<<

(2014) 33, 2413–2422 & 2014 Macmillan Publishers Limited All rights reserved 0950-9232/14 www.nature.com/onc

REVIEW Melanoma : new concepts and evolving paradigms

WE Damsky1,2, N Theodosakis1 and M Bosenberg1

Melanoma progression is typically depicted as a linear and stepwise process in which metastasis occurs relatively late in progression. Significant evidence suggests that in a subset of melanomas, progression is much more complex and less linear in nature. Epidemiologic and experimental observations in melanoma metastasis are reviewed here and are incorporated into a comprehensive model for melanoma metastasis, which takes into account the varied natural history of melanoma formation and progression.

Oncogene (2014) 33, 2413–2422; doi:10.1038/onc.2013.194; published online 3 June 2013 Keywords: ; melanoma; metastasis; BRAF; b-catenin

INTRODUCTION observations from human melanoma patients which add temporal Melanoma rates are rising worldwide.1 Most morbidity and and spatial complexity to metastasis. Many melanoma patients are mortality in melanoma is associated with metastatic disease, cured after excision of their primary tumor; however, some are which can occur even from thin primary tumors.2 Accordingly, not, and will have disease recurrence. Melanoma recurs less metastasis is a particularly ominous sign; when metastasis is frequently at the site of the original primary tumor, instead more clinically evident, is very poor. For example, melanoma commonly recurring at different sites in the body as metastatic patients with three or more sites of metastatic disease die within lesions.9 These recurrence patterns suggest that melanoma cells 1 year 495% of the time.3,4 New targeted and immunomo- had already spread before excision of the primary tumor, even dulatory therapies such as and have though this spread might not have been clinically apparent at offered the first improvements in overall survival to melanoma that time. In melanoma patients, metastatic recurrence can patients in several decades.5,6 Unfortunately, only a subset of occur within a relatively short period of time, but takes 45 patients benefit in a durable fashion from these therapies, while years in about 40% of patients.4,10–12 The period in between initial most will still ultimately succumb to metastatic disease. excision of the primary tumor and subsequent metastatic Melanoma results from the malignant proliferation of melano- recurrence is often referred to as metastatic dormancy, where cytes, the normal pigment producing cells in skin. Classically previously disseminated tumor cells are thought to persist in a melanoma progression and metastasis has been depicted using relatively non-proliferative state. the Clark model.7 In this model, progression of normal The mechanisms regulating metastatic dormancy are not to metastatic melanoma is linear, and consists of a particularly well understood, however, several hypotheses have series of steps including: benign precursor lesion formation been proposed. One hypothesis suggests that tumor cell extrinsic (melanocytic ), , progression through both factors such as the actively and purposefully radial and vertical growth phases of primary tumor growth, and constrain growth of disseminated cells.13,14 Another model ultimately metastasis (Figure 1). The accumulation of genetic and suggests that a relative inability of disseminated tumor cells to epigenetic changes is thought to drive progression through these recruit new blood vessels limits frank outgrowth of disseminated steps.8 In the Clark model, metastasis ensues late in disease disease.15 Finally, some authors have proposed that metastatic progression, only after all previous steps have been completed tumor cells simply growth arrest in these new stromal and relatively rare clones with metastatic competency emerge. microenvironments due to a relative mismatch between cell–cell Although this model likely depicts the natural history of some and cell–extracellular matrix interactions.16,17 Despite differences melanomas, several pathologic, epidemiologic, and experimental in the primary mechanism enforcing dormancy, these models are observations suggest that the progression of other melanomas similar in that they hypothesize that disseminated tumor cells must may be more complex and less linear in nature. Evolving overcome some barrier(s) before progression to a macrometastatic conceptual paradigms of melanoma progression that account lesion. Metastatic dormancy and subsequent reactivation of for these additional observations and incorporate the many growth in melanoma have been recently reviewed.12,16,18 individual steps in metastasis will be explored here. The seed-and-soil hypothesis of metastasis underscores the additional spatial complexity of metastasis. Seed-and-soil posits that patterns of metastasis observed in human TEMPORAL AND SPATIAL COMPLEXITY OF METASTASIS result from inherent compatibilities (or incompatibilities) between Metastasis involves a series of steps which must be completed different tumor cell types and different sites of metastasis.19 before the emergence of clinically meaningful metastatic disease For example, has an extraordinary proclivity for (Figure 1). These steps are further complicated by additional metastasis to the liver. Nearly 90% of patients with metastatic

1Department of , Yale University School of Medicine, New Haven, CT, USA and 2Department of , University of Vermont College of Medicine, Burlington, VT, USA. Correspondence: Dr WE Damsky, Department of Pathology, University of Vermont College of Medicine, Given Box A14, 189 Beaumont Avenue, Burlington, VT 05495, USA or Dr M Bosenberg, Department of Dermatology, Yale University School of Medicine, 333 Cedar Street, LCI 501, PO Box 208059, New Haven, CT 06520, USA. E-mail: [email protected] or [email protected] Received 2 March 2013; revised 22 April 2013; accepted 22 April 2013; published online 3 June 2013 Melanoma metastasis WE Damsky et al 2414

Figure 1. Melanoma formation and progression. Melanoma formation and metastasis are complex processes. Primary tumor formation has classically been depicted using a linear model in which normal melanocytes progress through several precursor lesions and ultimately to melanoma (individual steps colored in blue). However, substantial evidence suggests that such linear progression is only one pathway to and that not all steps always occur during progression of individual melanomas. Other pathways to primary tumor formation are also depicted (non-linear). There is additional evidence to suggest that dissemination of cells away from the primary lesion may not be a unique feature of invasive melanomas. The possibility of spread from these lesions is depicted with question marks. Approximately 4–12% of metastatic lesions are not associated with a primary cutaneous melanoma (depicted in orange). The individual steps in metastasis are depicted in green. Unlike primary tumor formation, steps in metastasis are likely to always occur in a linear fashion; although it is unclear whether passage of tumor cells through lymphatics is absolutely required for systemic dissemination. Direct entry of tumor cells into venous return may also be possible and is depicted with a question mark. The from micrometastasis to macrometastasis is likely a critical, rate-limiting step in the of many disseminated melanocytic lesions. This transition may take months to years (if it occurs at all) and is preceded by metastatic dormancy. Macroscopic metastatic lesions are thought to be able to seed additional metastases (21 spread). It is unclear whether 21 spread can also occur from micrometastatic lesions, which is depicted with a question mark.

uveal melanoma will have metastatic disease in the liver, epithelial cancers have even demonstrated that spread of which is often the only site of metastasis.20 In cutaneous abnormal cells can precede primary tumor formation.23,24 These melanomas, this principle is also important with common sites disseminated epithelial cells are capable of persisting and of clinically detectable visceral metastasis including: lung, brain, subsequently transforming to overt metastatic growths at these liver and bone, while metastases to other sites are either less distant sites.23,25 In this context, it is notable that 4–12% of common or rarely encountered clinically.3 Even among common patients with metastatic melanoma do not have a clinically sites of melanoma metastasis, certain epidemiologic patterns identifiable primary tumor.26–28 There are different explanations suggest additional biological complexity. For example, liver for this observation, though one is that some of these metastatic metastasis and are inversely correlated.3 Our lesions represent malignant progression of abnormal melanocytes current understanding of the molecular mechanisms underlying that had spread previously. melanoma metastasis to different organs has been recently In fact, non-malignant melanocytes are routinely visualized reviewed.21 outside of their normal epidermal location in humans. In compound melanocytic nevi, benign proliferations of melanocytes are found within the , demonstrating that spread through PREMALIGNANT DISSEMINATION AND PARALLEL the is not a unique feature of malignant PROGRESSION cells. Melanocytes can even be visualized in a subset of lymph In traditional models of metastasis, spread to spatially restricted nodes from normal, non-melanoma bearing patients.29–32 These sites is thought to be a unique feature of a subset of particularly melanocyte clusters in normal lymph nodes are referred to as aggressive tumor cells that arise late in the course of disease. nodal nevi. Interestingly, many nodal nevi bear BRAFV600E- However, some observations in melanoma and other cancers activating .33 BRAFV600E mutations are also found in argue that spread of tumor cells can occur much earlier during most benign cutaneous melanocytic nevi (mutations in melanoma primary tumor formation. For example, kinetic models of uveal are discussed in more detail below),34 leading some authors to melanoma based on tumor doubling times estimate that on speculate that nodal nevi could represent cells that have spread average, metastatic spread of tumor cells to the liver is initiated up from these benign lesions.35 However, it is very difficult to to 5 years before identification and removal of the primary definitely exclude the possibility that nodal melanocytes are tumor.22 Genetically engineered mouse models (GEMMs) of instead remnants of embryonic development, or embryonic rests.

Oncogene (2014) 2413 – 2422 & 2014 Macmillan Publishers Limited Melanoma metastasis WE Damsky et al 2415 Accordingly, direct evidence supporting early dissemination of Although the prevalence of MITF and b-catenin dysregulation in metastatic cells in melanoma is currently absent. melanoma is clear, the effect of this dysregulation with respect to The hypothesis that disseminated precancerous or even frankly melanoma progression and metastasis is debated. Increased MITF malignant cells can persist at distant sites and retain the potential levels and/or levels of melanocytic differentiation antigens in to progress to clinically relevant metastatic disease is often referred human melanomas have been associated with both a relatively to as the parallel progression model of metastasis.36,37 The parallel good outcome58,59 and a relatively poor outcome.60–62 Similarly, progression model has significant conceptual overlap with attempts to correlate b-catenin levels with outcome have been metastatic dormancy. In this model, continued genetic and/or challenging to interpret. Most consistently, increased b-catenin epigenetic evolution of tumor cells at distant sites facilitates has been associated with an improved outcome in some larger progression to overt metastasis. A prediction of the parallel studies, particularly when combined with additional markers into progression model is that DNA sequence and/or epigenetic prognostic models.63–65 Some smaller, previously published modifications should be relatively disparate between primary and reports did not find this relationship between increased metastatic lesions if continued evolution at distant sites is required b-catenin levels and poor outcome and some suggested, rather, for progression to overt metastasis. Recent efforts comparing DNA that b-catenin dysregulation is associated with disease sequence between matched primary and metastatic tumors in a progression and metastasis.52,66,67 Of course, correlating patient handful of melanoma patients have shown both a relative outcome with levels or staining patterns is difficult to homogeneity38,39 and a relative heterogeneity40 between interpret at a mechanistic level. b-catenin is thought to have matched lesions. Similar studies in epithelial cancers have more different functions depending on its subcellular localization, so commonly reported relative homogeneity between matched levels in tissue likely do not accurately represent functional primary and metastatic lesions, but significant heterogeneity in activity. Finally, prognosis is complex and represents several DNA sequence has also been reported.41 These observations aspects of disease biology, not only the likelihood of metastasis. suggest that relatively early dissemination with parallel progression Unfortunately, studies designed to functionally evaluate the and relatively late dissemination with limited evolution at distant roles of MITF and b-catenin in melanoma progression have also sites are both pathways with the potential to give rise to clinically been difficult to interpret as a whole.68 For example, increased meaningful metastatic disease. However, as it is not clear if- and/or levels of MITF have been shown to both increase69,70 and which- of these additional genetic changes have a functionally decrease71–74 metastasis and/or surrogate measures of important role in driving disease progression, these conclusions metastasis in experimental models of melanoma. Similarly, based on sequencing data are still fairly speculative. The degree to seemingly contradictory results are found with respect to the which early dissemination and parallel progression contribute to consequences of increased b-catenin in melanoma progression, metastatic melanoma pathogenesis on a population scale remains with some studies describing a pro-progression role for b-catenin to be seen. and others describing a progression restricting role.75–78 In an attempt to reconcile these observations, several hypo- theses have been proposed. Some groups have suggested that DIFFERENTIATION IN METASTASIS a relative balance in the degree of melanocytic differentiation is The degree and type of differentiation of tumor cells with respect important in driving metastasis, where both too much or too to their ability to metastasize is perhaps the most well-studied little can restrict progression.75,79 A separate hypothesis, the relationship in metastasis research. Epithelial–mesenchymal phenotypic plasticity (or phenotype switching) model, proposes transition is a model of metastasis developed in the study of that transient changes in the degree of melanocytic differentiation epithelial cancers, which posits that relative loss of epithelial in a particular melanoma may help to facilitate the different steps differentiation and relative gain of mesenchymal characteristics of metastasis for which they are advantageous.80–82 In this model, facilitates early steps in metastasis, such as local tissue increased melanocytic differentiation is thought to drive local and intravasation.42 Despite being a widely accepted paradigm for proliferation, while relative loss of melanocytic differentiation is cancer metastasis, epithelial–mesenchymal transition is not thought to facilitate the metastatic spread itself. This model is without controversy, even within epithelial cancers.43 Although somewhat consistent with recent studies in epithelial cancers that melanocytes share some features of differentiated epithelial cells, suggest different cellular programs drive the distinct processes of they are derived from the neural crest and have a differentiation tumor cell spread and tumor cell proliferation at distant sites.83,84 program related to pigment production,44 suggesting that the Other groups, including ours, have suggested instead that relationship between differentiation and metastasis in melanoma increased melanocytic differentiation has context-specific effects may be even more complex. with respect to progression and metastasis. A recent study by In melanocytes, differentiation is largely controlled by micro- Gallagher et al.85 suggested that while b-catenin activation in phthalmia-associated (MITF) and WNT/ melanocytes restricts cellular motility and migration, b-catenin b-catenin signaling. MITF is a transcription factor central to activation in melanomas enhances metastasis when measured melanocyte survival, proliferation and -pigment in a GEMM. Data from additional melanoma GEMMs production.45 Canonical WNT signaling through b-catenin also developed in our lab also suggests that the effects of b-catenin promotes increased melanocytic differentiation through direct and Mitf may be context-dependent.86 In these GEMMs, the effect transcriptional upregulation of MITF.46 b-catenin is also required of increased melanocytic differentiation on metastasis was for melanocyte survival47 and differentiated melanocyte dependent on concurrent genetic changes within the tumor. For function.48 Both MITF and b-catenin signaling are dysregulated example, in GEMMs with mutant Braf and inactivated Pten, in human melanomas. MITF is amplified in 10% of primary b-catenin substantially increased metastasis to the lungs, while in melanomas and 21% of metastases.49 Germline MITF mutations other GEMMs with wild-type Pten, b-catenin seemed to promote that alter MITF function are associated with an increased local tumor growth, but not metastasis.86 lifetime risk of melanoma.50,51 Oncogenic b-catenin mutations A last example of the additional layers of complexity in are found in B5–7% of melanomas and are thought to lead discerning the effect of b-catenin in melanoma progression is to constitutive activation of the canonical WNT pathway.52–55 illustrated by the recent observation that b-catenin in melanoma More broadly, WNT/b-catenin pathway activation can occur cells has substantial immunosuppressive effects in the local tumor through a variety of mechanisms in addition to of microenvironment.87 Assays that study melanoma metastasis, in b-catenin,56 with evidence of pathway activation in at least 1/3 of for example, an immunodeficient mouse model may not account melanomas.57 for these potential tumor-extrinsic effects of b-catenin activation.

& 2014 Macmillan Publishers Limited Oncogene (2014) 2413 – 2422 Melanoma metastasis WE Damsky et al 2416 Context is of critical importance when studying metastasis in which have reciprocal interactions with tumor cells. Tumor mouse models whether it is genetic background of the tumor, microenvironments vary during different steps in progression immune status of the host or another variable. A variety of and metastasis and can both inhibit and facilitate progres- approaches and mouse model systems have been employed, sion.16,17,41,88 In melanoma, changes in the tumor micro- many of which are summarized in Figure 2. Importantly, these environment can have very powerful effects on tumor cells. approaches do not study metastasis in a broadly comparable way, For example, embryonic and skeletal muscle microenvironments an additional consideration that must be taken into account when can restrict both proliferative and metastatic phenotypes.89,90 interpreting experimental data regarding metastasis. Context is In mouse models of melanoma metastasis, the microenvironment also of critical importance when interpreting staining patterns in at distant sites is thought to be critical in eliminating disseminated human melanomas. Staining patterns for a particular protein in tumor cells and preventing both formation of micrometastases, as primary versus metastatic, treated versus untreated or PTEN wild- well as progression of micrometastases to macrometastases.91 type versus PTEN mutant melanomas, for example, may not be Interestingly, the microenvironment can also positively influence broadly comparable. MITF and b-catenin in melanoma serve as an tumor progression. In epithelial cancers, for example, loss of the important example of these context-specific considerations in the transforming growth factor-bII receptor in stromal fibroblasts can study of metastasis. Mouse models and the particular advantages initiate neoplasia in otherwise unmodified epithelial cells.92 of GEMMs will be discussed further below. Immune cells are one particularly well-studied component of the in both melanoma and other cancers. Although the effects of the immune system in melanoma TUMOR MICROENVIRONMENT AND MACROENVIRONMENT will not be reviewed in detail here, in general, adaptive immunity Tumor microenvironments are defined by both cellular and is thought to help constrain progression of both primary and non-cellular constituents in the tissue immediately surrounding metastatic disease.14,93 Components of the innate immune system tumor cells. These factors include: stromal and immune cells, such as macrophages, on the other hand, have been shown to extracellular matrix, soluble factors and mechanical forces, all of facilitate both primary tumor progression and metastasis in a

Figure 2. Approaches for studying metastasis in mice. A variety of methods are used to study metastasis in mice, the most common of which are depicted here. Each of these approaches has relative advantages and disadvantages, including which individual steps of metastasis can be studied using the approach. The steps that can be studied using each method are shown as colored boxes. The immune status of the mouse host is also an important consideration. Immunodeficient mouse models do not account for the effect of the immune system on progression, while immunocompetent mouse models do not allow for the use of human melanoma cells. (a) In orthotopic models, melanoma cells are injected directly into the desired site of metastasis (i.e., the brain) leading to formation of a mass. Either human or murine melanoma cells can be injected into an immunodeficient mouse. Only murine melanoma cells can be injected into an immunocompetent mouse, which must be syngeneic (i.e., B16 melanoma cells into a c57bl/6 mouse). (b) Melanoma cells can be injected intravenously to simulate metastasis. With this approach either human or murine melanoma cells can be injected into an immunodeficient mouse. Murine melanoma cells can be injected into an immunocompetent, syngeneic mouse. (c) Melanoma cells can be injected subcutaneously leading to formation of a mass, which can subsequently metastasize to other sites. This approach allows for the use of either human or murine melanoma cells in immunodeficient mice or murine melanoma cells in immunocompetent, syngeneic mice. (d) GEMMs are based on the introduction of genetic changes commonly found in human melanomas, such as BrafV600E, specifically to the melanocytes of mice. Appropriate combinations of genetic changes result in the spontaneous formation of melanomas from endogenous melanocytes in an immunocompetent mouse. GEMMs recapitulate all steps of melanoma pathogenesis; however, study of human melanoma cells is not possible with GEMMs.

Oncogene (2014) 2413 – 2422 & 2014 Macmillan Publishers Limited Melanoma metastasis WE Damsky et al 2417 variety of experimental assays.94 In human melanomas, increased mutations to BRAF and NRAS are also found in the majority of macrophage staining is associated with poor outcome.95 The benign melanocytic nevi, suggesting that these changes represent relationship between the immune system and melanoma relatively early, although not independently sufficient, events in metastasis has been recently reviewed.96 melanomagenesis. Even the earliest genetic changes in primary Tumor cell extrinsic factors that act in an organism-wide tumor formation have been postulated to have a functionally fashion are also likely to be important in melanoma metastasis. important role in metastasis,114 which is consistent with the Perhaps the most well-known example of this type of ‘macro- observed early dissemination of abnormal cells in epithelial cancer environmental’ effect is the premetastatic niche. In this model, GEMMs discussed above. The hypothesis that early genetic soluble tumor-derived factors circulate systemically and can enact alterations drive many aspects of tumor progression is also complex programs that alter distant tissues in a way that facilitates consistent with the observation in human melanomas that later metastasis to those sites.97 Recent work has implicated proliferation rate115,116 and thickness4 of primary tumors are tumor-derived exosomes containing melanocytic antigens in very strongly correlated with the likelihood of metastasis. initiating premetastatic niche formation in melanoma.98,99 Epidemiologic observations from human melanomas suggest Other examples of interactions between established, spatially that there is no difference in rates of metastasis between BRAF and separated malignant lesions through endocrine-like mechanisms NRAS mutant melanomas,117,118 though both may exhibit higher have also been described,100,101 though the relative importance rates of metastasis than RAS/RAF wild-type melanomas.119 of these observations in human melanoma metastasis remains Interestingly, a recent study suggested that BRAFV600K-driven unclear. Additional macroenvironmental factors such as systemic melanomas may be more metastatic than BRAFV600E-driven inflammation,102–104 organismal metabolism,105–108 host genetic melanomas,120 suggesting that the metastasis promoting effect background109–111 and even male gender112 can all influence the of NRAS and/or BRAF may be more complex than mutation time course of progression and pattern of metastasis. An presence or absence alone. improved understanding of these macroenvironmental changes Functional studies of mutant RAS and RAF have reinforced the and the specific manner in which they affect tumor and tumor role of these in metastasis. For example, melanoma microenvironmental biology will be important moving forward. GEMMs have shown that NRAS mutant melanomas are inherently more metastatic than melanomas driven by other mutant RAS isoforms.121,122 In other mouse models, inhibition of mitogen- MUTATIONS AND METASTASIS activated protein /extracellular-signal-regulated kinase Our understanding of the genetic alterations commonly found in signaling can block lung metastasis.118 Although a specific human melanomas has advanced significantly over the past mechanism by which RAS or RAF mutation drives metastasis in several years (Figure 3). The mitogen-activated protein kinase/ melanoma is unclear, recent studies have proposed mechanisms extracellular-signal-regulated kinase pathway is the most com- that relate BRAF mutation to cell invasiveness.123,124 monly mutated pathway in melanoma, with BRAF and NRAS being The PI3K/AKT pathway is also frequently constitutively activated the most commonly mutated components.113 Activating in melanoma, most commonly through genetic or epigenetic

Figure 3. Recurrent alterations to key signaling pathways in melanoma. Proteins depicted in green are recurrently activated in melanoma. Mechanisms for activation include specific gain-of-function mutations, DNA copy number gains, and/or overexpression. Proteins depicted in blue are recurrently inactivated in melanoma. Mechanisms of inactivation include: point mutations, genomic losses and epigenetic silencing. Proteins depicted in gray are key effectors of pathways that are frequently hyper-activated in melanoma, but are not themselves commonly intrinsically dysregulated. Various negative feedback loops exist within and between pathways, though these are not depicted for simplicity. *Germline MC1R polymorphisms are related to altered pigment production and increased risk of melanoma. **Mutations occur in the TERT promoter rather than in coding DNA sequence.

& 2014 Macmillan Publishers Limited Oncogene (2014) 2413 – 2422 Melanoma metastasis WE Damsky et al 2418 inactivation of the PTEN tumor suppressor125,126 and copy number cells never progress to a macroscopic metastasis.153 Together, gains or overexpression of AKT3.127,128 PTEN has been shown to these experimental observations begin to shift the focus towards suppress melanoma metastasis in experimental assays.129 later steps in metastasis. Melanoma GEMMs support a critical role of Pten loss and Patterns of metastasis observed during autopsies of melanoma subsequent activation of the PI3K/Akt pathway in driving patients suggest that not all disseminated melanoma leads to metastasis of melanomas initiated by activating Ras and Raf clinically relevant metastatic disease. Metastatic tumor distribution mutations.130,131 In these models, however, activation of PI3K/Akt is often much more substantial than can be appreciated clinically is also a critical event in primary tumor formation, making specific while patients are alive. For example, intestinal metastasis is effects on metastasis difficult to infer. In additional melanoma detected in only 1–7% of patients clinically, but 26–58% of GEMMs, b-catenin-stabilizing mutations and Lkb1-inactivating patients will show evidence of intestinal metastasis at autopsy.3 mutations have been linked with increased metastasis and are Similarly, liver metastasis, while clinically evident in only 10–20% discussed further below. of patients with metastatic melanoma, is found in 54–77% In the past few years, genome-wide sequencing efforts patients at autopsy.3,154,155 In fact, it appears that subclinical have identified several new recurrent mutations in melanoma. melanoma metastases can manifest in almost any part of the Some of these mutations include: RAC1,55,132 PREX2,55 GRM3,133 body, including sites rarely encountered clinically.3,155,156 More GRIN2A133 and the TERT promoter.134 Discerning the functional sensitive assays for detecting disseminated tumor cells also consequences of these and other recurrent genetic aberrations in suggest that clinically inevident tumor burden may be much melanoma have the potential to uncover additional pathways higher than can be appreciated in melanoma patients. For regulating progression and metastasis (Figure 3). For example, example, micrometastatic disease can be detected by reverse PREX family guanine-nucleotide exchange factors have recently transcription–PCR and immunohistochemistry in large proportion been shown to be required for melanoma metastasis in a of patients and often in lieu of clinically detectable metastatic mouse model.135 Additional efforts have led to identification of disease.157 recurrent BAP1 mutations in the majority of metastatic uveal Other potential clues that growth at distant sites, rather than melanomas.109,136 simply arriving at distant sites, may be a critical rate-limiting step Other large scale approaches have been undertaken in an in the pathogenesis of melanoma metastasis comes from analysis attempt to identify metastasis regulators in melanoma by of draining lymph nodes from melanoma patients. The presence comparing DNA copy number changes137–139 and global mRNA or absence of tumor cells in sentinel lymph nodes has important expression between primary and metastatic melanomas.140 prognostic implications for melanoma patients.4 However, it has Unfortunately, to date these analyses have had little overlap in been shown that melanoma patients with histologic evidence of identified.140 Several other molecular mediators of melanoma spread to lymph nodes, but with tumor cell foci melanoma metastasis have been identified by a variety of o0.1 mm in size, have an equivalent prognosis to patients without approaches and include: Kiss-1,84 NEDD9,141 RhoC,142 MDA-9/ any evidence of spread to lymph nodes at all.158–160 Interestingly, syntenin,143 NM23144 and BRMS1,145 among others.146 complete removal of the draining bed does not improve survival in melanoma patients with evidence of spread.161,162 These observations may suggest that the presence RATE-LIMITING STEP IN MELANOMA METASTASIS? of tumor cells in the draining lymph nodes reflects two aspects of Metastasis is a complicated process with many different steps tumor biology: (1) the ability to disseminate and persist in distant (Figure 1). Traditionally, metastasis research has focused on early sites, and (2) the ability to progress to more clinically meaningful steps in metastasis such as: acquisition of cellular motility, local lesions at these sites. These additional observations, though tissue invasion and intravasation. Conversely, relatively less certainly somewhat speculative, may help to begin to synthesize emphasis has been placed on later steps in metastasis such as: newer experimental data with patterns of metastasis in human extravasation, survival, and both persistence and proliferation at melanoma patients. distant sites. Although these later steps in metastasis are These many considerations emphasize the importance of later inherently more difficult to study, observations in human steps in metastasis and suggest that progression of clinically melanoma patients suggest that understanding these processes undetectable disseminated melanoma to clinically detectable is critical for a comprehensive understanding of the pathogenesis macrometastatic disease may be a particularly important target for of metastatic melanoma. continued development of new adjuvant therapeutic approaches. Early metastasis work in the 1970s, suggested that dissemina- For example, bisphosphonate therapy, a strategy to make bone tion of malignant cells from a primary tumor is a fairly common microenvironments less hospitable to disseminated tumor cells, is event, with 41 million cells estimated to be shed per gram of now being used in an attempt to eliminate latent micrometastatic tumor per day.147 In addition to being relatively common, it was disease in the bones of some breast and prostate cancer also found that circulating tumor cells do not necessarily predict patients.163,164 Analogous, non-conventional adjuvant strategies whether metastasis has already or will ever occur.148 Despite more based on improved understanding of disseminated tumor cell recent technological improvements in the detection of circulating biology may be useful in targeting dormant micrometastatic tumor cells, newer studies still have not consistently found that disease in human melanoma patients. the presence of circulating tumor cells is related to outcome in melanoma.149 Further complicating this picture, and consistent with the early dissemination hypothesis, benign melanocytes and MOUSE MODELS nevus cells are routinely detected in the circulation of patients Mouse models have been critical in advancing our understanding without melanoma.150,151 of metastasis. Mouse models offer the ability to study different Studies tracking the fate of circulating tumor cells in experi- steps of cancer metastasis in a relatively physiologic manner.165 mental models of melanoma metastasis suggest that the Mice have been employed in a variety of different ways to study persistence of disseminated tumor cells at distant sites, the metastasis, each with intrinsic advantages and disadvantages progression of individual tumor cells to micrometastatic clusters of (Figure 2). In general, GEMMs are thought to particularly cells and the outgrowth of micrometastasis to clinically relevant accurately recapitulate the complex biology of tumor initiation, macrometastases are relatively rate-limiting events in metastasis progression and metastasis compared with other in vivo pathogenesis.100,152 Other studies using a variety of different approaches.166 In melanoma GEMMs, the introduction of approaches have also suggested that most disseminated tumor genetic changes specifically to endogenous melanocytes of mice

Oncogene (2014) 2413 – 2422 & 2014 Macmillan Publishers Limited Melanoma metastasis WE Damsky et al 2419 results in spontaneous progression of melanocytes to metastatic 6 Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB et al. melanoma in a background that is otherwise unbiased and Improved survival with ipilimumab in patients with metastatic melanoma. N Engl 166–168 unmanipulated. Further, GEMMs are not immuno- JMed2010; 363: 711–723. compromised and incorporate potentially critical effects of the 7 Clark Jr WH, Elder DE, Guerry Dt, Epstein MN, Greene MH, Van Horn M. A study of immune system in disease progression. Recently developed tumor progression: the precursor lesions of superficial spreading and GEMMs offer high rates of metastasis and will likely melanoma. Hum Pathol 1984; 15: 1147–1165. serve as new tools for in-depth study of the mechanisms of 8 Miller AJ, Mihm Jr MC. Melanoma. N Engl J Med. 2006; 355: 51–65. metastasis.86,169 In the Pten/Braf/b-catenin model developed in our 9 Balch CM, Urist MM, Karakousis CP, Smith TJ, Temple WJ, Drzewiecki K et al. Efficacy of 2-cm surgical margins for intermediate-thickness melanomas (1 to lab, very high rates of spontaneous metastasis occur reproducibly 86 4 mm). Results of a multi-institutional randomized surgical trial. Ann Surg 1993; to lymph nodes and lungs. 218: 262–267 (discussion 7–9). New GEMMs such as the Pten/Braf/b-catenin model offer unique 10 Crowley NJ, Seigler HF. Late recurrence of malignant melanoma. Analysis of 168 opportunities for the study of metastasis. For example, incorpor- patients. Ann Surg 1990; 212: 173–177. ating fluorescent reporters into these models will allow for an 11 Slingluff Jr CL, Dodge RK, Stanley WE, Seigler HF. The annual risk of melanoma unprecedented ability to visualize and purify cells from various progression. Implications for the concept of cure. Cancer 1992; 70: 1917–1927. steps of experimentally unmanipulated spontaneous metastasis. 12 Pierard GE, Pierard-Franchimont C, Reginster MA, Quatresooz P. Smouldering Intravital imaging technologies allow real-time visualization of malignant melanoma and metastatic dormancy: an update and review. Dermatol these processes and have been recently reviewed.170 Processes Res Pract 2012; 2012: 461278. 13 Koebel CM, Vermi W, Swann JB, Zerafa N, Rodig SJ, Old LJ et al. Adaptive such as early dissemination of abnormal cells, metastatic immunity maintains occult cancer in an equilibrium state. Nature 2007; 450: dormancy and progression of micrometastatic lesions to frank 903–907. metastases can be studied and manipulated in these models. 14 Eyles J, Puaux AL, Wang X, Toh B, Prakash C, Hong M et al. Tumor cells disseminate early, but immunosurveillance limits metastatic outgrowth, in a mouse model of melanoma. J Clin Invest 2010; 120: 2030–2039. CONCLUSIONS AND FUTURE DIRECTIONS 15 Folkman J. Role of angiogenesis in tumor growth and metastasis. Semin Oncol 2002; 29(6 Suppl 16): 15–18. A fundamental shift is underway in our conceptualization of 16 Ossowski L, Aguirre-Ghiso JA. Dormancy of metastatic melanoma. Pigment Cell melanoma metastasis. Although the linear Clark model has been Melanoma Res 2010; 23: 41–56. very useful in studying melanoma progression and metastasis, it is 17 Bissell MJ, Hines WC. Why don’t we get more cancer? A proposed role of the becoming clear that this model probably only represents the microenvironment in restraining cancer progression. Nat Med 2011; 17: 320–329. progression of a subset of melanomas, with many melanomas 18 Izraely S, Sagi-Assif O, Klein A, Meshel T, Tsarfaty G, Pasmanik-Chor M et al. The having more complex and less linear natural histories. As there are metastatic microenvironment: brain-residing melanoma metastasis and dormant many routes to primary tumor formation, there are likely also micrometastasis. Int J Cancer 2012; 131: 1071–1082. many routes to the establishment of clinically meaningful 19 Paget S. The distribution of secondary growths in cancer of the breast. Lancet 1889; 133: 571–573. metastatic disease. When studying melanoma metastasis, pro- 20 Bakalian S, Marshall JC, Logan P, Faingold D, Maloney S, Di Cesare S et al. gression may be better visualized as a network rather than a linear Molecular pathways mediating liver metastasis in patients with uveal melanoma. series, in which sequential progression through every step Clin Cancer Res 2008; 14: 951–956. represents only one pathway from normal melanocyte to 21 Damsky WE, Rosenbaum LE, Bosenberg M. Decoding melanoma metastasis. metastatic melanoma (Figure 1). Cancers 2010; 3: 126–163. Moving forward, it will be important to keep the context of new 22 Eskelin S, Pyrhonen S, Summanen P, Hahka-Kemppinen M, Kivela T. Tumor data regarding melanoma metastasis paramount when interpret- doubling times in metastatic malignant melanoma of the uvea: tumor pro- ing and integrating these observations into existing paradigms. gression before and after treatment. Ophthalmology 2000; 107: 1443–1449. Additionally, incorporating other mediators of tumor biology, such 23 Husemann Y, Geigl JB, Schubert F, Musiani P, Meyer M, Burghart E et al. Systemic spread is an early step in breast cancer. Cancer Cell 2008; 13: 58–68. as noncoding RNAs and newer paradigms in tumor biology, such 24 Rhim AD, Mirek ET, Aiello NM, Maitra A, Bailey JM, McAllister F et al. EMT and as cancer stem cells, into evolving models of metastasis will be dissemination precede pancreatic tumor formation. Cell 2012; 148: 349–361. helpful as the role of these entities becomes more clearly defined 25 Podsypanina K, Du YC, Jechlinger M, Beverly LJ, Hambardzumyan D, Varmus H. in melanoma. It is an exciting time in in which Seeding and propagation of untransformed mouse mammary cells in the lung. new tools are helping us to understand disease pathogenesis at a Science 2008; 321: 1841–1844. more fundamental level, and new technologies and infrastructure 26 Das Gupta T, Brasfield R. Metastatic melanoma: a clinicopathologic study. Cancer are allowing more rapid translation of these discoveries into novel 1964; 17: 1323–1339. treatments for melanoma patients. 27 Giuliano AE, Moseley HS, Morton DL. Clinical aspects of unknown primary melanoma. Ann Surg 1980; 191: 98–104. 28 Reintgen DS, McCarty KS, Woodard B, Cox E, Seigler HF. Metastatic malignant melanoma with an unknown primary. Surg Gynecol Obstet 1983; 156: 335–340. CONFLICT OF INTEREST 29 Bautista NC, Cohen S, Anders KH. Benign cells in axillary The authors declare no conflict of interest. lymph nodes. A prospective and immunohistochemical study with literature review. Am J Clin Pathol 1994; 102: 102–108. 30 Carson KF, Wen DR, Li PX, Lana AM, Bailly C, Morton DL et al. Nodal nevi and cutaneous melanomas. Am J Surg Pathol 1996; 20: 834–840. REFERENCES 31 Fisher CJ, Hill S, Millis RR. Benign lymph node inclusions mimicking metastatic 1 Little EG, Eide MJ. Update on the current state of melanoma incidence. Dermatol carcinoma. J Clin Pathol 1994; 47: 245–247. Clin 2012; 30: 355–361. 32 McCarthy SW, Palmer AA, Bale PM, Hirst E. Naevus cells in lymph nodes. 2 Bedrosian I, Faries MB, Guerry Dt, Elenitsas R, Schuchter L, Mick R et al. Pathology 1974; 6: 351–358. Incidence of sentinel node metastasis in patients with thin primary melanoma 33 Taube JM, Begum S, Shi C, Eshleman JR, Westra WH. Benign nodal nevi (o or ¼ 1 mm) with vertical growth phase. Ann Surg Oncol 2000; 7: 262–267. frequently harbor the activating BRAF mutation. Am J Surg Pathol 2009; 3 Balch CM, Houghton AN, Sober AJ, Soong SJ. Cutaneous Melanoma, 4th edn. 33: 568–571. Quality Medical Publishing: St Louis, MO, USA, 2003. 34 Kumar R, Angelini S, Snellman E, Hemminki K. BRAF mutations are common 4 Balch CM, Gershenwald JE, Soong SJ, Thompson JF, Atkins MB, Byrd DR et al. somatic events in melanocytic nevi. J Invest Dermatol 2004; 122: 342–348. Final version of 2009 AJCC melanoma staging and classification. J Clin Oncol 35 Ross AL, Sanchez MI, Grichnik JM. Nevogenesis: a benign metastatic process? 2009; 27: 6199–6206. ISRN Dermatol 2011; 2011: 813513. 5 Chapman PB, Hauschild A, Robert C, Haanen JB, Ascierto P, Larkin J et al. 36 Stoecklein NH, Klein CA. Genetic disparity between primary tumours, Improved survival with vemurafenib in melanoma with BRAF V600E mutation. disseminated tumour cells, and manifest metastasis. Int J Cancer 2010; 126: N Engl J Med 2011; 364: 2507–2516. 589–598.

& 2014 Macmillan Publishers Limited Oncogene (2014) 2413 – 2422 Melanoma metastasis WE Damsky et al 2420 37 Klein CA. Parallel progression of primary tumours and metastases. Nat Rev 63 Meyer S, Fuchs TJ, Bosserhoff AK, Hofstadter F, Pauer A, Roth V et al. A seven- Cancer 2009; 9: 302–312. marker signature and clinical outcome in malignant melanoma: a large-scale 38 Gartner JJ, Davis S, Wei X, Lin JC, Trivedi NS, Teer JK et al. Comparative exome tissue-microarray study with two independent patient cohorts. PLoS One 2012; 7: sequencing of metastatic lesions provides insights into the mutational pro- e38222. gression of melanoma. BMC Genomics 2012; 13: 505. 64 Gould Rothberg BE, Berger AJ, Molinaro AM, Subtil A, Krauthammer MO, Camp 39 Turajlic S, Furney SJ, Lambros MB, Mitsopoulos C, Kozarewa I, Geyer FC et al. RL et al. Melanoma prognostic model using tissue microarrays and genetic Whole genome sequencing of matched primary and metastatic acral melano- algorithms. J Clin Oncol 2009; 27: 5772–5780. mas. Genome Res 2012; 22: 196–207. 65 Kageshita T, Hamby CV, Ishihara T, Matsumoto K, Saida T, Ono T. Loss of beta- 40 Pleasance ED, Cheetham RK, Stephens PJ, McBride DJ, Humphray SJ, Greenman catenin expression associated with disease progression in malignant melanoma. CD et al. A comprehensive catalogue of somatic mutations from a human cancer Br J Dermatol 2001; 145: 210–216. genome. Nature 2010; 463: 191–196. 66 Kielhorn E, Provost E, Olsen D, D’Aquila TG, Smith BL, Camp RL et al. Tissue 41 Valastyan S, Weinberg RA. Tumor metastasis: molecular insights and evolving microarray-based analysis shows phospho-beta-catenin expression in malignant paradigms. Cell 2011; 147: 275–292. melanoma is associated with poor outcome. Int J Cancer 2003; 103: 652–656. 42 Kalluri R, Weinberg RA. The basics of epithelial-mesenchymal transition. J Clin 67 Tucci MG, Lucarini G, Brancorsini D, Zizzi A, Pugnaloni A, Giacchetti A et al. Invest 2009; 119: 1420–1428. Involvement of E-cadherin, beta-catenin, Cdc42 and CXCR4 in the progression 43 Tarin D, Thompson EW, Newgreen DF. The fallacy of epithelial mesenchymal and prognosis of cutaneous melanoma. Br J Dermatol 2007; 157: 1212–1216. transition in neoplasia. Cancer Res 2005; 65: 5996–6000 (discussion-1). 68 Webster MR, Weeraratna AT. A Wnt-er migration: the confusing role of beta- 44 Ernfors P. Cellular origin and developmental mechanisms during the formation catenin in melanoma metastasis. Sci Signal 2013; 6: pe11. of skin melanocytes. Exp Cell Res 2010; 316: 1397–1407. 69 McGill GG, Haq R, Nishimura EK, Fisher DE. c-Met expression is regulated by Mitf 45 Mitra D, Fisher DE. Transcriptional regulation in melanoma. Hematol Oncol Clin in the melanocyte lineage. J Biol Chem 2006; 281: 10365–10373. North Am 2009; 23: 447–465. 70 Seong I, Min HJ, Lee JH, Yeo CY, Kang DM, Oh ES et al. Sox10 controls migration 46 Widlund HR, Horstmann MA, Price ER, Cui J, Lessnick SL, Wu M et al. Beta- of B16F10 melanoma cells through multiple regulatory target genes. PLoS One catenin-induced melanoma growth requires the downstream target Micro- 2012; 7: e31477. phthalmia-associated transcription factor. J Cell Biol 2002; 158: 1079–1087. 71 Arozarena I, Bischof H, Gilby D, Belloni B, Dummer R, Wellbrock C. In melanoma, 47 Hari L, Brault V, Kleber M, Lee HY, Ille F, Leimeroth R et al. Lineage-specific beta-catenin is a suppressor of invasion. Oncogene 2011; 30: 4531–4543. requirements of beta-catenin in neural crest development. J Cell Biol 2002; 159: 72 Carreira S, Goodall J, Denat L, Rodriguez M, Nuciforo P, Hoek KS et al. Mitf 867–880. regulation of Dia1 controls melanoma proliferation and invasiveness. Genes Dev 48 Rabbani P, Takeo M, Chou W, Myung P, Bosenberg M, Chin L et al. Coordinated 2006; 20: 3426–3439. activation of Wnt in epithelial and melanocyte stem cells initiates pigmented 73 Javelaud D, Alexaki VI, Pierrat MJ, Hoek KS, Dennler S, Van Kempen L et al. GLI2 hair regeneration. Cell 2011; 145: 941–955. and M-MITF transcription factors control exclusive expression programs 49 Garraway LA, Widlund HR, Rubin MA, Getz G, Berger AJ, Ramaswamy S et al. and inversely regulate invasion in human melanoma cells. Pigment Cell Mela- Integrative genomic analyses identify MITF as a lineage survival oncogene noma Res 2011; 24: 932–943. amplified in malignant melanoma. Nature 2005; 436: 117–122. 74 Cheli Y, Giuliano S, Fenouille N, Allegra M, Hofman V, Hofman P et al. Hypoxia 50 Bertolotto C, Lesueur F, Giuliano S, Strub T, de Lichy M, Bille K et al. and MITF control metastatic behaviour in mouse and human melanoma cells. A SUMOylation-defective MITF germline mutation predisposes to melanoma and Oncogene 2012; 31: 2461–2470. renal carcinoma. Nature 2011; 480: 94–98. 75 Yang PT, Anastas JN, Toroni RA, Shinohara MM, Goodson JM, Bosserhoff AK et al. 51 Yokoyama S, Woods SL, Boyle GM, Aoude LG, MacGregor S, Zismann V et al. WLS inhibits melanoma cell proliferation through the beta-catenin signalling A novel recurrent mutation in MITF predisposes to familial and sporadic pathway and induces spontaneous metastasis. EMBO Mol Med 2012; 4: melanoma. Nature 2011; 480: 99–103. 1294–1307. 52 Demunter A, Libbrecht L, Degreef H, De Wolf-Peeters C, van den Oord JJ. Loss of 76 Takahashi Y, Nishikawa M, Suehara T, Takiguchi N, Takakura Y. Gene silencing of membranous expression of beta-catenin is associated with tumor progression in beta-catenin in melanoma cells retards their growth but promotes the formation cutaneous melanoma and rarely caused by exon 3 mutations. Mod Pathol 2002; of pulmonary metastasis in mice. Int J Cancer 2008; 123: 2315–2320. 15: 454–461. 77 Sinnberg T, Menzel M, Kaesler S, Biedermann T, Sauer B, Nahnsen S et al. 53 Forbes SA, Tang G, Bindal N, Bamford S, Dawson E, Cole C et al. COSMIC Suppression of casein kinase 1alpha in melanoma cells induces a switch in (the Catalogue of Somatic Mutations in Cancer): a resource to investigate acquired mutations in human cancer. Nucleic Acids Res 2010; 38: (Database issue) beta-catenin signaling to promote metastasis. Cancer Res 2010; 70: 6999–7009. D652–D657. 78 Grossmann AH, Yoo JH, Clancy J, Sorensen LK, Sedgwick A, Tong Z et al. The 54 Omholt K, Platz A, Ringborg U, Hansson J. Cytoplasmic and nuclear accumulation small GTPase ARF6 stimulates beta-catenin transcriptional activity during of beta-catenin is rarely caused by CTNNB1 exon 3 mutations in cutaneous WNT5A-mediated melanoma invasion and metastasis. Sci Signal 2013; 6: ra14. malignant melanoma. Int J Cancer 2001; 92: 839–842. 79 Yajima I, Kumasaka MY, Thang ND, Goto Y, Takeda K, Iida M et al. Molecular 55 Hodis E, Watson IR, Kryukov GV, Arold ST, Imielinski M, Theurillat JP et al. A network associated with MITF in Skin Melanoma development and progression. landscape of driver mutations in melanoma. Cell 2012; 150: 251–263. J 2011; 2011: 730170. 56 Larue L, Delmas V. The WNT/Beta-catenin pathway in melanoma. Front Biosci 80 Pinner S, Jordan P, Sharrock K, Bazley L, Collinson L, Marais R et al. Intravital 2006; 11: 733–742. imaging reveals transient changes in pigment production and Brn2 expression 57 Rimm DL, Caca K, Hu G, Harrison FB, Fearon ER. Frequent nuclear/cytoplasmic during metastatic melanoma dissemination. Cancer Res 2009; 69: 7969–7977. localization of beta-catenin without exon 3 mutations in malignant melanoma. 81 Eichhoff OM, Weeraratna A, Zipser MC, Denat L, Widmer DS, Xu M et al. Dif- Am J Pathol 1999; 154: 325–329. ferential LEF1 and TCF4 expression is involved in melanoma cell phenotype 58 Nazarian RM, Prieto VG, Elder DE, Duncan LM. Melanoma expression switching. Pigment Cell Melanoma Res 2011; 24: 631–642. in melanocytic tumor progression: a tissue microarray study. J Cutan Pathol 82 Hoek KS, Goding CR. Cancer stem cells versus phenotype-switching in mela- 2010; 37(Suppl 1): 41–47. noma. Pigment Cell Melanoma Res 2010; 23: 746–759. 59 Makhzami S, Rambow F, Delmas V, Larue L. Efficient gene expression profiling of 83 Tsai JH, Donaher JL, Murphy DA, Chau S, Yang J. Spatiotemporal regulation of laser-microdissected melanoma metastases. Pigment Cell Melanoma Res 2012; epithelial-mesenchymal transition is essential for 25: 783–791. metastasis. Cancer Cell 2012; 22: 725–736. 60 Ugurel S, Houben R, Schrama D, Voigt H, Zapatka M, Schadendorf D et al. 84 Ocana OH, Corcoles R, Fabra A, Moreno-Bueno G, Acloque H, Vega S et al. Microphthalmia-associated transcription factor gene amplification in metastatic Metastatic colonization requires the repression of the epithelial-mesenchymal melanoma is a prognostic marker for patient survival, but not a predictive transition inducer prrx1. Cancer Cell 2012; 22: 709–724. marker for chemosensitivity and response. Clin Cancer Res 2007; 85 Gallagher SJ, Rambow F, Kumasaka M, Champeval D, Bellacosa A, Delmas V et al. 13: 6344–6350. Beta-catenin inhibits melanocyte migration but induces melanoma metastasis. 61 Gradilone A, Gazzaniga P, Ribuffo D, Bottoni U, Frati L, Aglian AM et al. Prog- Oncogene 2013; 32: 2230–2238. nostic significance of expression in for 86 Damsky WE, Curley DP, Santhanakrishnan M, Rosenbaum LE, Platt JT, Gould ultra-thin, thin, and thick melanomas. Eur Rev Med Pharmacol Sci 2012; 16: Rothberg BE et al. Beta-catenin signaling controls metastasis in Braf-activated 1367–1376. Pten-deficient melanomas. Cancer Cell 2011; 20: 741–754. 62 Journe F, Id Boufker H, Van Kempen L, Galibert MD, Wiedig M, Sales F et al. 87 Yaguchi T, Goto Y, Kido K, Mochimaru H, Sakurai T, Tsukamoto N et al. Immune TYRP1 mRNA expression in melanoma metastases correlates with clinical out- suppression and resistance mediated by constitutive activation of Wnt/beta- come. Br J Cancer 2011; 105: 1726–1732. catenin signaling in human melanoma cells. J Immunol 2012; 189: 2110–2117.

Oncogene (2014) 2413 – 2422 & 2014 Macmillan Publishers Limited Melanoma metastasis WE Damsky et al 2421 88 Wirtz D, Konstantopoulos K, Searson PC. The physics of cancer: the role of 113 Lopez-Bergami P. The role of mitogen- and stress-activated protein kinase physical interactions and mechanical forces in metastasis. Nat Rev Cancer 2011; pathways in melanoma. Pigment Cell Melanoma Res 2011; 24: 902–921. 11: 512–522. 114 Bernards R, Weinberg RA. A progression puzzle. Nature 2002; 418: 823. 89 Parlakian A, Gomaa I, Solly S, Arandel L, Mahale A, Born G et al. Skeletal muscle 115 Frahm SO, Schubert C, Parwaresch R, Rudolph P. High proliferative activity may phenotypically converts and selectively inhibits metastatic cells in mice. PLoS predict early metastasis of thin melanomas. Hum Pathol 2001; 32: 1376–1381. One 2010; 5: e9299. 116 Boni R, Doguoglu A, Burg G, Muller B, Dummer R. MIB-1 immunoreactivity cor- 90 Hendrix MJ, Seftor EA, Seftor RE, Kasemeier-Kulesa J, Kulesa PM, Postovit LM. relates with metastatic dissemination in primary thick cutaneous melanoma. Reprogramming metastatic tumour cells with embryonic microenvironments. J Am Acad Dermatol 1996; 35(3 Pt 1): 416–418. Nat Rev Cancer 2007; 7: 246–255. 117 Colombino M, Capone M, Lissia A, Cossu A, Rubino C, De Giorgi V et al. BRAF/ 91 Luzzi KJ, MacDonald IC, Schmidt EE, Kerkvliet N, Morris VL, Chambers AF et al. NRAS mutation frequencies among primary tumors and metastases in patients Multistep nature of metastatic inefficiency: dormancy of solitary cells after with melanoma. J Clin Oncol 2012; 30: 2522–2529. successful extravasation and limited survival of early micrometastases. Am J 118 Rozenberg GI, Monahan KB, Torrice C, Bear JE, Sharpless NE. Metastasis in an Pathol 1998; 153: 865–873. orthotopic murine model of melanoma is independent of RAS/RAF mutation. 92 Bhowmick NA, Chytil A, Plieth D, Gorska AE, Dumont N, Shappell S et al. TGF-beta Melanoma Res 2010; 20: 361–371. signaling in fibroblasts modulates the oncogenic potential of adjacent epithelia. 119 Jakob JA, Bassett Jr RL, Ng CS, Curry JL, Joseph RW, Alvarado GC et al. NRAS Science 2004; 303: 848–851. mutation status is an independent prognostic factor in metastatic melanoma. 93 Carmi Y, Rinott G, Dotan S, Elkabets M, Rider P, Voronov E et al. Microenviron- Cancer 2012; 118: 4014–4023. ment-derived IL-1 and IL-17 interact in the control of lung metastasis. J Immunol 120 El-Osta H, Falchook G, Tsimberidou A, Hong D, Naing A, Kim K et al. BRAF 2011; 186: 3462–3471. mutations in advanced cancers: clinical characteristics and outcomes. PLoS One 94 Qian BZ, Pollard JW. Macrophage diversity enhances tumor progression and 2011; 6: e25806. metastasis. Cell 2010; 141: 39–51. 121 Chin L, Pomerantz J, Polsky D, Jacobson M, Cohen C, Cordon-Cardo C et al. 95 Jensen TO, Schmidt H, Moller HJ, Hoyer M, Maniecki MB, Sjoegren P et al. Cooperative effects of INK4a and ras in melanoma susceptibility in vivo. Genes Macrophage markers in serum and tumor have prognostic impact in American Dev 1997; 11: 2822–2834. Joint Committee on Cancer stage I/II melanoma. J Clin Oncol 2009; 27: 122 Ackermann J, Frutschi M, Kaloulis K, McKee T, Trumpp A, Beermann F. Metas- 3330–3337. tasizing melanoma formation caused by expression of activated N-RasQ61K on 96 Melnikova VO, Bar-Eli M. Inflammation and melanoma metastasis. Pigment Cell an INK4a-deficient background. Cancer Res 2005; 65: 4005–4011. Melanoma Res 2009; 22: 257–267. 123 Arozarena I, Sanchez-Laorden B, Packer L, Hidalgo-Carcedo C, Hayward R, Viros A 97 Peinado H, Lavotshkin S, Lyden D. The secreted factors responsible for pre- et al. Oncogenic BRAF induces melanoma cell invasion by downregulating the cGMP-specific phosphodiesterase PDE5A. Cancer Cell 2011; 19: 45–57. metastatic niche formation: old sayings and new thoughts. Semin Cancer Biol 124 Ferguson J, Arozarena I, Ehrhardt M, Wellbrock C. Combination of MEK and SRC 2011; 21: 139–146. inhibition suppresses melanoma cell growth and invasion. Oncogene 2013; 32: 98 Hood JL, San RS, Wickline SA. Exosomes released by melanoma cells prepare 86–96. sentinel lymph nodes for tumor metastasis. Cancer Res 2011; 71: 3792–3801. 125 Tsao H, Zhang X, Benoit E, Haluska FG. Identification of PTEN/MMAC1 alterations 99 Peinado H, Aleckovic M, Lavotshkin S, Matei I, Costa-Silva B, Moreno-Bueno G in uncultured melanomas and melanoma cell lines. Oncogene 1998; 16: et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro- 3397–3402. metastatic phenotype through MET. Nat Med 2012; 18: 883–891. 126 Zhou XP, Gimm O, Hampel H, Niemann T, Walker MJ, Eng C. Epigenetic PTEN 100 McAllister SS, Gifford AM, Greiner AL, Kelleher SP, Saelzler MP, Ince TA et al. silencing in malignant melanomas without PTEN mutation. Am J Pathol 2000; Systemic endocrine instigation of indolent tumor growth requires . 157: 1123–1128. Cell 2008; 133: 994–1005. 127 Ko JM, Velez NF, Tsao H. Pathways to melanoma. Semin Cutan Med Surg 2010; 29: 101 Demicheli R, Retsky MW, Hrushesky WJ, Baum M. Tumor dormancy and surgery- 210–217. driven interruption of dormancy in breast cancer: learning from failures. Nat Clin 128 Stahl JM, Sharma A, Cheung M, Zimmerman M, Cheng JQ, Bosenberg MW et al. Pract Oncol 2007; 4: 699–710. Deregulated Akt3 activity promotes development of malignant melanoma. 102 Rothwell PM, Wilson M, Price JF, Belch JF, Meade TW, Mehta Z. Effect of daily Cancer Res 2004; 64: 7002–7010. aspirin on risk of cancer metastasis: a study of incident cancers during rando- 129 Madhunapantula SV, Robertson GP. The PTEN-AKT3 signaling cascade as a mised controlled trials. Lancet 2012; 379: 1591–1601. therapeutic target in melanoma. Pigment Cell Melanoma Res 2009; 22: 400–419. 103 Algra AM, Rothwell PM. Effects of regular aspirin on long-term cancer incidence 130 Dankort D, Curley DP, Cartlidge RA, Nelson B, Karnezis AN, Damsky Jr WE et al. and metastasis: a systematic comparison of evidence from observational studies Braf(V600E) cooperates with Pten loss to induce metastatic melanoma. Nat versus randomised trials. Lancet Oncol 2012; 13: 518–527. Genet 2009; 41: 544–552. 104 Karnezis T, Shayan R, Caesar C, Roufail S, Harris NC, Ardipradja K et al. VEGF-D 131 Nogueira C, Kim KH, Sung H, Paraiso KH, Dannenberg JH, Bosenberg M et al. promotes tumor metastasis by regulating prostaglandins produced by the Cooperative interactions of PTEN deficiency and RAS activation in melanoma collecting lymphatic endothelium. Cancer Cell 2012; 21: 181–195. metastasis. Oncogene 2010; 29: 6222–6232. 105 Calle EE, Rodriguez C, Walker-Thurmond K, Thun MJ. Overweight, , and 132 Krauthammer M, Kong Y, Ha BH, Evans P, Bacchiocchi A, McCusker JP et al. mortality from cancer in a prospectively studied cohort of U.S. adults. NEnglJ Exome sequencing identifies recurrent somatic RAC1 mutations in melanoma. Med 2003; 348: 1625–1638. Nat Genet 2012; 44: 1006–1014. 106 Haakinson DJ, Leeds SG, Dueck AC, Gray RJ, Wasif N, Stucky CC et al. The impact 133 Prickett TD, Wei X, Cardenas-Navia I, Teer JK, Lin JC, Walia V et al. Exon capture of obesity on breast cancer: a retrospective review. Ann Surg Oncol 2012; 19: analysis of G protein-coupled receptors identifies activating mutations in GRM3 3012–3018. in melanoma. Nat Genet 2011; 43: 1119–1126. 107 Wu C, Aronson WJ, Terris MK, Presti Jr JC, Kane CJ, Amling CL et al. Diabetes 134 Huang FW, Hodis E, Xu MJ, Kryukov GV, Chin L, Garraway LA. Highly recurrent predicts metastasis after radical prostatectomy in obese men: results from the TERT promoter mutations in human melanoma. Science 2013; 339: 957–959. SEARCH database. BJU Int (e-pub ahead of print 10 January 2013; doi: 10.1111/ 135 Lindsay CR, Lawn S, Campbell AD, Faller WJ, Rambow F, Mort RL et al. P-Rex1 is j.1464-410X.2012.11687.x). required for efficient melanoblast migration and melanoma metastasis. Nat 108 Nagel G, Bjorge T, Stocks T, Manjer J, Hallmans G, Edlinger M et al. Metabolic risk Commun 2011; 2: 555. factors and skin cancer in the Metabolic Syndrome and Cancer Project (Me-Can). 136 Harbour JW, Onken MD, Roberson ED, Duan S, Cao L, Worley LA et al. Frequent Br J Dermatol 2012; 167: 59–67. mutation of BAP1 in metastasizing uveal melanomas. Science 2010; 330: 109 Njauw CN, Kim I, Piris A, Gabree M, Taylor M, Lane AM et al. Germline BAP1 1410–1413. inactivation is preferentially associated with metastatic ocular melanoma and 137 Kabbarah O, Nogueira C, Feng B, Nazarian RM, Bosenberg M, Wu M et al. Inte- cutaneous-ocular melanoma families. PLoS One 2012; 7: e35295. grative genome comparison of primary and metastatic melanomas. PLoS One 110 Crawford NP, Ziogas A, Peel DJ, Hess J, Anton-Culver H, Hunter KW. Germline 2010; 5: e10770. polymorphisms in SIPA1 are associated with metastasis and other indicators of 138 Valsesia A, Rimoldi D, Martinet D, Ibberson M, Benaglio P, Quadroni M et al. poor prognosis in breast cancer. Breast Cancer Res 2006; 8: R16. Network-guided analysis of genes with altered somatic copy number and gene 111 Faraji F, Pang Y, Walker RC, Nieves Borges R, Yang L, Hunter KW. Cadm1 is a expression reveals pathways commonly perturbed in metastatic melanoma. metastasis susceptibility gene that suppresses metastasis by modifying tumor PLoS One 2011; 6: e18369. interaction with the cell-mediated immunity. PLoS Genet 2012; 8: e1002926. 139 Scott KL, Nogueira C, Heffernan TP, van Doorn R, Dhakal S, Hanna JA et al. 112 Mervic L. Time course and pattern of metastasis of cutaneous melanoma differ Proinvasion metastasis drivers in early-stage melanoma are . Cancer between men and women. PLoS One 2012; 7: e32955. Cell 2011; 20: 92–103.

& 2014 Macmillan Publishers Limited Oncogene (2014) 2413 – 2422 Melanoma metastasis WE Damsky et al 2422 140 Timar J, Gyorffy B, Raso E. Gene signature of the metastatic potential of cuta- 157 Borthwick NJ, Thombs J, Polak M, Gabriel FG, Hungerford JL, Damato B et al. neous melanoma: too much for too little? Clin Exp Metastasis 2010; 27: 371–387. The biology of micrometastases from uveal melanoma. J Clin Pathol 2011; 64: 141 Kim M, Gans JD, Nogueira C, Wang A, Paik JH, Feng B et al. Comparative 666–671. oncogenomics identifies NEDD9 as a melanoma metastasis gene. Cell 2006; 125: 158 van Akkooi AC, Nowecki ZI, Voit C, Schafer-Hesterberg G, Michej W, de Wilt JH 1269–1281. et al. Sentinel node tumor burden according to the Rotterdam criteria is the 142 Clark EA, Golub TR, Lander ES, Hynes RO. Genomic analysis of metastasis reveals most important prognostic factor for survival in melanoma patients: a an essential role for RhoC. Nature 2000; 406: 532–535. multicenter study in 388 patients with positive sentinel nodes. Ann Surg 2008; 143 Das SK, Bhutia SK, Kegelman TP, Peachy L, Oyesanya RA, Dasgupta S et al. 248: 949–955. MDA-9/syntenin: a positive gatekeeper of melanoma metastasis. Front Biosci 159 van der Ploeg AP, van Akkooi AC, Schmitz PI, Koljenovic S, Verhoef C, Eggermont 2012; 17: 1–15. AM. EORTC Melanoma Group sentinel node protocol identifies high rate of 144 Jarrett SG, Novak M, Harris N, Merlino G, Slominski A, Kaetzel DM. NM23 submicrometastases according to Rotterdam Criteria. Eur J Cancer 2010; 46: deficiency promotes metastasis in a UV radiation-induced mouse model of 2414–2421. human melanoma. Clin Exp Metastasis 2013; 30: 25–36. 160 van der Ploeg AP, van Akkooi AC, Rutkowski P, Nowecki ZI, Michej W, Mitra A 145 Riker AI, Samant RS. Location, location, location: The BRMS1 protein and mela- et al. Prognosis in patients with sentinel node-positive melanoma is accurately noma progression. BMC Med 2012; 10:19. defined by the combined Rotterdam tumor load and Dewar topography criteria. 146 Orgaz JL, Sanz-Moreno V. Emerging molecular targets in melanoma invasion and J Clin Oncol 2011; 29: 2206–2214. metastasis. Pigment Cell Melanoma Res 2013; 26: 39–57. 161 Kingham TP, Panageas KS, Ariyan CE, Busam KJ, Brady MS, Coit DG. Outcome of 147 Butler TP, Gullino PM. Quantitation of cell shedding into efferent blood of patients with a positive sentinel lymph node who do not undergo completion mammary . Cancer Res 1975; 35: 512–516. . Ann Surg Oncol 2010; 17: 514–520. 148 Fidler IJ. Metastasis: guantitative analysis of distribution and fate of tumor 162 Wong SL, Morton DL, Thompson JF, Gershenwald JE, Leong SP, Reintgen DS embolilabeled with 125 I-5-iodo-20-deoxyuridine. J Natl Cancer Inst 1970; 45: et al. Melanoma patients with positive sentinel nodes who did not undergo 773–782. completion lymphadenectomy: a multi-institutional study. Ann Surg Oncol 2006; 149 Ireland A, Millward M, Pearce R, Lee M, Ziman M. Genetic factors in metastatic 13: 809–816. progression of cutaneous melanoma: the future role of circulating melanoma 163 Coleman RE. Adjuvant bisphosphonates in breast cancer: are we witnessing the emergence of a new therapeutic strategy? Eur J Cancer 2009; 45: cells in prognosis and management. Clin Exp Metastasis 2011; 28: 327–336. 1909–1915. 150 De Giorgi V, Pinzani P, Salvianti F, Grazzini M, Orlando C, Lotti T et al. Circulating 164 Gnant M, Mlineritsch B, Schippinger W, Luschin-Ebengreuth G, Postlberger S, benign nevus cells detected by ISET technique: warning for melanoma Menzel C et al. Endocrine therapy plus zoledronic acid in premenopausal breast molecular diagnosis. Arch Dermatol 2010; 146: 1120–1124. cancer. N Engl J Med 2009; 360: 679–691. 151 Clawson GA, Kimchi E, Patrick SD, Xin P, Harouaka R, Zheng S et al. Circulating 165 Bos PD, Nguyen DX, Massague J. Modeling metastasis in the mouse. Curr Opin tumor cells in melanoma patients. PLoS One 2012; 7: e41052. Pharmacol 2010; 10: 571–577. 152 Kienast Y, von Baumgarten L, Fuhrmann M, Klinkert WE, Goldbrunner R, Herms J 166 Sharpless NE, Depinho RA. The mighty mouse: genetically engineered et al. Real-time imaging reveals the single steps of brain metastasis formation. mouse models in cancer drug development. Nat Rev Drug Discov 2006; 5: Nat Med 2010; 16: 116–122. 741–754. 153 Chambers AF, Groom AC, MacDonald IC. Dissemination and growth of cancer 167 Damsky Jr WE, Bosenberg M. Mouse melanoma models and cell lines. Pigment cells in metastatic sites. Nat Rev Cancer 2002; 2: 563–572. Cell Melanoma Res 2010; 23: 853–859. 154 Lee YT. Malignant melanoma: pattern of metastasis. CA Cancer J Clin 1980; 30: 168 Walker GJ, Soyer HP, Terzian T, Box NF. Modelling melanoma in mice. Pigment 137–142. Cell Melanoma Res 2011; 24: 1158–1176. 155 Patel JK, Didolkar MS, Pickren JW, Moore RH. Metastatic pattern of malignant 169 Liu W, Monahan KB, Pfefferle AD, Shimamura T, Sorrentino J, Chan KT et al. melanoma. A study of 216 autopsy cases. Am J Surg 1978; 135: 807–810. LKB1/STK11 inactivation leads to expansion of a prometastatic tumor sub- 156 Schon CA, Gorg C, Ramaswamy A, Barth PJ. Splenic metastases in a large population in melanoma. Cancer Cell 2012; 21: 751–764. unselected autopsy series. Pathol Res Pract 2006; 202: 351–356. 170 Pittet MJ, Weissleder R. Intravital imaging. Cell 2011; 147: 983–991.

Oncogene (2014) 2413 – 2422 & 2014 Macmillan Publishers Limited