;27(1):3–9. doi: 10.1016/j.clindermatol.2008.09.001. [DOI] [PubMed] [Google Scholar]
- 30.Baumert J, et al. Time trends in tumour thickness vary in subgroups: analysis of 6475 patients by age, tumour site and melanoma subtype. Melanoma Res. 2009;19(1):24–30. doi: 10.1097/CMR.0b013e32831c6fe7. [DOI] [PubMed] [Google Scholar]
- 31.Demierre MF, et al. Early detection of thick melanomas in the United States: beware of the nodular subtype. Archives of dermatology. 2005;141(6):745–750. doi: 10.1001/archderm.141.6.745. [DOI] [PubMed] [Google Scholar]
- 32.Criscione VD, Weinstock MA. Melanoma thickness trends in the United States, 1988–2006. The Journal of investigative dermatology. 2010;130(3):793–797. doi: 10.1038/jid.2009.328. [DOI] [PubMed] [Google Scholar]
- 33.Lipsker DM, et al. Striking increase of thin melanomas contrasts with stable incidence of thick melanomas. Archives of dermatology. 1999;135(12):1451–1456. doi: 10.1001/archderm.135.12.1451. [DOI] [PubMed] [Google Scholar]
- 34.Richardson A, et al. The incidence and thickness of cutaneous malignant melanoma in New Zealand 1994–2004. The New Zealand medical journal. 2008;121(1279):18–26. [PubMed] [Google Scholar]
- 35.Crocetti E, Carli P. Unexpected reduction of mortality rates from melanoma in males living in central Italy. European journal of cancer. 2003;39(6):818–821. doi: 10.1016/s0959-8049(03)00056-x. [DOI] [PubMed] [Google Scholar]
- 36.Tejera-Vaquerizo A, et al. Thick melanoma: the problem continues. Journal of the European Academy of Dermatology and Venereology : JEADV. 2008;22(5):575–579. doi: 10.1111/j.1468-3083.2007.02517.x. [DOI] [PubMed] [Google Scholar]
- 37.Linos E, et al. Increasing burden of melanoma in the United States. The Journal of investigative dermatology. 2009;129(7):1666–1674. doi: 10.1038/jid.2008.423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Chamberlain AJ, Fritschi L, Kelly JW. Nodular melanoma: patients' perceptions of presenting features and implications for earlier detection. Journal of the American Academy of Dermatology. 2003;48(5):694–701. doi: 10.1067/mjd.2003.216. [DOI] [PubMed] [Google Scholar]
- 39.Kelly JW, et al. Nodular melanoma. No longer as simple as ABC. Australian family physician. 2003;32(9):706–709. [PubMed] [Google Scholar]
- 40.Rigel DS, et al. ABCDE--an evolving concept in the early detection of melanoma. Archives of dermatology. 2005;141(8):1032–1034. doi: 10.1001/archderm.141.8.1032. [DOI] [PubMed] [Google Scholar]
- 41.Demierre MF. Epidemiology and prevention of cutaneous melanoma. Current treatment options in oncology. 2006;7(3):181–186. doi: 10.1007/s11864-006-0011-z. [DOI] [PubMed] [Google Scholar]
- 42.Friedlander P, Hodi FS. Advances in targeted therapy for melanoma. Clinical advances in hematology & oncology : H&O. 2010;8(9):619–627. [PubMed] [Google Scholar]
- 43.Edlundh-Rose E, et al. NRAS and BRAF mutations in melanoma tumours in relation to clinical characteristics: a study based on mutation screening by pyrosequencing. Melanoma research. 2006;16(6):471–478. doi: 10.1097/01.cmr.0000232300.22032.86. [DOI] [PubMed] [Google Scholar]
- 44.Saldanha G, et al. Cutaneous melanoma subtypes show different BRAF and NRAS mutation frequencies. Clinical cancer research : an official journal of the American Association for Cancer Research. 2006;12(15):4499–4505. doi: 10.1158/1078-0432.CCR-05-2447. [DOI] [PubMed] [Google Scholar]
- 45.Ball NJ, et al. Ras mutations in human melanoma: a marker of malignant progression. The Journal of investigative dermatology. 1994;102(3):285–290. doi: 10.1111/1523-1747.ep12371783. [DOI] [PubMed] [Google Scholar]
- 46.van Elsas A, et al. Analysis of N-ras mutations in human cutaneous melanoma: tumor heterogeneity detected by polymerase chain reaction/single-stranded conformation polymorphism analysis. Recent results in cancer research. Fortschritte der Krebsforschung. Progres dans les recherches sur le cancer. 1995;139:57–67. doi: 10.1007/978-3-642-78771-3_5. [DOI] [PubMed] [Google Scholar]
- 47.van 't Veer LJ, et al. N-ras mutations in human cutaneous melanoma from sun-exposed body sites. Molecular and cellular biology. 1989;9(7):3114–3116. doi: 10.1128/mcb.9.7.3114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Albino AP, et al. Analysis of ras oncogenes in malignant melanoma and precursor lesions: correlation of point mutations with differentiation phenotype. Oncogene. 1989;4(11):1363–1374. [PubMed] [Google Scholar]
- 49.Albino AP, et al. Transforming ras genes from human melanoma: a manifestation of tumour heterogeneity? Nature. 1984;308(5954):69–72. doi: 10.1038/308069a0. [DOI] [PubMed] [Google Scholar]
- 50.Barbacid M. ras genes. Annual review of biochemistry. 1987;56:779–827. doi: 10.1146/annurev.bi.56.070187.004023. [DOI] [PubMed] [Google Scholar]
- 51.Omholt K, et al. NRAS and BRAF mutations arise early during melanoma pathogenesis and are preserved throughout tumor progression. Clinical cancer research : an official journal of the American Association for Cancer Research. 2003;9(17):6483–6488. [PubMed] [Google Scholar]
- 52.Reifenberger J, et al. Frequent alterations of Ras signaling pathway genes in sporadic malignant melanomas. International journal of cancer. Journal international du cancer. 2004;109(3):377–384. doi: 10.1002/ijc.11722. [DOI] [PubMed] [Google Scholar]
- 53.Lee JH, Choi JW, Kim YS. Frequencies of BRAF and NRAS mutations are different in histological types and sites of origin of cutaneous melanoma: a meta-analysis. The British journal of dermatology. 2011;164(4):776–784. doi: 10.1111/j.1365-2133.2010.10185.x. [DOI] [PubMed] [Google Scholar]
- 54.Jafari M, et al. Analysis of ras mutations in human melanocytic lesions: activation of the ras gene seems to be associated with the nodular type of human malignant melanoma. Journal of cancer research and clinical oncology. 1995;121(1):23–30. doi: 10.1007/BF01202725. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Viros A, et al. Improving melanoma classification by integrating genetic and morphologic features. PLoS medicine. 2008;5(6):e120. doi: 10.1371/journal.pmed.0050120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Broekaert SM, et al. Genetic and morphologic features for melanoma classification. Pigment cell & melanoma research. 2010;23(6):763–770. doi: 10.1111/j.1755-148X.2010.00778.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Davies H, et al. Mutations of the BRAF gene in human cancer. Nature. 2002;417(6892):949–954. doi: 10.1038/nature00766. [DOI] [PubMed] [Google Scholar]
- 58.Pollock PM, Meltzer PS. A genome-based strategy uncovers frequent BRAF mutations in melanoma. Cancer Cell. 2002;2(1):5–7. doi: 10.1016/s1535-6108(02)00089-2. [DOI] [PubMed] [Google Scholar]
- 59.Brose MS, et al. BRAF and RAS mutations in human lung cancer and melanoma. Cancer research. 2002;62(23):6997–7000. [PubMed] [Google Scholar]
- 60.Pollock PM, et al. High frequency of BRAF mutations in nevi. Nature genetics. 2003;33(1):19–20. doi: 10.1038/ng1054. [DOI] [PubMed] [Google Scholar]
- 61.Liu W, et al. Distinct clinical and pathological features are associated with the BRAF(T1799A(V600E)) mutation in primary melanoma. J Invest Dermatol. 2007;127(4):900–905. doi: 10.1038/sj.jid.5700632. [DOI] [PubMed] [Google Scholar]
- 62.Maldonado JL, et al. Determinants of BRAF mutations in primary melanomas. Journal of the National Cancer Institute. 2003;95(24):1878–1890. doi: 10.1093/jnci/djg123. [DOI] [PubMed] [Google Scholar]
- 63.Thomas NE, et al. Number of nevi and early-life ambient UV exposure are associated with BRAF-mutant melanoma. Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. 2007;16(5):991–997. doi: 10.1158/1055-9965.EPI-06-1038. [DOI] [PubMed] [Google Scholar]
- 64.Sasaki Y, et al. BRAF point mutations in primary melanoma show different prevalences by subtype. The Journal of investigative dermatology. 2004;123(1):177–183. doi: 10.1111/j.0022-202X.2004.22722.x. [DOI] [PubMed] [Google Scholar]
- 65.Lang J, MacKie RM. Prevalence of exon 15 BRAF mutations in primary melanoma of the superficial spreading, nodular, acral, and lentigo maligna subtypes. The Journal of investigative dermatology. 2005;125(3):575–579. doi: 10.1111/j.0022-202X.2005.23833.x. [DOI] [PubMed] [Google Scholar]
- 66.Johnson N, et al. A comparative study of genome-wide SNP, CGH microarray and protein expression analysis to explore genotypic and phenotypic mechanisms of acquired antiestrogen resistance in breast cancer. Breast cancer research and treatment. 2008;111(1):55–63. doi: 10.1007/s10549-007-9758-6. [DOI] [PubMed] [Google Scholar]
- 67.Vanison C, Tanna N, Murthy AS. Comparative genomic hybridization for the diagnosis of melanoma. Eur J Plast Surg. 2010;33(1):45–48. doi: 10.1007/s00238-009-0369-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Bauer J, Bastian BC. Distinguishing melanocytic nevi from melanoma by DNA copy number changes: comparative genomic hybridization as a research and diagnostic tool. Dermatol Ther. 2006;19(1):40–49. doi: 10.1111/j.1529-8019.2005.00055.x. [DOI] [PubMed] [Google Scholar]
- 69.Trolet J, et al. Genomic profiling and identification of high-risk uveal melanoma by array CGH analysis of primary tumors and liver metastases. Invest Ophthalmol Vis Sci. 2009;50(6):2572–2580. doi: 10.1167/iovs.08-2296. [DOI] [PubMed] [Google Scholar]
- 70.Mensink HW, et al. Molecular cytogenetic analysis of archival uveal melanoma with known clinical outcome. Cancer Genet Cytogenet. 2008;181(2):108–111. doi: 10.1016/j.cancergencyto.2007.12.001. [DOI] [PubMed] [Google Scholar]
- 71.White JS, et al. Correlation of comparative genomic hybridization results of 100 archival uveal melanomas with patient survival. Cancer Genet Cytogenet. 2006;170(1):29–39. doi: 10.1016/j.cancergencyto.2006.05.004. [DOI] [PubMed] [Google Scholar]
- 72.Hughes S, et al. Microarray comparative genomic hybridisation analysis of intraocular uveal melanomas identifies distinctive imbalances associated with loss of chromosome 3. Br J Cancer. 2005;93(10):1191–1196. doi: 10.1038/sj.bjc.6602834. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Kabbarah O, et al. Integrative genome comparison of primary and metastatic melanomas. PLoS One. 2010;5(5):e10770. doi: 10.1371/journal.pone.0010770. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Balazs M, et al. Chromosomal imbalances in primary and metastatic melanomas revealed by comparative genomic hybridization. Cytometry. 2001;46(4):222–232. doi: 10.1002/cyto.1131. [DOI] [PubMed] [Google Scholar]
- 75.Liotta L, Petricoin E. Molecular profiling of human cancer. Nature reviews. Genetics. 2000;1(1):48–56. doi: 10.1038/35049567. [DOI] [PubMed] [Google Scholar]
- 76.Ramaswamy S, Golub TR. DNA microarrays in clinical oncology. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2002;20(7):1932–1941. doi: 10.1200/JCO.2002.20.7.1932. [DOI] [PubMed] [Google Scholar]
- 77.Liang P, Pardee AB. Analysing differential gene expression in cancer. Nature reviews. Cancer. 2003;3(11):869–876. doi: 10.1038/nrc1214. [DOI] [PubMed] [Google Scholar]
- 78.Djuranovic S, Nahvi A, Green R. A parsimonious model for gene regulation by miRNAs. Science. 2011;331(6017):550–553. doi: 10.1126/science.1191138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Kosik KS. MicroRNAs and cellular phenotypy. Cell. 2010;143(1):21–26. doi: 10.1016/j.cell.2010.09.008. [DOI] [PubMed] [Google Scholar]
- 80.Friedman RC, et al. Most mammalian mRNAs are conserved targets of microRNAs. Genome Res. 2009;19(1):92–105. doi: 10.1101/gr.082701.108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Lim LP, et al. Microarray analysis shows that some microRNAs downregulate large numbers of target mRNAs. Nature. 2005;433(7027):769–773. doi: 10.1038/nature03315. [DOI] [PubMed] [Google Scholar]
- 82.Neveu P, et al. MicroRNA profiling reveals two distinct p53-related human pluripotent stem cell states. Cell Stem Cell. 2010;7(6):671–681. doi: 10.1016/j.stem.2010.11.012. [DOI] [PubMed] [Google Scholar]
- 83.Volinia S, et al. Reprogramming of miRNA networks in cancer and leukemia. Genome Res. 2010;20(5):589–599. doi: 10.1101/gr.098046.109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Landi MT, et al. MicroRNA expression differentiates histology and predicts survival of lung cancer. Clin Cancer Res. 2010;16(2):430–441. doi: 10.1158/1078-0432.CCR-09-1736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Jukic DM, et al. Microrna profiling analysis of differences between the melanoma of young adults and older adults. J Transl Med. 2010;8:27. doi: 10.1186/1479-5876-8-27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Dacic S, et al. miRNA expression profiling of lung adenocarcinomas: correlation with mutational status. Mod Pathol. 2010;23(12):1577–1582. doi: 10.1038/modpathol.2010.152. [DOI] [PubMed] [Google Scholar]
- 87.Chan E, et al. MicroRNA signatures differentiate melanoma subtypes. Cell Cycle. 2011;10(11):1845–1852. doi: 10.4161/cc.10.11.15777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Grichnik JM, et al. Melanoma, a tumor based on a mutant stem cell? The Journal of investigative dermatology. 2006;126(1):142–153. doi: 10.1038/sj.jid.5700017. [DOI] [PubMed] [Google Scholar]
- 89.Klein WM, et al. Increased expression of stem cell markers in malignant melanoma. Mod Pathol. 2007;20(1):102–107. doi: 10.1038/modpathol.3800720. [DOI] [PubMed] [Google Scholar]
- 90.van der Rhee JI, et al. Clinical and histologic characteristics of malignant melanoma in families with a germline mutation in CDKN2A. Journal of the American Academy of Dermatology. 2011;65(2):281–288. doi: 10.1016/j.jaad.2010.06.044. [DOI] [PubMed] [Google Scholar]
- 91.Pavey SJ, et al. Loss of p16 expression is associated with histological features of melanoma invasion. Melanoma research. 2002;12(6):539–547. doi: 10.1097/00008390-200212000-00003. [DOI] [PubMed] [Google Scholar]
- 92.Wang YL, et al. Immunohistochemical detection of CDK4 and p16INK4 proteins in cutaneous malignant melanoma. The British journal of dermatology. 1996;134(2):269–275. [PubMed] [Google Scholar]
- 93.Reed JA, et al. Loss of expression of the p16/cyclin-dependent kinase inhibitor 2 tumor suppressor gene in melanocytic lesions correlates with invasive stage of tumor progression. Cancer research. 1995;55(13):2713–2718. [PubMed] [Google Scholar]
- 94.Radhi JM. Malignant melanoma arising from nevi, p53, p16, and Bcl-2: expression in benign versus malignant components. Journal of cutaneous medicine and surgery. 1999;3(6):293–297. doi: 10.1177/120347549900300603. [DOI] [PubMed] [Google Scholar]
- 95.Funk JO, et al. p16INK4a expression is frequently decreased and associated with 9p21 loss of heterozygosity in sporadic melanoma. Journal of cutaneous pathology. 1998;25(6):291–296. doi: 10.1111/j.1600-0560.1998.tb01748.x. [DOI] [PubMed] [Google Scholar]
- 96.Talve L, et al. Loss of expression of the p16INK4/CDKN2 gene in cutaneous malignant melanoma correlates with tumor cell proliferation and invasive stage. International journal of cancer. Journal international du cancer. 1997;74(3):255–259. doi: 10.1002/(sici)1097-0215(19970620)74:3<255::aid-ijc4>3.0.co;2-y. [DOI] [PubMed] [Google Scholar]
- 97.Roth JM, et al. Inhibition of experimental metastasis by targeting the HUIV26 cryptic epitope in collagen. The American journal of pathology. 2006;168(5):1576–1586. doi: 10.2353/ajpath.2006.050176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Xu J, et al. Proteolytic exposure of a cryptic site within collagen type IV is required for angiogenesis and tumor growth in vivo. The Journal of cell biology. 2001;154(5):1069–1079. doi: 10.1083/jcb.200103111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Gagne PJ, et al. Temporal exposure of cryptic collagen epitopes within ischemic muscle during hindlimb reperfusion. The American journal of pathology. 2005;167(5):1349–1359. doi: 10.1016/S0002-9440(10)61222-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Cretu A, et al. Disruption of endothelial cell interactions with the novel HU177 cryptic collagen epitope inhibits angiogenesis. Clinical cancer research : an official journal of the American Association for Cancer Research. 2007;13(10):3068–3078. doi: 10.1158/1078-0432.CCR-06-2342. [DOI] [PubMed] [Google Scholar]
- 101.Ng B, et al. Shedding of distinct cryptic collagen epitope (HU177) in sera of melanoma patients. Clin Cancer Res. 2008;14(19):6253–6258. doi: 10.1158/1078-0432.CCR-07-4992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.A P, L L, B P. AY 43-9006 (sorafenib-BAY) alters proliferation pathways and mutant specific-PCR (MS-PCR) improves detection of BRAF mutations in metastatic melanoma (MM) Journal of Clinical Oncology. 2007;25 p. abstract 8534. [Google Scholar]
- 103.Romano E, et al. Treatment implications of the emerging molecular classification system for melanoma. Lancet Oncol. 2011;12(9):913–922. doi: 10.1016/S1470-2045(10)70274-6. [DOI] [PubMed] [Google Scholar]
- 104.Carvajal RD, et al. KIT as a therapeutic target in metastatic melanoma. JAMA. 2011;305(22):2327–2334. doi: 10.1001/jama.2011.746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Guo J, 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;29(21):2904–2909. doi: 10.1200/JCO.2010.33.9275. [DOI] [PubMed] [Google Scholar]
- 106.Van Raamsdonk CD, et al. Mutations in GNA11 in uveal melanoma. N Engl J Med. 2010;363(23):2191–2199. doi: 10.1056/NEJMoa1000584. [DOI] [PMC free article] [PubMed] [Google Scholar]
