Introduction
Melanoma is increasing in incidence. In the United States in 2014, there were over 76,000 new melanoma cases and over 9,000 melanoma-related deaths [1]. Current American Joint Committee on Cancer’s (AJCC) staging for cutaneous melanoma is based on primary tumor thickness and presence of ulceration, mitoses, nodal spread, and distant metastases as determinants of prognosis. However, survival outcomes can vary widely even within the same stage. Given the clinical and biological heterogeneity of primary melanoma, new prognostic tools are needed to more precisely identify the patients most likely to develop advanced disease. Such tools would affect clinical surveillance strategies and aid in patient selection for adjuvant therapy. Here, we discuss the evolution of primary melanoma prognostic determinants, their limitations, and the current literature of melanoma prognostic markers from a molecular and immunologic perspective.
Evolution of Cutaneous Melanoma Staging Over Time
In the mid-twentieth century, melanoma staging, and thus prognosis, were driven by thickness and level of invasion. In 1969, Clark et al. [2] described five levels of primary melanoma tumor invasion that inversely correlated with overall survival (OS) in a study of 208 primary melanomas [2]. Breslow et al. [3] also observed that tumor thickness combined with Clark’s level produced the most accurate prognoses in a cohort of 98 patients with primary melanomas examined for Clark’s level, tumor thickness, and cross-sectional area (thickness times diameter). No melanoma recurrences or metastases from primary tumors < 0.76 mm thick in Clark’s levels II and III occurred within 5 years of follow-up, thus 0.76 mm was recommended as a cut-off point to stratify patients for lymph node dissection [3].
Since these seminal studies, melanoma staging has continued to evolve as patient cohorts have expanded and additional prognostic features have been described. Today, the AJCC oversees melanoma staging guidelines, which incorporate tumor thickness and other histopathological parameters into the “TNM” (tumor, node, metastasis) criteria used for most solid tumors. In 2002, the AJCC made significant changes to the melanoma staging system, including increasing thickness thresholds for T staging from 0.75, 1.5, and 4 mm to 1, 2, and 4 mm, and including ulceration status as a second determinant of T staging, based on a multivariate analysis of 13,581 primary melanoma patients [4–5]. Additionally, in a subset of 4,750 patients without clinically-detected nodal metastases, Clark’s level was found to be predictive of outcome in thin melanomas (<1 mm), whereas ulceration was the most prognostic factor in thicker melanomas (> 1 mm) [5]. Other AJCC staging updates at that time included shifting thick T4 melanomas from stage IIIA to stage IIC, inclusion of satellite metastases in N staging, and exclusion of the size of nodal metastases, which was no longer found to be prognostic [5, 6, 7].
In 2010, mitotic rate (≥ 1/mm2) replaced Clark’s level to characterize T1 melanomas when a multivariate analysis of 4,861 T1 primary melanomas demonstrated that thickness, mitotic rate, and ulceration significantly predicted survival outcomes, while Clark’s level did not [7]. Inclusion of mitoses in melanoma staging was considered practice-changing. Due to a 10% risk of sentinel lymph node (SLN) positivity for primary tumor thickness of 0.76–1 mm with mitoses, surgeons might now be prompted to offer SLN biopsy to patients with thinner melanomas if mitoses are present [8]. Additionally, a lymph node was now defined as positive if any isolated tumor cells were detected histologically or by immunohistochemistry (IHC), whereas at least 0.2 mm of tumor was previously required for positivity. This also changed prognostic discussions that clinicians have with these patients, who are now being upstaged to at least IIIA melanoma for even a single tumor cell noted in the lymph node, conferring a worse prognosis [7–8].
Limitations of AJCC Staging System
While the AJCC cutaneous melanoma staging system has evolved over time to more accurately reflect patient prognosis, improvements are still needed. TNM staging does not always reliably predict patient outcomes by stage. For example, stage I melanomas ≤ 1 mm comprise the majority (∼80%) of melanoma diagnoses reported to the SEER Cancer Registry and carry an excellent prognosis with 5 year OS rates of 92–97% [9–10]. However, 15% of these patients die from melanoma [10]. Furthermore, stage IIC melanomas have a worse 5-year OS (53%) compared to stage IIIA melanomas (78%) and stage III patients have highly variable 5-year OS rates, ranging from 78%, 59%, and 50% for stage IIIA, IIIB, and IIIC, respectively [9, 11].
It has been hypothesized that adding measurements of host anti-tumor immune response and/or molecular features to staging may improve primary melanoma prognostication. New tumor-, host-, and/or blood-based biomarkers to address the inconsistencies of an ever-evolving, imperfect staging system are critically needed. We review the literature on prognostic molecular and immunologic markers in primary cutaneous melanoma, their associations with clinicopathologic and survival outcomes, and their potential for incorporation into current staging models.
Tissue-based Molecular Genetic Markers
Identifying tissue-based prognostic molecular markers in melanoma has been challenging. One major limitation is that most primary melanomas are preserved as formalin-fixed paraffin-embedded (FFPE) rather than fresh frozen tissues due to the small size of the specimens. High quality genetic material is less commonly extractable from FFPE, precluding their use with early generation sequencing methods [12]. Relative stability of mRNA is also inconsistent in FFPE samples [13]. However, improved extraction methods, newer technologies, and novel, stable RNA molecules, such as microRNA, have allowed exome- or genome-wide mutational analysis or RNA expression profiling from small amounts of genetic material isolated from FFPE [12]. Such technical and biological advances have yielded new insights into melanoma biology and new molecules to assess prognostically.
Genes that were initially identified as genetically altered drivers of melanoma tumorigenesis (BRAF, NRAS, PTEN, etc.) were captured in candidate-based analyses focused on known proliferation and survival pathways. Next generation sequencing (NGS) has afforded an unprecedented depth of knowledge of the mutational landscape of entire melanoma genomes, which has confirmed and expanded on these findings [14, 15, 16, 17]. However, challenges identifying genetic drivers of melanomagenesis and progression remain. A comprehensive analysis of whole exome sequencing (WES) data from 21 tumor types (TCGA and Broad Institute) calculated that sequencing information for >5000 samples is required to identify all enriched mutated genes present in greater than 2% of melanomas at 90% power [18], highlighting the particular challenge of interpreting sequencing data for melanoma. Moreover, among the genes nominated from melanoma NGS studies, few reports have associated individual mutations with clinical outcomes or studied their use as prognostic biomarkers in cutaneous melanoma. None-the-less, there has been a movement to genetically define melanomas, and here we review associations with patient prognosis for commonly mutated genes in melanoma.
BRAF
Activating BRAF mutations are present in 50%-60% of melanoma, the majority of which are V600E (∼90%) or V600K (∼10%) [19]. BRAF mutations have been described in high frequencies in benign nevi, although occurring less frequently in radial growth phase (RGP) compared to vertical growth phase (VGP) melanomas, suggesting that oncogenic BRAF mutations associate with melanoma progression more strongly than with melanomagenesis [20].
There are conflicting data on BRAF mutation status as an independent prognostic marker in melanoma. A meta-analysis found that patients with BRAF mutant tumors have a 1.7 fold increased risk of death compared to BRAF wild-type (WT) tumors [21]. In contrast, many small studies of primary melanomas cite no independent association between BRAF mutation and survival (Table 1) [22–25], though Ellerhorst et al. did find that BRAF mutant primary melanomas may have more adverse pathological characteristics (increased thickness and ulceration) compared to WT tumors, and tend to present clinically with a higher disease stage [25].
Table 1.
Mutation | Study Population | Clinicopathological associations |
Survival associations | Reference |
---|---|---|---|---|
BRAF | 89 primary melanomas |
Truncal location, Superficial spreading histologic subtype, younger age |
No association with development of metastases or MSS |
Maldonado et al. 2003 |
223 primary melanomas |
Higher rate of ulceration |
No association with survival outcomes |
Ellerhorst et al. 2010 | |
249 primary melanomas |
Worse RFS in multivariate analysis adjusting for age, sex, tumor site, tumor stage, mitosis, and ulceration, but not in a second multivariate analysis which also controlled for thickness |
Devitt et al. 2011 | ||
Meta-analysis of 4 melanoma studies |
1.7 fold increased risk of death |
Ardekani et al. 2012 | ||
51 primary nodular melanomas and 18 paired metastases |
Higher rate of ulceration in primary tumor |
No association with RFS or MSS |
Akslen et al. 2005 | |
79 melanoma nodal metastases and their associated primaries |
Worse survival outcome | Mann et al. 2013 | ||
912 primary melanomas |
Younger age, Superficial spreading histologic subtype, present mitoses |
3 fold-increased risk of melanoma-related death compared to WT for high risk patients after adjustment for age, sex, study center, anatomic site, and TIL grade |
Thomas et al. 2015 | |
NRAS | 223 primary melanomas |
Thicker, upper extremity location |
No association with OS | Ellerhorst et al. 2010 |
249 primary melanomas |
Thicker, higher mitotic rates, trend towards higher proliferation as measured by Ki-67 |
Association with shorter MSS | Devitt et al. 2011 | |
51 primary melanomas and 18 paired metastases |
No association with RFS or MSS |
Akslen et al. 2008 | ||
79 melanoma nodal metastases and their associated primaries |
Worse survival outcome | Mann et al. 2013 | ||
912 primary melanomas |
Lower TIL grade, present mitoses, anatomic site other than scalp/neck |
3 fold-increased risk of melanoma-related death compared to WT for high risk patients after adjustment for age, sex, study center, anatomic site, and TIL grade |
Thomas et al. 2015 |
MSS = melanoma-specific survival; DFS = disease-free survival; RFS = recurrence-free survival; OS = overall survival; WT = wild type; TIL = tumor-infiltrating lymphocytes
NRAS
NRAS mutations, which occur in about 15% of cutaneous melanomas, most commonly at codon 61 (Q61K or Q61R), also lead to constitutive activation of the MAPK pathway [25, 26]. Similar to BRAF mutations, prognostic implications of NRAS mutations have varied in the literature (Table 1).
Devitt et al. prospectively studied 249 primary melanomas and found that patients with NRAS mutant melanoma (codon 61) had shorter melanoma-specific survival (MSS) (HR=2.51, p=0.05), and trended towards worse recurrence-free survival (RFS) in multivariate analysis (HR 2.2, p=0.09), but NRAS mutation had no impact on OS (HR 1.37, p=0.37) [27]. NRAS mutated primary melanomas tended to be thicker, have more mitoses, and more frequently be of nodular subtype when compared to BRAF mutant or NRAS/BRAF WT tumors [26]. Multiple other studies of primary melanomas have shown no association of NRAS mutation status with RFS, MSS, or OS [24–25].
Most recently, investigators from the Genes, Environment, and Melanoma (GEM) study reported on associations of BRAF and NRAS status with clinicopathologic variables and MSS in 912 primary melanoma patients. The 7.6 year median follow-up time was completed prior to 2011, which limits the possible confounders of targeted or immunotherapy administration in patients who progressed. While there were no differences in 5 year OS between patients with BRAF mutant, NRAS mutant or WT tumors, there was a 3-fold increase in melanoma-related death in BRAF and NRAS mutated patients specifically in the high-risk category (T stage T2b or above) compared to patients with WT tumors [28].
KIT
KIT is a type III receptor tyrosine kinase for stem cell factor (SCF) and functions as an oncogene, involved in melanocytic differentiation, migration, proliferation, and survival [27, 29]. KIT mutations or gene amplifications occur in 10–25% of melanomas arising from areas of acral, mucosal, or sun-damaged skin, are rare (0–2%) in cutaneous melanomas without chronic sun-damage, and can rarely co-exist with BRAF mutations [27, 30–31]. There is limited information on the prognostic significance of KIT in melanoma, although melanoma subtypes associated with KIT mutation have a worse overall prognosis compared to more common cutaneous melanomas [32].
PTEN
PTEN is a tumor suppressor that functions as a phosphatase that when inactivated, results in constitutive activation of PI3K/AKT signaling and increased cell survival [33]. Up to 10% of cutaneous melanomas harbor loss-of-function alterations in PTEN, which include mutations, deletions, and promoter methylation [34]. In a study of 127 primary melanomas, decreased PTEN protein abundance was associated with primary tumor ulceration but not survival outcomes [33]. In contrast, a recent study reported that tumors with PTEN methylation have worse OS compared to unmethylated PTEN tumors [35]. PTEN is inactivated by multiple mechanisms and future outcome association studies should ideally incorporate all mechanisms that contribute to and define PTEN-inactivated tumors.
Newly identified genetic mutations
NGS studies have expanded the known mutational landscape of melanoma and nominated novel mutations as drivers of melanoma tumorigenesis. Literature review has revealed only a few studies so far that have examined prognostic associations of these newly identified genetic mutations.
A mix of recurrent and non-recurrent mutations in the protein phosphatase PPP6C were recently identified in 9–12% of melanomas [15, 17]. Consistent with previous reports, our group recently reported PPP6C mutations in 10.7% (25/233) and 10.4% (8/77) of primary and metastatic melanoma samples, respectively [36]. Interestingly, we documented that in AJCC stage I-III patients, those with stop mutations trended toward worse RFS, while those with non-stop mutations trended toward improved RFS compared with that of patients with PPP6C WT tumors.
Though present in only 5–9% of melanomas, mutations in RAC1 are predominantly P29S missense mutations that yield aberrant RAC activity, which drives melanoma proliferation and tumorigenesis [15, 17]. A recent study found that only 3.3% (27/814) of primary cutaneous melanomas harbor RAC1 P29S mutations [37], suggesting that these mutations occur as later events in the natural history of melanoma and may be key drivers of metastatic spread. Consistent with this notion, RAC1 mutations were associated with nodal spread at diagnosis, in addition to other negative prognostic indicators. Unfortunately, this study did not report associations of RAC1 P29S mutation status with distant metastasis or patient outcomes, precluding definitive assessment.
Recurrent mutations in regulatory regions of several genes that aberrantly activate expression of these genes have also recently been described [38–40]. Of these, recurrent mutations in the promoter of TERT, which create novel ETS family transcription factor binding sites, have been documented in up to 70% of melanoma patient samples. TERT promoter mutations result in increased telomerase expression, thus favoring chromosomal stability through maintenance of telomere length, which may improve the fitness of cancer cells harboring such mutations. Two recent studies found that TERT promoter mutations (43% and 22% of patients) in primary melanomas associated with key prognostic parameters (tumor thickness, ulceration, and mitotic rates), and poor disease free survival (DFS) and OS [41–42].
Collectively, these findings support the potential relevance of newly identified genetic mutations as clinically useful prognostic factors, and further studies to analyze associations of genetic mutations with patient outcomes are warranted. Increased collaboration among research centers to stimulate sample and data pooling and to standardize statistical methods and analyses will aid in a more definitive assessment of genetic mutations as prognostic markers.
Expression of protein-coding genes
Expression level-based analyses of mRNAs and proteins provide immensely alluring molecular markers, as there are tens of thousands of potential markers. In theory, proteins have a distinct advantage of being the functional end products of protein-coding genes. Moreover, their stability, modifications, and localization multiply their potential as prognostic markers. However, tools to measure proteins genome-wide are still developing and protein-based (immunohistochemical) biomarker studies thus far reported in melanoma have largely taken candidate approaches. Several comprehensive reviews and/or meta-analyses have examined protein-based biomarker studies in melanoma [43–46], thus we refer readers to these excellent manuscripts instead of addressing them in depth here.
Briefly, we do note 3 important multi-marker prognostic signature studies that have been developed with the use of tissue microarray-based (TMA) immunohistochemistry [43, 47]. Gould Rothberg et al. built a 5-protein signature (high non-nuclear/nuclear ATF2, high nuclear p21, high total β-catenin, low non-nuclear/nuclear p16, and low total fibronectin; from 20 candidates evaluated) indicative of favorable prognosis (OS) using immunofluorescence-based automated quantitative analysis (AQUA) histochemistry in 192 AJCC stage II FFPE specimens [43]. They validated this scoring method in a set of 226 additional primary melanoma patients. In contrast, Meyer et al. developed a 7-marker signature (Bax, Bcl-X, PTEN, COX-2, β-catenin loss, MTAP loss, and presence of CD20+ B-lymphocytes, from 70 candidates evaluated) indicative of poor prognosis (RFS and OS) in training (n = 362) and validation (n = 225) patient cohort sets [46]. Both studies identified β-catenin as a protective prognostic indicator. Of the remaining 6 reported by Meyer et al., none were evaluated in Gould Rothberg et al. Of the remaining 4 reported by Gould Rothberg et al., p21 and p16 were evaluated by Meyer et al., though this study did not examine prognostic impact of nuclear staining only (p21) or non-nuclear/nuclear ratios (p16).
Additionally, in a cohort of 395 primary melanomas, Kashani-Sabet et al. confirmed a protein-based prognostic signature containing NCOA3, SPP1 (osteopontin), and RGS1, which they previously identified as prognostic in a cDNA microarray study [48]. A multimarker index score of expression was found to be an independent predictor of disease-specific survival (DSS) and sentinel lymph node status, and was validated in an independent patient cohort. Interestingly, osteopontin has been demonstrated as a prognostic factor across multiple studies, as we detail below.
In contrast to protein, mRNA-based biomarkers can be robustly and quantifiably measured genome-wide using several methodologies and should be easily translatable into a highly quantitative RT-qPCR-based clinical assay. Such studies, however, are limited by poor quality mRNA obtained in archival FFPE tissues [13, 49]. Moreover, contamination from infiltrating cells and surrounding microenvironment likely result in significant expression bias and complicate cross-study comparisons. There are numerous reports documenting mRNA expression profiling of melanoma cell lines, metastatic melanoma tissues or comparisons across melanoma progression (nevi, primary, metastasis) [50–55]. However, only a few studies have examined gene expression profiling or developed mRNA-based prognostic signatures from primary melanoma tissues of different outcomes (Table 2).
Table 2.
Selected Genes |
Study Population and Method |
Clinicopathological or survival associations | Reference | |
---|---|---|---|---|
mRNA |
MCM3, MCM4, MCM6, KPNA2 |
83 fresh- frozen primary melanomas microarray IHC |
254 genes differentially expressed based on less than or greater than 4-year distant metastasis-free survival (n = 58). IHC validation for 8 of 23 genes analyzed, with 4 associating with overall survival in validation cohort (n = 176). |
Winnepennninckx et al. 2006 |
KRT9, DCD, PIP, SCGB1D2, SCGB2A2, COL6A6, KBTBD10, ECRG2, HES6 |
41 fresh Primary melanomas microarray RT-qPCR |
92 genes differentially expressed in primary melanomas between short- (OS < 4years, n = 20) and long-term (OS > 5years, n =21) survivors. |
Brunner et al. 2008 | |
RT-qPCR analysis of previously identified 92 genes from Brunner et al. (2008). 11 genes associated with OS in univariate Cox analyses were further measured in 53 additional primary tumors. 9 genes retained significant association with OS. |
Brunner et al. 2013 | |||
CD24, EVL | 116 non- metastasizing (stage I/II) and 72 metastasized (stage III/IV) Primary melanomas SAGE |
Top 50 differentially expressed genes used for class prediction in 28 independent primary tumors. CD24 and EVL, alone, or combined with Breslow thickness provided strongest predictor of outcome. |
Gschaider et al. 2012 | |
BAP1, MGP, SPP1, CXCL14, CLCA2, S100A8, S100A9, BTG1, SAP130, ARG1, KRT6B, KRT14, GJA1, EIF1B, RBM23, LTA4H, CRABP2, DSC1, ROBO1, AQP3, SPRR1B, CST6, TRYP1, ID2, TACSD2, PPL |
268 primary Cutaneous melanomas, with semi- random stratification to two cohorts RT-qPCR |
20 genes selected from mining of public datasets for genes consistently altered in primary vs. metastatic melanoma, 5 additional genes prognostic in uveal melanoma, BAP1 (3’ and 5’ end), and 1 gene originally selected as 1 of 4 reference genes comprise RT-qPCR- based 28-gene prognostic signature. Radial Basis Machine modeling stratified patients into low- or high- risk groups, which clearly separated in Kaplan-Meier disease-free survival curves. Multivariate Cox analyses with key prognostic parameters indicated this signature might be independently prognostic. |
Gerami et al. 2015 | |
Same as above |
Subset of above |
Follow-up study examined 28-gene signature in 217 primary melanoma patients who had undergone SLNB. In this patient cohort, the expression signature performed as well or better as an independent predictor of DFS, DMFS, and OS. |
Gerami et al. 2015 | |
HOXA1 Targets genes |
N/A |
HOXA1 pro-invasion transcriptional signature identified. Signature classified patient samples in Winnepenninckx et al. |
Wardwell-Ozgo et al. 2014 | |
CD2 | 47 and 48 stage II/III FFPE primary melanomas Nanostring |
A 53-gene panel of immunoregulatory genes in stage II and III melanoma patients predicted non-progression, RFS, and DSS |
Sivendran et al. 2014 | |
miRNA |
miR-382, miR-516b |
92 and 119 FFPE primary melanomas miRNA microarray |
Reported miRNAs associated with tumor thickness and recurrence status, but focused on functional contributions to metastatic spread. Two metastasis suppressor miRNAs (miR-382, miR-516b) were not clearly associated with survival parameters in multivariate analyses (data not reported). |
Hanniford et al. 2015 |
134 miRNA | 82 and 97 FFPE primary melanomas miRNA microarray |
Reported miRNA associated with nodular vs. superficial spreading melanoma subtypes in primary melanoma tissues. Prognostic implications within each subtype were not reported. |
Poliseno et al. 2012 | |
miR-200 family, miR-203 |
23 fresh- frozen primary melanomas RT-qPCR |
11 miRNA (of 159 measured) associated with tumor thickness. In validation patient set, study observed progressive loss of miR-200a, c, and miR-203 expression from nevi to primary to metastasis, and at the invasive front of primary tumors. |
Van Kempen et al. 2012 | |
miR-30d | 92 FFPE Primary melanomas miRNA microarray |
Pro-metastatic miR-30d expression associated with stage, thickness, histological subtype, and RFS and OS in primary melanoma samples. |
Gaziel-Sovran et al. 2011 | |
miR-1908, miR-199a- 3p, miR- 199a-5p |
71 primary melanomas RT-qPCR |
miR-1908, miR-199a-3p and −5p were up-regulated in a melanoma metastasis in vivo selection model. Studyobserved univariate associations with metastasis- free survival in patient samples. |
Pencheva et al. 2012 | |
miR-15b | 128 primary melanomas RT-qPCR |
Expression of pro-proliferative miR-15b associated with poor RFS and OS in multivariate analyses. miR-34a and miR-210 did not associate with survival parameters. |
Satzger et al. 2010 |
Winnepenninckx et al. reported mRNA expression profiles for 83 fresh-frozen primary cutaneous melanomas, in which they defined a set of 254 genes expressed differentially based on 4-year distant metastasis-free survival (DMFS) and identified high expression of minichromosome maintenance (MCM) genes as key predictors of reduced DMFS [55]. A follow-up study re-analyzed this data in the context of biological pathways and found that high expression of DNA repair genes was associated with poor DMFS [56]. Interestingly, another recent study, which defined a pro-invasive HOXA1 transcriptional signature, found that patients from the Winnepenninckx et al. study with a ‘high’ HOXA1 signature had poor DMFS, adding an intriguing underlying transcriptional mechanism to those original expression findings, though they did not note if DNA repair pathway genes were highly represented in their signature [57]. A second primary cutaneous melanoma expression profiling study identified 92 genes associated with OS [58], and a subsequent follow-up study, which further assessed these 92 genes, reported a 9-gene signature (6 protective and 3 unfavorable) associated with OS in a 91 patient cohort [59]. A recent report utilized serial analysis of gene expression (SAGE) to identify differentially expressed transcripts between 116 primary tumors that had not metastasized (stage I/II) and 72 primary tumors that had already spread at initial diagnosis (stage III/IV) [60]. Subsequent multivariate class prediction analyses of the top 50 differentially expressed genes identified CD24 and EVL as strong predictors of metastatic risk in an independent set of 28 primary melanoma samples. Conway et al. [61] performed DASL (cDNA-mediated annealing, selection, extension, and ligation) expression profiling of 502 cancer genes for >350 FFPE cutaneous primary melanomas from two patient cohorts, identifying high osteopontin (SPP1) expression associated with reduced RFS, which confirmed previous findings [50, 62–63]. Osteopontin has been implicated in cell motility, proliferation, and in activation of the NF-kappa B pathway, which may add mechanistic explanations of its prognostic capacity [62]. A subsequent re-analysis of these data by the same group identified increased expression of DNA repair genes as associated with poor recurrence-free survival [64], consistent with the described re-analysis of Winnepenninckx et al. data [56].
Collectively, these studies reported from tens to hundreds of genes associated with metastatic risk and/or survival outcomes in primary cutaneous melanoma. No reported genes overlap in more than two studies. Of the 50 genes reported in Gschaider et al. [60], 7 genes overlap with Winnepenninckx et al. [55], though, interestingly, the cellular source of these RNAs is likely to be keratinocytes, endothelial cells, or infiltrating immune cells, which would support that microenvironment and/or host response are key prognostic markers for patient outcomes in primary cutaneous melanoma (Supplementary Table 1). Five genes from a combined list from Conway et al. and Jewell et al. overlap with Winnepenninckx et al., and osteopontin overlapped between the Conway/Jewell and Gschaider et al. studies. None of the 92 genes originally reported to be associated with OS in Brunner et al. [58] and further assessed in a follow-up study [59] overlapped with genes reported in the other studies, though we note that none of the other studies defined genes directly associated with OS.
Two recent studies by Gerami and colleagues selected a set of 28 gene targets (melanoma metastasis-associated genes identified from data mining, previously defined prognostic uveal melanoma genes, and BAP1) for analysis as a prognostic molecular signature in melanoma [65–66] . In the first study, the gene set was measured in 268 primary melanoma patient samples, which were mostly randomly stratified to training (n=164) and validation (n=104) cohorts [66]. In the second, analysis of gene set measurements was restricted to patients who had undergone sentinel lymph node biopsy (SLNB) procedure. Prognostic modeling was performed using Radial Basis Machine (RBM) modeling to separate patients into two risk classes that were plotted in KM survival curves and compared with AJCC stage, Breslow depth, ulceration, mitotic rate, and age or SLN status in univariate and multivariate analyses. Each study found the defined signature to be an independent predictor of metastatic risk. These studies are the basis of a clinical prognostic test (DecisionDx) offered by Castle Biosciences, which we discuss below.
Expression of microRNA
A newer class of RNA, microRNA (miRNA), has been of particular interest to cancer researchers over the last decade. While fewer in number than mRNA and not translated to protein, miRNA are ideal potential biomarkers because they are quantifiable genome-wide and highly stable in FFPE tissues [67–68], although they are equally affected by sample purity as mRNA. MiRNA expression profiling has typically been performed using microarray (genome-wide, but inconsistently quantitative) or RT-qPCR (fewer miRNA, but highly quantitative), though newer technologies, including RNA-sequencing and Nanostring, allow highly quantitative genome-wide analysis. Early studies of global miRNA expression in primary melanoma did not focus on prognosis, but rather on identification of miRNA that play functional roles in melanoma tumorigenesis [69–73]. Our group reported 18 miRNA associated with post-recurrence survival [74], with a subset of 6 of these microRNA (mir-150, mir-342-3p, mir-455-3p, mir-145, mir-155, and mir-497) defined as a post-recurrence survival signature. Interestingly, miR-342-3p is located in an intron of its host gene EVL, which was recently reported to be associated with metastatic risk in primary melanoma [60]. A recent study replicated the prognostic importance of miR-150 and miR-142 (from the 18 associated with post-recurrence survival [74]) in stage III lymph node metastases and patient sera [75].
We recently reported identification of metastasis-suppressive miRNA that we originally identified as differentially expressed in array-based global miRNA profiling of primary cutaneous melanoma tissues (2 cohorts containing 92 and 119 patient samples) [76]. We found these miRNA to be associated with thickness, recurrence status, and/or RFS. From this data, we defined a 4-miRNA signature (miR-150, miR-15b, miR-16-5p, and miR-374b-3p) that, in combination with AJCC stage, was predictive of development of brain metastasis [77]. Our group also reported a series of 134 miRNAs differentially expressed in nodular (n = 56) vs. superficial spreading melanoma (n = 26) histological subtypes, controlling for stage [78]; however, the possibility that selected miRNA signatures have independent prognostic importance for these subtypes has not yet been explored.
Van Kempen et al. performed expression profiling of 159 miRNAs by RT-qPCR in 23 fresh-frozen melanoma samples [79]. 11 miRNAs associated with tumor thickness in this patient cohort. In a validation patient set, expression analysis of these 11 and further in situ hybridization revealed loss of miR-200 family (miR-200a and c) and miR-203 expression during melanoma progression and at the invasive front of primary tumors. Of the miRNA we found consistently correlated with tumor thickness in our patient cohorts (n = 27), 10 were measured in the van Kempen study. 4 of 10 (miR-200b and c, miR-203, and miR-205) correlated with thickness, and each maintained the same direction as in our data (Supplementary Table 2). Of the 11 miRNA the van Kempen study reported, there was evidence of a correlation with thickness for 9 in at least 1 of our data sets, and all correlation directions were maintained between studies.
Several other studies identified miRNA that play functional roles in the proliferative or invasive/metastatic capacity of melanoma cells, based on analysis of miRNA expression and associations with patient outcomes. Our group previously reported that high expression in primary melanoma tissues of miR-30d, a pro-metastatic miRNA that regulates glycosylation and immune suppression in melanoma, associates with advanced stage, increased thickness, nodular histological subtype, and poor RFS and OS [80]. Another recent study used in vivo selection in mice to generate highly metastatic melanoma cell line variants [81]. MiR-1908, miR-199a-3p, and miR-199a-5p, which were highly up-regulated in these metastatic variants and critical drivers of melanoma progression, were inversely associated with metastasis-free survival in a cohort of 71 primary melanoma samples, though multivariate analyses were not performed and patient characteristics were not reported.
Several reports have defined loss of miR-211 expression as a key feature of melanoma cells, and its over-expression is suppressive of melanoma cell proliferation and invasion in vitro [71, 73, 82]. Surprisingly, however, no studies have yet reported associations of miR-211 expression in primary tumors with patient outcomes. In this regard, one of our recently published datasets, which included a set of congenital nevi, is supportive of decreased abundance of miR-211 in primary melanoma compared to nevi [76]. However, miR-211 expression in these primary tumors did not clearly associate with prognostic parameters or patient outcome in these data (not shown). Finally, Satzger et al. reported that high expression of the pro-proliferative miRNA miR-15b in a set of 128 primary melanoma tissues significantly associated with poor outcomes (RFS and OS) in multivariate analyses [83].
Overall, these studies demonstrate potential utility of miRNA expression-based prognostic assays, however, there is limited overlap of the key miRNA reported as prognostic in these studies and sample sizes remain modest. miR-150 and miR-15b are the most consistently associated with prognosis in primary and metastatic melanoma, based on tissue and serum (described below) studies.
Immune Markers of the Host
Tumor-infiltrating lymphocytes
Immunologic markers are being studied as prognostic tools that may enhance outcome predictions and be incorporated into traditional TNM melanoma staging. For example, the Immunoscore has been designed to prognosticate early stage colon cancer patients based on CD3+, CD8+, and CD45+ lymphocyte densities within tumors. High immunoscores in colon cancer are associated with improved RFS and OS [84]. Tumor-infiltrating lymphocytes (TILs) have been investigated across multiple malignancies for decades as a surrogate for anti-tumor immune response, however current cancer staging systems do not incorporate TIL status.
Debate exists if TIL status is clearly associated with melanoma prognosis (Table 3). Clark et al. reviewed 98 primary melanomas for 8 pathologic variables, including tumor-infiltrating lymphocytes (TILs) which were defined as one of three grades as measured in the vertical growth phase (VGP): brisk (lymphocytes infiltrate the entire VGP or tumor base), non-brisk (lymphocytes are focally present in the VGP), or absent (lymphocytes may be present in fibrotic areas or near vessels, but do not infiltrate the VGP). TIL grade significantly correlated with 8-year OS [84]. Since then, data in the literature both supports [86–89] and refutes [90–93] the prognostic relevance of absent, non-brisk, and brisk TILs in primary melanomas.
Table 3.
Number of cases |
Survival Associations | Comments | Reference |
---|---|---|---|
264 | Predicts 8 year OS (brisk 88%, non-brisk 75%, absent 59%) |
Clark et al. 1989 | |
548 | No survival differences | Compared absent to present TILs only |
Barnhill et al. 1996 |
285 | Predicts 5 year OS (brisk 77%, non-brisk 53%, absent 37%) |
Clemente et al. 1996 | |
259 | Predicts 5 year OS (brisk 100%, non-brisk and absent 71%) |
Groups non-brisk and absent TILs in their analysis |
Tuthill et al. 2002 |
887 | No survival differences | Groups non-brisk and brisk TILs in their analysis |
Taylor et al. 2007 |
1251 | No survival differences | Mandala et al. 2009 | |
515 | Predicts 5 year OS (brisk 95%, non-brisk 84%) |
Did not address absent TILs |
Burton et al. 2011 |
1865 | Predicts 5 year OS | Used their own TIL grading system assigning a score from 0 to 3 |
Azimi et al. 2012 |
2845 | Predicts 5 year OS (brisk 97%, non-brisk 94%, absent 93%) |
Thomas et al. 2012 |
Azimi et al. scored 1,865 primary melanomas with a different TIL grading system using a score of 0–3 and showed that TIL grade was inversely associated with SLN status. TIL grade 0 was associated with 27.8% rate of SLN positivity compared to a rate of only 5.6% for TIL grade 3 [86]. Higher TIL grade was also significantly associated with longer RFS and MSS (survival rate of 100% vs. 75% for TIL grades 3 and 0, respectively, with median follow-up time of 43 months). Multivariate analysis demonstrated that the strongest predictors of RFS were thickness, ulceration, and TIL grade [86]. Similarly, in 887 patients who underwent SLN mapping, Taylor et al. found that absent TIL was predictive of SLN positivity in both univariate and multivariate analysis; however, in contrast to the results of Azimi et al., there was no difference in DFS or OS [91].
There are several reasons for the discrepancies in TIL grade among studies. Studies differed by sample size, patient population, characteristics of the primary tumors, and even method of grading TILs. For example, Clemente et al. examined 285 primary melanomas of which 82% were thicker than 2 mm, and found an association of TIL grade with survival, [91]. In contrast, Barnhill et al. studied 548 primary melanomas with a higher proportion of thinner melanomas (43% were less than 0.76 mm) in which there was no significant difference in OS based on TIL grade [90, 94]. TILs are thought to be more frequent in thin melanomas [91], and in a study of 293 high risk primary melanomas (thickness > 4 mm), TIL grade did not significantly correlate with survival outcomes, although there was a trend towards improved relapse-free survival for TIL presence [95]. Furthermore, studies vary in the TIL groups they compare for their survival analysis. Some only compare absent vs. present TIL grade [90–91] without separating out the brisk and non-brisk groups, while others group non-brisk and absent TIL grade [87] or do not address absent TILs at all [93], which makes cross-study comparisons challenging.
The identification of balance between immunostimulatory and immunosuppressive T-cell populations within the lymphocytic infiltrates is an overlooked consideration. For example, in a study of CD3, CD8, CD20, and Foxp3 immunohistochemical expression on lymph nodes positive for melanoma, the ratios of peritumoral to intratumoral TILs for both CD3 and CD8 expression were lower in patients who developed recurrent melanoma [96]. In metastatic melanoma, Erdag et al. characterized immunotypes A, B, and C, which roughly compare with absent, non-brisk, and brisk TIL grades, and showed a difference in median OS rates (15, 23, and 130 months, respectively) as well as differences in the cellular composition of the immune infiltrate present in each immunotype [97]. Higher densities of CD8+ T cells and CD45+ leukocytes, T cells, and B cells were associated with improved survival. There was no correlation of Foxp3, a regulatory T cell marker, with survival. The proportion of B cells was highest in immunotype C and proportion of macrophages was lowest in immunotype C [97].
Immunoregulatory Gene Expression Levels
A newer area of investigation beyond pathologic TIL classifications is aimed at identifying differentially expressed genes with immunoregulatory functions, with the hope of characterizing prognostic immune biomarkers. This is now possible with the advent of efficient, high-throughput technology such as Nanostring that can quantify gene expression levels for up to 800 genes of interest from small amounts of RNA isolated from FFPE tissue. Recently, a 53-gene immune panel proposed from an initial set of 446 genes was tested for prognostic value using Nanostring technology in RNA isolated from melanoma tumors of 47 stage II and III patients [98]. The gene signature was found to be predictive of non-progression, RFS, and DSS. The candidate genes are important for T-cell and natural killer cell function, leukocyte migration, and are involved in immune surveillance pathways. For example, CCR5, CD8, CD3, and IZKF1 were more highly expressed in the patients who did not progress. Specifically, the most differentially expressed gene was CD2, a co-stimulatory molecule expressed on T and NK cells. Increased CD2 positive cells correlated with melanoma non-progression, as confirmed by IHC [98].
The Cancer Genome Atlas recently reported a distinct immunologically-characterized subgroup of cutaneous melanomas that predict improved survival outcomes. Regional melanoma lymph nodes were examined for a lymphocyte score (“LScore”) calculated based on the density and location of TILs, with higher LScores correlating with prolonged survival and stratification into the immune subclass. Additionally, elevated expression of LCK and SYK, non-receptor tyrosine kinases involved in lymphocyte signaling, were found in the immune subgroup, with a correlation of increased LCK gene expression and improved survival [99].
Although the differing study designs and patient populations challenge our understanding of the prognostic value of TILs, however, the role of the host anti-tumor immune response in melanoma cannot be ignored. Future studies characterizing immune phenotypes, and perhaps an immunoscore or quantitative molecular markers of host responses are needed to better assess correlation of TILs with clinical outcome. Gene expression analyses of immunoregulatory genes like that of Sivendran et al. may identify key immunologic markers that can be linked with current known AJCC prognostic factors or molecular markers for more accurate outcome predictions.
Serum Markers
While the serum marker lactate dehydrogenase (LDH) is incorporated into the AJCC staging system for substaging of metastatic (stage IV) melanoma, no serum-based biomarkers have been identified that would meet criteria for incorporation into the staging system for earlier-stage melanoma (predictive of relapse) or to provide better prognostic or therapeutic guidelines in advanced melanoma.
Melanoma inhibitory activity protein (MIA) is produced and secreted by melanoma cells. In a cohort of 84 patients prospectively followed with stage I and II cutaneous melanoma, high vs. low serum MIA concentrations were associated with increased recurrence (66% vs. 5%) and reduced DFS at 4 years (30.3% vs. 94.5%) [100]. Serum levels of S100B, a protein involved in microtubule assembly and inhibition of the tumor suppressor p53, increase progressively in each stage of melanoma and associate negatively with survival [101–102].
C-reactive protein (CRP) is an acute phase reactant elevated in numerous clinical scenarios, including states of infection, inflammation, and malignancy. Measurement of elevated serum CRP level may be superior to serum LDH level in detecting patients who progress to metastatic melanoma [103]. Serum CRP levels measured at time of primary melanoma resection and then sequentially thereafter showed an association of elevated CRP with worse OS, MSS, and DFS in stage I and II patients in multivariate analyses,, suggesting serum CRP may be a valuable prognostic biomarker in melanoma [104].
Two studies by our group examined the utility of serum-based miRNA abundance for prediction and surveillance of recurrence in melanoma patients [105–106]. Friedman et al. defined a 5-miRNA signature from sera isolated from at-diagnosis blood of 80 primary melanoma patients that classified patients into high- and low-risk groups, which separated well in KM curves for RFS. In a follow-up study, we also developed a refined 4-miRNA sera-based signature (miR-150, miR-15b, miR-30d, and miR-425) adjusted for AJCC stage to define low- and high-risk patients, which stratified patients by RFS and OS in two cohorts (n = 201, n = 82) [105].
Finally, there is interest in measuring circulating tumor DNA (ctDNA) in advanced melanoma as an early indicator of response to treatment [107–108]. While ctDNA measurements are not currently used in clinical practice, prospective studies will address whether ctDNA can be used as a blood-based biomarker to detect melanoma recurrence.
Discussion
Our review has identified numerous reports of individual or sets of biomarkers that are prognostic or associate with prognostic factors in the primary melanoma cohorts in which they were studied. Only a few have been examined in multiple similar studies by independent groups and in independent patient cohorts. Collectively, assessed biomarkers encompass melanoma cell intrinsic and host-derived gene-, RNA-, and protein-based factors, as well as host-based cellular factors (e.g. TILs). Ideally, the biologic functions of these markers would mechanistically explain their prognostic impact, and as such, focus on markers with biological plausibility might be prudent; however, many putative biomarkers, which may be useful prognostically, have underlying biology that has not yet been clearly elucidated. In spite of the many studies reported, translation of genetic, molecular, or host-based factors into clinical usage in primary melanoma has not yet occurred.
Immunostaining-based molecular markers are routinely assessed for certain cancers, such as HER2 amplification in breast cancer [109]; however, none of the protein-based studies in primary melanoma have advanced into clinical practice. Cancer centers are rapidly incorporating genetic sequencing platforms into routine clinical practice to identify somatic variants in patient tumors, however, primary melanoma patients are unlikely to receive such testing until there is clarity of the mutations that clearly associate with patient outcome or until additional adjuvant therapies become available. RNA expression-based molecular markers will be more challenging to implement into routine practice and will likely require commercial development and validation of these assays as superior to the somatic mutational-based prognostic markers that have achieved broad uptake in the oncologic community. Examples of prognostic gene expression signatures already in practice include Mammaprint and OncotypeDX in early stage breast cancers [110–111] and OncotypeDX in stage II colon cancer [112]. The development of new biomarker-based systems for prognosticating clinical outcomes in early-stage melanoma can serve to select patients for clinical trials of adjuvant therapies.
Recently, Castle Biosciences released the DecisionDx Melanoma gene panel which predicts the risk (high (69%) or low (3%)) of distant metastasis at 5 years for stage I-III patients [66, 113]. This panel may be used in clinical practice, however it is not currently recommended by any standard treatment guidelines, including the National Comprehensive Cancer Network (NCCN). Definitive assessment of the sensitivity and specificity of this test should be conducted in prospective analyses of patients who are representative of the general primary melanoma patient population. Moreover, guidance on interpretation of DecisionDx results is of critical importance. Clinicians may suggest that patients predicted by this test to be at high risk for metastasis undergo more frequent clinical evaluations and surveillance imaging, particularly in patients who have negative sentinel lymph node biopsies. However, in settings in which adjuvant therapies are either contraindicated or not available, this categorization may increase patient anxiety without real benefit to the patient. Would increased surveillance of these patients result in earlier detection of recurrent disease? More importantly, would early detection of recurrence improve overall survival or help reduce melanoma mortality rates of these patients?
As previously noted [43], there are a number of challenges that have limited prognostic biomarker study interpretations and cross-study comparisons. Since 2005, publishing groups are recommended to follow the REMARK criteria as reporting standards [114], which aim to encourage transparency and improve study reporting. REMARK guidelines cover the reporting of study design, preplanned hypotheses, patient and specimen characteristics, assay methods, and statistical analysis methods. We advocate that groups performing future prognostic studies should consistently use REMARK criteria as a guide during study design, implementation, and analysis, as well as final reporting [115]. In addition to these guidelines, there are other technical considerations for molecular prognostic studies that are not frequently considered or discussed. For non-histological studies, sample isolation and purity are critical considerations. For tumor cell-intrinsic biomarkers, pure tumor sample is ideal for accurate measurements, cross-sample, and cross-study comparisons. In contrast, molecular-based measurements of immune cell infiltrates or micro-environmental factors from surrounding tissue may also represent good prognostic markers. As such, consideration of the cellular source(s) of the markers to be measured can greatly inform strategies for sample isolation and the level of tumor purity that is desirable. In contrast, the tumor- or host-derived nature of histological-based markers is clear, because cellular architecture is preserved. Moreover, for protein biomarkers cellular localization can be considered (e.g. nuclear staining, nuclear/cytoplasmic ratio, etc.), as well as post-translational modifications. However, the quantitative nature of the assessment being performed is a key concern for histological-based markers, with the most rigorous studies being based on automated quantitative analysis (AQUA). In addition, histological studies are typically limited to a single, 1-cell thick plane (or even small tumor areas within such planes, i.e. tissue microarrays [TMAs]) of a much larger 3-dimensional object. This limitation is also present in non-histological marker studies, but to a slightly lesser extent, since multiple sections (5–10) or whole TMA cores are typically required to extract sufficient material for analysis. In these studies, which typically rely on small portions of a much larger tumor, tumor-cell heterogeneity, if present, may result in sampling bias that has the potential to increase false negatives and false positives, and thereby confound study interpretations.
In addition to these considerations, markers are measured at single points in time and thus cannot adequately address any lead-time bias associated with the dimension of time. To the extent that AJCC stage represents some measure of time from tumor onset, it would be advisable to adjust for or stratify by tumor stage in multivariate analyses. Among other factors to control for, histological subtype (superficial spreading, nodular, acral), which vary in terms of pathologic features and may represent different biological entities, is an important consideration [116–117]. While the data in this review are focused on cutaneous melanoma, the rarer and more therapy-resistant melanoma types such as uveal and mucosal will also require further study to identify markers of both prognostic and predictive value.
Additionally, future prognostic studies must consider which primary outcomes to examine (PFS, DMFS, OS, etc.) and understand how these outcomes may be affected by frequency of clinical surveillance strategies, including imaging, and treatment strategies. Although DMFS may be associated with lead-time bias, it may provide more accurate measures of associations with putative prognostic markers than OS, which will increasingly be confounded by efficacious systemic therapies administered to advanced melanoma patients. The effect of a changing treatment landscape on defining prognostic molecular markers in future studies warrants serious consideration. As archival FFPE samples are exhausted from patient cohorts for whom clearly efficacious therapeutics were not available, future prognostic studies will depend on samples from patients treated with MAPK inhibitors, immunotherapeutic regimens, and other therapies or combinations, which will have varying responses and survival outcomes. However, prognostic factors that are independent of therapeutic response will be more difficult to define, due to the large number of factors that will need to be controlled for during study design or adjusted for in multivariate analyses. Collectively, these limitations suggest that collaborative, multi-institutional working groups that design and conduct well-reasoned, pre-defined studies evaluating multiple markers and/or marker types in large, well-defined patient cohorts would be the most efficient deployment of resources and the most likely to identify reproducible markers. Such studies could also be designed to allow multi-modal, multi-parameter risk signatures to be defined, which might dramatically enhance prognostication of primary melanoma patients. Any promising prognostic markers identified should ideally be validated in prospectively-collected tissue sets, preferably in the setting of a clinical trial.
Conclusion
We continue to believe that improved prognostic stratification has the potential to transform primary melanoma patient management, as clear differences in outcome exist for patients with histologically similar lesions. Our understanding of the particular factors (genetic mutation, expression alteration, host response, etc.) that are critical for predicting patient outcomes is incomplete. Overall, the studies we have considered in this review have not defined prognostic markers that could be readily incorporated into the current staging system. As such, efforts should be continued in these and other (e.g. proteomics) directions to maximize the likelihood of identifying clinically useful prognostic biomarkers. Each of these directions informs the others, and we may find that each is independently important to derive a more advanced prognostic model that stratifies patients accurately and robustly.
Supplementary Material
Acknowledgments
Funding: Part of this work was supported by: Department of Defense W81XWH-10-1-0803 (PIs: EH, IO); NIH/NCI 1R01CA163891-01A1 (PIs: EH, IO)
Footnotes
There are no conflict of interest disclosures for any of the authors.
References
- 1.Siegel R, Ma J, Zou Z, Jemal A. Cancer statistics, 2014. CA Cancer J Clin. 2014;64:9–29. doi: 10.3322/caac.21208. [DOI] [PubMed] [Google Scholar]
- 2.Clark WH, Jr, From L, Bernardino EA, Mihm MC. The histogenesis and biologic behavior of primary human malignant melanomas of the skin. Cancer Res. 1969;29:705–727. [PubMed] [Google Scholar]
- 3.Breslow A. Thickness, cross-sectional areas and depth of invasion in the prognosis of cutaneous melanoma. Ann Surg. 1970;172:902–908. doi: 10.1097/00000658-197011000-00017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Balch CM, Buzaid AC, Soong SJ, et al. Final version of the American Joint Committee on Cancer staging system for cutaneous melanoma. J Clin Oncol. 2001;19:3635–3648. doi: 10.1200/JCO.2001.19.16.3635. [DOI] [PubMed] [Google Scholar]
- 5.Balch CM, Soong SJ, Gershenwald JE, et al. Prognostic factors analysis of 17,600 melanoma patients: validation of the American Joint Committee on Cancer melanoma staging system. J Clin Oncol. 2001;19:3622–3634. doi: 10.1200/JCO.2001.19.16.3622. [DOI] [PubMed] [Google Scholar]
- 6.Buzaid AC, Ross MI, Balch CM, et al. Critical analysis of the current American Joint Committee on Cancer staging system for cutaneous melanoma and proposal of a new staging system. J Clin Oncol. 1997;15:1039–1051. doi: 10.1200/JCO.1997.15.3.1039. [DOI] [PubMed] [Google Scholar]
- 7.Balch CM, Gershenwald JE, Soong SJ, et al. Final version of 2009 AJCC melanoma staging and classification. J Clin Oncol. 2009;27:6199–6206. doi: 10.1200/JCO.2009.23.4799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Piris A, Mihm MC, Jr, Duncan LM. AJCC melanoma staging update: impact on dermatopathology practice and patient management. J Cutan Pathol. 2011;38:394–400. doi: 10.1111/j.1600-0560.2011.01699.x. [DOI] [PubMed] [Google Scholar]
- 9.American Cancer Society. [[accessed April 26, 2015]];What are the survival rates for melanoma skin cancer, by stage? Availbale from URL: http://www.cancer.org/cancer/skincancer-melanoma/detailedguide/melanoma-skin-cancer-survival-rates.
- 10.Gimotty PA, Elder DE, Fraker DL, et al. Identification of high-risk patients among those diagnosed with thin cutaneous melanomas. J Clin Oncol. 2007;25:1129–1134. doi: 10.1200/JCO.2006.08.1463. [DOI] [PubMed] [Google Scholar]
- 11.Balch CM, Gershenwald JE, Soong SJ, et al. Multivariate analysis of prognostic factors among 2,313 patients with stage III melanoma: comparison of nodal micrometastases versus macrometastases. J Clin Oncol. 2010;28:2452–2459. doi: 10.1200/JCO.2009.27.1627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Harbst K, Staaf J, Lauss M, et al. Molecular profiling reveals low- and high-grade forms of primary melanoma. Clin Cancer Res. 2012;18:4026–4036. doi: 10.1158/1078-0432.CCR-12-0343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.von Smolinski D, Leverkoehne I, von Samson-Himmelstjerna G, Gruber AD. Impact of formalin-fixation and paraffin-embedding on the ratio between mRNA copy numbers of differently expressed genes. Histochem Cell Biol. 2005;124:177–188. doi: 10.1007/s00418-005-0013-0. [DOI] [PubMed] [Google Scholar]
- 14.Berger MF, Hodis E, Heffernan TP, et al. Melanoma genome sequencing reveals frequent PREX2 mutations. Nature. 2012;485:502–506. doi: 10.1038/nature11071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Hodis E, Watson IR, Kryukov GV, et al. A landscape of driver mutations in melanoma. Cell. 2012;150:251–263. doi: 10.1016/j.cell.2012.06.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Jeck WR, Parker J, Carson CC, et al. Targeted next generation sequencing identifies clinically actionable mutations in patients with melanoma. Pigment Cell Melanoma Res. 2014;27:653–663. doi: 10.1111/pcmr.12238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Krauthammer M, Kong Y, Ha BH, et al. Exome sequencing identifies recurrent somatic RAC1 mutations in melanoma. Nat Genet. 2012;44:1006–1014. doi: 10.1038/ng.2359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Lawrence MS, Stojanov P, Mermel CH, et al. Discovery and saturation analysis of cancer genes across 21 tumour types. Nature. 2014;505:495–501. doi: 10.1038/nature12912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Dong J, Phelps RG, Qiao R, et al. BRAF oncogenic mutations correlate with progression rather than initiation of human melanoma. Cancer Res. 2003;63:3883–3885. [PubMed] [Google Scholar]
- 21.Safaee Ardekani G, Jafarnejad SM, Tan L, Saeedi A, Li G. The prognostic value of BRAF mutation in colorectal cancer and melanoma: a systematic review and meta-analysis. PLoS One. 2012;7:e47054. doi: 10.1371/journal.pone.0047054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Maldonado JL, Fridlyand J, Patel H, et al. Determinants of BRAF mutations in primary melanomas. J Natl Cancer Inst. 2003;95:1878–1890. doi: 10.1093/jnci/djg123. [DOI] [PubMed] [Google Scholar]
- 23.Shinozaki M, Fujimoto A, Morton DL, Hoon DS. Incidence of BRAF oncogene mutation and clinical relevance for primary cutaneous melanomas. Clin Cancer Res. 2004;10:1753–1757. doi: 10.1158/1078-0432.ccr-1169-3. [DOI] [PubMed] [Google Scholar]
- 24.Akslen LA, Angelini S, Straume O, et al. BRAF and NRAS mutations are frequent in nodular melanoma but are not associated with tumor cell proliferation or patient survival. J Invest Dermatol. 2005;125:312–317. doi: 10.1111/j.0022-202X.2005.23788.x. [DOI] [PubMed] [Google Scholar]
- 25.Ellerhorst JA, Greene VR, Ekmekcioglu S, et al. Clinical correlates of NRAS and BRAF mutations in primary human melanoma. Clin Cancer Res. 2011;17:229–235. doi: 10.1158/1078-0432.CCR-10-2276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Devitt B, Liu W, Salemi R, et al. Clinical outcome and pathological features associated with NRAS mutation in cutaneous melanoma. Pigment Cell Melanoma Res. 2011;24:666–672. doi: 10.1111/j.1755-148X.2011.00873.x. [DOI] [PubMed] [Google Scholar]
- 27.Woodman SE, Davies MA. Targeting KIT in melanoma: a paradigm of molecular medicine and targeted therapeutics. Biochem Pharmacol. 2010;80:568–574. doi: 10.1016/j.bcp.2010.04.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Thomas NE, Edmiston SN, Alexander A, Groben PA, Parrish E, Kricker A, et al. Association Between and Mutational Status and Melanoma-Specific Survival Among Patients With Higher Risk Primary Melanoma. JAMA Oncol. 2015;1(3):359–68. doi: 10.1001/jamaoncol.2015.0493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Todd JR, Scurr LL, Becker TM, Kefford RF, Rizos H. The MAPK pathway functions as a redundant survival signal that reinforces the PI3K cascade in c-Kit mutant melanoma. Oncogene. 2014;33:236–245. doi: 10.1038/onc.2012.562. [DOI] [PubMed] [Google Scholar]
- 30.Curtin JA, Busam K, Pinkel D, Bastian BC. Somatic activation of KIT in distinct subtypes of melanoma. J Clin Oncol. 2006;24:4340–4346. doi: 10.1200/JCO.2006.06.2984. [DOI] [PubMed] [Google Scholar]
- 31.Beadling C, Jacobson-Dunlop E, Hodi FS, et al. KIT gene mutations and copy number in melanoma subtypes. Clin Cancer Res. 2008;14:6821–6828. doi: 10.1158/1078-0432.CCR-08-0575. [DOI] [PubMed] [Google Scholar]
- 32.Bradford PT, Goldstein AM, McMaster ML, Tucker MA. Acral lentiginous melanoma: incidence and survival patterns in the United States, 1986–2005. Arch Dermatol. 2009;145:427–434. doi: 10.1001/archdermatol.2008.609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Mikhail M, Velazquez E, Shapiro R, et al. PTEN expression in melanoma: relationship with patient survival, Bcl-2 expression, and proliferation. Clin Cancer Res. 2005;11:5153–5157. doi: 10.1158/1078-0432.CCR-05-0397. [DOI] [PubMed] [Google Scholar]
- 34.Aguissa-Toure AH, Li G. Genetic alterations of PTEN in human melanoma. Cell Mol Life Sci. 2012;69:1475–1491. doi: 10.1007/s00018-011-0878-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Lahtz C, Stranzenbach R, Fiedler E, Helmbold P, Dammann RH. Methylation of PTEN as a prognostic factor in malignant melanoma of the skin. J Invest Dermatol. 2010;130:620–622. doi: 10.1038/jid.2009.226. [DOI] [PubMed] [Google Scholar]
- 36.Gold HL, Wengrod J, de Miera EV, et al. PP6C hotspot mutations in melanoma display sensitivity to Aurora kinase inhibition. Mol Cancer Res. 2014;12:433–439. doi: 10.1158/1541-7786.MCR-13-0422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Mar VJ, Wong SQ, Logan A, et al. Clinical and pathological associations of the activating RAC1 P29S mutation in primary cutaneous melanoma. Pigment Cell Melanoma Res. 2014;27:1117–1125. doi: 10.1111/pcmr.12295. [DOI] [PubMed] [Google Scholar]
- 38.Dutton-Regester K, Gartner JJ, Emmanuel R, et al. A highly recurrent RPS27 5’UTR mutation in melanoma. Oncotarget. 2014;5:2912–2917. doi: 10.18632/oncotarget.2048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Huang FW, Hodis E, Xu MJ, Kryukov GV, Chin L, Garraway LA. Highly recurrent TERT promoter mutations in human melanoma. Science. 2013;339:957–959. doi: 10.1126/science.1229259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Horn S, Figl A, Rachakonda PS, et al. TERT promoter mutations in familial and sporadic melanoma. Science. 2013;339:959–961. doi: 10.1126/science.1230062. [DOI] [PubMed] [Google Scholar]
- 41.Griewank KG, Murali R, Puig-Butille JA, et al. TERT promoter mutation status as an independent prognostic factor in cutaneous melanoma. J Natl Cancer Inst. 2014:106. doi: 10.1093/jnci/dju246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Populo H, Boaventura P, Vinagre J, et al. TERT promoter mutations in skin cancer: the effects of sun exposure and X-irradiation. J Invest Dermatol. 2014;134:2251–2257. doi: 10.1038/jid.2014.163. [DOI] [PubMed] [Google Scholar]
- 43.Gould Rothberg BE, Berger AJ, Molinaro AM, et al. Melanoma prognostic model using tissue microarrays and genetic algorithms. J Clin Oncol. 2009;27:5772–5780. doi: 10.1200/JCO.2009.22.8239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Gould Rothberg BE, Bracken MB, Rimm DL. Tissue biomarkers for prognosis in cutaneous melanoma: a systematic review and meta-analysis. J Natl Cancer Inst. 2009;101:452–474. doi: 10.1093/jnci/djp038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Gould Rothberg BE, Rimm DL. Biomarkers: the useful and the not so useful--an assessment of molecular prognostic markers for cutaneous melanoma. J Invest Dermatol. 2010;130:1971–1987. doi: 10.1038/jid.2010.149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Rothberg BE, Rimm DL. Construction and analysis of multiparameter prognostic models for melanoma outcome. Methods Mol Biol. 2014;1102:227–258. doi: 10.1007/978-1-62703-727-3_13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Meyer S, Fuchs TJ, Bosserhoff AK, et al. A seven-marker signature and clinical outcome in malignant melanoma: a large-scale tissue-microarray study with two independent patient cohorts. PLoS One. 2012;7:e38222. doi: 10.1371/journal.pone.0038222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Kashani-Sabet M, Venna S, Nosrati M, et al. A multimarker prognostic assay for primary cutaneous melanoma. Clin Cancer Res. 2009;15:6987–6992. doi: 10.1158/1078-0432.CCR-09-1777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Klopfleisch R, Weiss A, Gruber A. Excavation of a buried treasure--DNA, mRNA, miRNA and protein analysis in formalin fixed, paraffin embedded tissues. Histology and histopathology. 2011;26:797–1607. doi: 10.14670/HH-26.797. [DOI] [PubMed] [Google Scholar]
- 50.Jaeger J, Koczan D, Thiesen HJ, et al. Gene expression signatures for tumor progression, tumor subtype, and tumor thickness in laser-microdissected melanoma tissues. Clin Cancer Res. 2007;13:806–815. doi: 10.1158/1078-0432.CCR-06-1820. [DOI] [PubMed] [Google Scholar]
- 51.Bittner M, Meltzer P, Chen Y, et al. Molecular classification of cutaneous malignant melanoma by gene expression profiling. Nature. 2000;406:536–540. doi: 10.1038/35020115. [DOI] [PubMed] [Google Scholar]
- 52.Haqq C, Nosrati M, Sudilovsky D, et al. The gene expression signatures of melanoma progression. Proc Natl Acad Sci U S A. 2005;102:6092–6097. doi: 10.1073/pnas.0501564102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Talantov D, Mazumder A, Yu J, et al. Novel genes associated with malignant melanoma but not benign melanocytic lesions. Clin Cancer Res. 2005;11:7234–7276. doi: 10.1158/1078-0432.CCR-05-0683. [DOI] [PubMed] [Google Scholar]
- 54.Riker A, Enkemann S, Fodstad O, et al. The gene expression profiles of primary and metastatic melanoma yields a transition point of tumor progression and metastasis. BMC medical genomics. 2008;1:13. doi: 10.1186/1755-8794-1-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Winnepenninckx V, Lazar V, Michiels S, et al. Gene expression profiling of primary cutaneous melanoma and clinical outcome. J Natl Cancer Inst. 2006;98:472–482. doi: 10.1093/jnci/djj103. [DOI] [PubMed] [Google Scholar]
- 56.Kauffmann A, Rosselli F, Lazar V, et al. High expression of DNA repair pathways is associated with metastasis in melanoma patients. Oncogene. 2008;27:565–573. doi: 10.1038/sj.onc.1210700. [DOI] [PubMed] [Google Scholar]
- 57.Wardwell-Ozgo J, Dogruluk T, Gifford A, et al. HOXA1 drives melanoma tumor growth and metastasis and elicits an invasion gene expression signature that prognosticates clinical outcome. Oncogene. 2014;33:1017–1026. doi: 10.1038/onc.2013.30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Brunner G, Reitz M, Schwipper V, et al. Increased expression of the tumor suppressor PLZF is a continuous predictor of long-term survival in malignant melanoma patients. Cancer Biother Radiopharm. 2008;23:451–459. doi: 10.1089/cbr.2008.0473. [DOI] [PubMed] [Google Scholar]
- 59.Brunner G, Reitz M, Heinecke A, et al. A nine-gene signature predicting clinical outcome in cutaneous melanoma. J Cancer Res Clin Oncol. 2013;139:249–258. doi: 10.1007/s00432-012-1322-z. [DOI] [PubMed] [Google Scholar]
- 60.Gschaider M, Neumann F, Peters B, et al. An attempt at a molecular prediction of metastasis in patients with primary cutaneous melanoma. PLoS One. 2012;7:e49865. doi: 10.1371/journal.pone.0049865. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Conway C, Mitra A, Jewell R, et al. Gene expression profiling of paraffin-embedded primary melanoma using the DASL assay identifies increased osteopontin expression as predictive of reduced relapse-free survival. Clin Cancer Res. 2009;15:6939–6946. doi: 10.1158/1078-0432.CCR-09-1631. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Rangel J, Nosrati M, Torabian S, et al. Osteopontin as a molecular prognostic marker for melanoma. Cancer. 2008;112:144–150. doi: 10.1002/cncr.23147. [DOI] [PubMed] [Google Scholar]
- 63.Zhou Y, Dai DL, Martinka M, et al. Osteopontin expression correlates with melanoma invasion. J Invest Dermatol. 2005;124:1044–1052. doi: 10.1111/j.0022-202X.2005.23680.x. [DOI] [PubMed] [Google Scholar]
- 64.Jewell R, Conway C, Mitra A, et al. Patterns of expression of DNA repair genes and relapse from melanoma. Clin Cancer Res. 2010;16:5211–5221. doi: 10.1158/1078-0432.CCR-10-1521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Gerami P, Cook RW, Russell MC, et al. Gene expression profiling for molecular staging of cutaneous melanoma in patients undergoing sentinel lymph node biopsy. J Am Acad Dermatol. 2015;72:780–785. doi: 10.1016/j.jaad.2015.01.009. e783. [DOI] [PubMed] [Google Scholar]
- 66.Gerami P, Cook RW, Wilkinson J, et al. Development of a prognostic genetic signature to predict the metastatic risk associated with cutaneous melanoma. Clin Cancer Res. 2015;21:175–183. doi: 10.1158/1078-0432.CCR-13-3316. [DOI] [PubMed] [Google Scholar]
- 67.Hall JS, Taylor J, Valentine HR, et al. Enhanced stability of microRNA expression facilitates classification of FFPE tumour samples exhibiting near total mRNA degradation. Br J Cancer. 2012;107:684–694. doi: 10.1038/bjc.2012.294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Xi Y, Nakajima G, Gavin E, et al. Systematic analysis of microRNA expression of RNA extracted from fresh frozen and formalin-fixed paraffin-embedded samples. RNA. 2007;13:1668–1674. doi: 10.1261/rna.642907. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Caramuta S, Egyh√°zi S, Rodolfo M, et al. MicroRNA expression profiles associated with mutational status and survival in malignant melanoma. J Invest Dermatol. 2010;130:2062–2132. doi: 10.1038/jid.2010.63. [DOI] [PubMed] [Google Scholar]
- 70.Chen J, Feilotter H, Paré G, et al. MicroRNA-193b represses cell proliferation and regulates cyclin D1 in melanoma. Am J Pathol. 2010;176:2520–2529. doi: 10.2353/ajpath.2010.091061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Mazar J, DeYoung K, Khaitan D, et al. The regulation of miRNA-211 expression and its role in melanoma cell invasiveness. PLoS One. 2010:5. doi: 10.1371/journal.pone.0013779. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Philippidou D, Schmitt M, Moser D, et al. Signatures of microRNAs and selected microRNA target genes in human melanoma. Cancer Res. 2010;70:4163–4236. doi: 10.1158/0008-5472.CAN-09-4512. [DOI] [PubMed] [Google Scholar]
- 73.Levy C, Khaled M, Iliopoulos D, et al. Intronic miR-211 assumes the tumor suppressive function of its host gene in melanoma. Mol Cell. 2010;40:841–849. doi: 10.1016/j.molcel.2010.11.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Segura M, Belitskaya-Lévy I, Rose A, et al. Melanoma MicroRNA signature predicts post-recurrence survival. Clin Cancer Res. 2010;16:1577–1663. doi: 10.1158/1078-0432.CCR-09-2721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Tembe V, Schramm SJ, Stark MS, et al. MicroRNA and mRNA expression profiling in metastatic melanoma reveal associations with BRAF mutation and patient prognosis. Pigment Cell Melanoma Res. 2015;28:254–266. doi: 10.1111/pcmr.12343. [DOI] [PubMed] [Google Scholar]
- 76.Hanniford D, Segura MF, Zhong J, et al. Identification of metastasis-suppressive microRNAs in primary melanoma. J Natl Cancer Inst. 2015:107. doi: 10.1093/jnci/dju494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Hanniford D, Zhong J, Koetz L, et al. A miRNA-based signature detected in primary melanoma tissue predicts development of brain metastasis. Clin Cancer Res. 2015 doi: 10.1158/1078-0432.CCR-14-2566. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Poliseno L, Haimovic A, Segura MF, et al. Histology-specific microRNA alterations in melanoma. J Invest Dermatol. 2012;132:1860–1868. doi: 10.1038/jid.2011.451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.van Kempen LC, van den Hurk K, Lazar V, et al. Loss of microRNA-200a and c, and microRNA-203 expression at the invasive front of primary cutaneous melanoma is associated with increased thickness and disease progression. Virchows Arch. 2012;461:441–448. doi: 10.1007/s00428-012-1309-9. [DOI] [PubMed] [Google Scholar]
- 80.Gaziel-Sovran A, Segura M, Di Micco R, et al. miR-30b/30d regulation of GalNAc transferases enhances invasion and immunosuppression during metastasis. Cancer Cell. 2011;20:104–122. doi: 10.1016/j.ccr.2011.05.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Pencheva N, Tran H, Buss C, et al. Convergent multi-miRNA targeting of ApoE drives LRP1/LRP8-dependent melanoma metastasis and angiogenesis. Cell. 2012;151:1068–1082. doi: 10.1016/j.cell.2012.10.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Boyle G, Woods S, Bonazzi V, et al. Melanoma cell invasiveness is regulated by miR-211 suppression of the BRN2 transcription factor. Pigment Cell Melanoma Res. 2011;24:525–562. doi: 10.1111/j.1755-148X.2011.00849.x. [DOI] [PubMed] [Google Scholar]
- 83.Satzger I, Mattern A, Kuettler U, et al. MicroRNA-15b represents an independent prognostic parameter and is correlated with tumor cell proliferation and apoptosis in malignant melanoma. International journal of cancer. Journal international du Cancer. 2010;126:2553–2615. doi: 10.1002/ijc.24960. [DOI] [PubMed] [Google Scholar]
- 84.Galon J, Pages F, Marincola FM, et al. Cancer classification using the Immunoscore: a worldwide task force. J Transl Med. 2012;10:205. doi: 10.1186/1479-5876-10-205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Clark WH, Jr, Elder DE, Guerry Dt, et al. Model predicting survival in stage I melanoma based on tumor progression. J Natl Cancer Inst. 1989;81:1893–1904. doi: 10.1093/jnci/81.24.1893. [DOI] [PubMed] [Google Scholar]
- 86.Azimi F, Scolyer RA, Rumcheva P, et al. Tumor-infiltrating lymphocyte grade is an independent predictor of sentinel lymph node status and survival in patients with cutaneous melanoma. J Clin Oncol. 2012;30:2678–2683. doi: 10.1200/JCO.2011.37.8539. [DOI] [PubMed] [Google Scholar]
- 87.Tuthill RJ, Unger JM, Liu PY, Flaherty LE, Sondak VK. Risk assessment in localized primary cutaneous melanoma: a Southwest Oncology Group study evaluating nine factors and a test of the Clark logistic regression prediction model. Am J Clin Pathol. 2002;118:504–511. doi: 10.1309/WBF7-N8KH-71KT-RVQ9. [DOI] [PubMed] [Google Scholar]
- 88.Thomas NE, Busam KJ, From L, et al. Tumor-infiltrating lymphocyte grade in primary melanomas is independently associated with melanoma-specific survival in the population-based genes, environment and melanoma study. J Clin Oncol. 2013;31:4252–4259. doi: 10.1200/JCO.2013.51.3002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Fortes C, Mastroeni S, Manooranparanpampil T, et al. Tumor-infiltrating lymphocytes predict cutaneous melanoma survival. J Transl Med. 2015;13:2066. doi: 10.1097/CMR.0000000000000164. [DOI] [PubMed] [Google Scholar]
- 90.Barnhill RL, Fine JA, Roush GC, Berwick M. Predicting five-year outcome for patients with cutaneous melanoma in a population-based study. Cancer. 1996;78:427–432. doi: 10.1002/(SICI)1097-0142(19960801)78:3<427::AID-CNCR8>3.0.CO;2-G. [DOI] [PubMed] [Google Scholar]
- 91.Taylor RC, Patel A, Panageas KS, Busam KJ, Brady MS. Tumor-infiltrating lymphocytes predict sentinel lymph node positivity in patients with cutaneous melanoma. J Clin Oncol. 2007;25:869–875. doi: 10.1200/JCO.2006.08.9755. [DOI] [PubMed] [Google Scholar]
- 92.Mandala M, Imberti GL, Piazzalunga D, et al. Clinical and histopathological risk factors to predict sentinel lymph node positivity, disease-free and overall survival in clinical stages I-II AJCC skin melanoma: outcome analysis from a single-institution prospectively collected database. Eur J Cancer. 2009;45:2537–2545. doi: 10.1016/j.ejca.2009.05.034. [DOI] [PubMed] [Google Scholar]
- 93.Burton AL, Roach BA, Mays MP, et al. Prognostic significance of tumor infiltrating lymphocytes in melanoma. Am Surg. 2011;77:188–192. [PubMed] [Google Scholar]
- 94.Clemente CG, Mihm MC, Jr, Bufalino R, Zurrida S, Collini P, Cascinelli N. Prognostic value of tumor infiltrating lymphocytes in the vertical growth phase of primary cutaneous melanoma. Cancer. 1996;77:1303–1310. doi: 10.1002/(SICI)1097-0142(19960401)77:7<1303::AID-CNCR12>3.0.CO;2-5. [DOI] [PubMed] [Google Scholar]
- 95.Rao UN, Lee SJ, Luo W, Mihm MC, Jr, Kirkwood JM. Presence of tumor-infiltrating lymphocytes and a dominant nodule within primary melanoma are prognostic factors for relapse-free survival of patients with thick (t4) primary melanoma: pathologic analysis of the e1690 and e1694 intergroup trials. Am J Clin Pathol. 2010;133:646–653. doi: 10.1309/AJCPTXMEFOVYWDA6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Capone M. Immunoscore: a new possible approach for melanoma classification. Journal for ImmunoTherapy of Cancer. 2014;2:193. [Google Scholar]
- 97.Erdag G, Schaefer JT, Smolkin ME, et al. Immunotype and immunohistologic characteristics of tumor-infiltrating immune cells are associated with clinical outcome in metastatic melanoma. Cancer Res. 2012;72:1070–1080. doi: 10.1158/0008-5472.CAN-11-3218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Sivendran S, Chang R, Pham L, et al. Dissection of immune gene networks in primary melanoma tumors critical for antitumor surveillance of patients with stage II-III resectable disease. J Invest Dermatol. 2014;134:2202–2211. doi: 10.1038/jid.2014.85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Cancer Genome Atlas Network. Genomic Classification of Cutaneous Melanoma. Cell. 2015;161:1681–1696. doi: 10.1016/j.cell.2015.05.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Sandru A, Panaitescu E. Prognostic value of melanoma inhibitory activity protein in localized cutaneous malignant melanoma. 2014;2014:843214. doi: 10.1155/2014/843214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Martenson ED, Hansson LO, Nilsson B, et al. Serum S-100b protein as a prognostic marker in malignant cutaneous melanoma. J Clin Oncol. 2001;19:824–831. doi: 10.1200/JCO.2001.19.3.824. [DOI] [PubMed] [Google Scholar]
- 102.Bonfrer JM, Korse CM, Nieweg OE, Rankin EM. The luminescence immunoassay S-100: a sensitive test to measure circulating S-100B: its prognostic value in malignant melanoma. Br J Cancer. 1998;77:2210–2214. doi: 10.1038/bjc.1998.368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Deichmann M, Kahle B, Moser K, Wacker J, Wust K. Diagnosing melanoma patients entering American Joint Committee on Cancer stage IV, C-reactive protein in serum is superior to lactate dehydrogenase. Br J Cancer. 2004;91:699–702. doi: 10.1038/sj.bjc.6602043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Fang S, Wang Y, Sui D, et al. C-Reactive Protein As a Marker of Melanoma Progression. J Clin Oncol. 2015 doi: 10.1200/JCO.2014.58.0209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Fleming NH, Zhong J, da Silva IP, et al. Serum-based miRNAs in the prediction and detection of recurrence in melanoma patients. Cancer. 2015;121:51–59. doi: 10.1002/cncr.28981. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Friedman EB, Shang S, de Miera EV, et al. Serum microRNAs as biomarkers for recurrence in melanoma. J Transl Med. 2012;10:155. doi: 10.1186/1479-5876-10-155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Lipson EJ, Velculescu VE, Pritchard TS, et al. Circulating tumor DNA analysis as a real-time method for monitoring tumor burden in melanoma patients undergoing treatment with immune checkpoint blockade. J Immunother Cancer. 2014;2:42. doi: 10.1186/s40425-014-0042-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Sanmamed MF, Fernandez-Landazuri S, Rodriguez C, et al. Quantitative cell-free circulating BRAFV600E mutation analysis by use of droplet digital PCR in the follow-up of patients with melanoma being treated with BRAF inhibitors. Clin Chem. 2015;61:297–304. doi: 10.1373/clinchem.2014.230235. [DOI] [PubMed] [Google Scholar]
- 109.Ellis CM, Dyson MJ, Stephenson TJ, Maltby EL. HER2 amplification status in breast cancer: a comparison between immunohistochemical staining and fluorescence in situ hybridisation using manual and automated quantitative image analysis scoring techniques. J Clin Pathol. 2005;58:710–714. doi: 10.1136/jcp.2004.023424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.van de Vijver MJ, He YD, van’t Veer LJ, et al. A gene-expression signature as a predictor of survival in breast cancer. N Engl J Med. 2002;347:1999–2009. doi: 10.1056/NEJMoa021967. [DOI] [PubMed] [Google Scholar]
- 111.Cronin M, Sangli C, Liu ML, et al. Analytical validation of the Oncotype DX genomic diagnostic test for recurrence prognosis and therapeutic response prediction in node-negative, estrogen receptor-positive breast cancer. Clin Chem. 2007;53:1084–1091. doi: 10.1373/clinchem.2006.076497. [DOI] [PubMed] [Google Scholar]
- 112.Webber EM, Lin JS, Evelyn PW. Oncotype DX tumor gene expression profiling in stage II colon cancer. Application: prognostic, risk prediction. PLoS Curr. 2010:2. doi: 10.1371/currents.RRN1177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.DecisionDx-Melanoma Test. Test overview: identifying early stage melanomas at high risk of metastasis. [[accessed June 18, 2015]];Castle Biosciences. Available from: URL: http://skinmelanoma.com/decisiondx-melanoma-test/test-overview-value/
- 114.McShane LM, Altman DG, Sauerbrei W, et al. Reporting recommendations for tumor marker prognostic studies. J Clin Oncol. 2005;23:9067–9072. doi: 10.1200/JCO.2004.01.0454. [DOI] [PubMed] [Google Scholar]
- 115.Altman DG, McShane LM, Sauerbrei W, Taube SE. Reporting Recommendations for Tumor Marker Prognostic Studies (REMARK): explanation and elaboration. PLoS Med. 2012;9:e1001216. doi: 10.1371/journal.pmed.1001216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Greenwald HS, Friedman EB, Osman I. Superficial spreading and nodular melanoma are distinct biological entities: a challenge to the linear progression model. Melanoma Res. 2012;22:1–8. doi: 10.1097/CMR.0b013e32834e6aa0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Warycha MA, Christos PJ, Mazumdar M, et al. Changes in the presentation of nodular and superficial spreading melanomas over 35 years. Cancer. 2008;113:3341–3348. doi: 10.1002/cncr.23955. [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.