You are on page 1of 15

The n e w e ng l a n d j o u r na l of m e dic i n e

review article

mechanisms of disease

Melanoma
Arlo J. Miller, M.D., Ph.D., and Martin C. Mihm, Jr., M.D.

A
lthough melanoma accounts for only 4 percent of all derma- From the Dermatopathology Unit, Massa-
tologic cancers, it is responsible for 80 percent of deaths from skin cancer; chusetts General Hospital, and Harvard
Medical School — both in Boston. Ad-
only 14 percent of patients with metastatic melanoma survive for five years.1 dress reprint requests to Dr. Mihm at the
The intractability of advanced melanoma shows how much we have to learn about Department of Dermatopathology, Massa-
the changes that facilitate the vertical growth and deep invasion of melanoma and chusetts General Hospital, 55 Fruit St.,
Warren 827, Boston, MA 02114.
about the mechanisms that block the effectiveness of chemotherapy.
The Clark model of the progression of melanoma emphasizes the stepwise trans- N Engl J Med 2006;355:51-65.
formation of melanocytes to melanoma (Fig. 1). The model depicts the proliferation Copyright © 2006 Massachusetts Medical Society.

of melanocytes in the process of forming nevi and the subsequent development of


dysplasia, hyperplasia, invasion, and metastasis.2 Numerous molecular events, many
of them revealed by genomic3 and proteomic4 methods, have been associated with
the development of melanoma. But rather than catalogue all the molecular lesions
in this tumor, we will focus on connections between molecular pathways and risk
factors for melanoma, the different steps of neoplastic transformation, and the
patterns of molecular changes in melanoma (Fig. 2).

En v ironmen ta l a nd Gene t ic In t er ac t ions

Risk Factors
The strongest risk factors for melanoma are a family history of melanoma, multiple
benign or atypical nevi, and a previous melanoma. Immunosuppression, sun sensi-
tivity, and exposure to ultraviolet radiation are additional risk factors. Each of these
risk factors corresponds to a genetic predisposition or an environmental stressor
that contributes to the genesis of melanoma. Each factor is understood to various
degrees at a molecular level. For example, 25 to 40 percent of the members of
melanoma-prone families have mutations in cyclin-dependent kinase inhibitor 2A
(CDKN2A)5 (Table 1 lists all genes mentioned in this article), and a few rare kindreds
have mutations in cyclin-dependent kinase 4 (CDK4). There is a rational basis for a
link between susceptibility to melanoma and a mutation in CDKN2A or CDK4 since
both are tumor-suppressor genes. They will be discussed later in the context of
disease progression. In addition, sensitivity to ultraviolet light is associated with a
polymorphic genetic determinant that affects susceptibility to melanoma, thereby
highlighting an important genetic–environmental interaction.

Photosensitivity, Tanning, and Melanoma


The effect of exposure to ultraviolet light is governed by variations in particular
genes (polymorphisms) that affect both the defensive response of the skin to ultra-
violet light and the risk of melanoma. Ultraviolet radiation causes genetic changes
in the skin, impairs cutaneous immune function, increases the local production of
growth factors, and induces the formation of DNA-damaging reactive oxygen species
that affect keratinocytes and melanocytes.6,7 The tanning response is a defensive
measure in which melanocytes synthesize melanin and transfer it to keratinocytes,

n engl j med 355;1 www.nejm.org july 6, 2006 51

The New England Journal of Medicine


Downloaded from nejm.org at GLASGOW UNIVERSITY LIBRARY on April 6, 2013. For personal use only. No other uses without permission.
Copyright © 2006 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

where it absorbs and dissipates ultraviolet energy.7


Figure 1 (facing page). The Clark Model (Hematoxylin
Clinically, variations in pigmentation and the tan- and Eosin).
ning response to ultraviolet light are associated Melanocytes progress through a series of steps toward
with variations in susceptibility to melanoma.8,9 malignant transformation. The frequency of both the
At the molecular level, exposure to ultraviolet progression of nevi toward becoming malignant and
light increases skin pigmentation, in part through the regression of nevi is unknown. The model empha-
sizes the histopathological changes that occur in the
the action of α-melanocyte–stimulating hormone
progression of melanoma. Normal melanocytes pro-
(α-MSH) on its receptor, the melanocortin recep- gressively develop a malignant phenotype through the
tor 1 (MC1R) (Fig. 3). Binding of the hormone to acquisition of various phenotypic features. The partic-
the receptor stimulates intracellular signaling in ular histologic features characterizing each step of pro-
melanocytes, and this signaling increases the ex- gression are the visible manifestations of underlying
genetic changes.
pression of enzymes involved in the production
of melanin. Light-skinned and redheaded people
often carry germ-line polymorphisms in the MC1R ed by the activation of oncogenic pathways is lim-
gene10,11 that reduce the activity of the receptor.12 ited.14 At a molecular level, abnormal activation
Such polymorphisms increase the risk of mel- of the mitogen-activated protein kinase (MAPK)
anoma considerably.13 In light-skinned people, signaling pathway (also called extracellular-related
therefore, the basis of increased susceptibility to kinase [ERK]) stimulates growth in melanoma
melanoma is a genetic impairment in the produc- cells (Fig. 4A).15-17 Activation of this pathway is the
tion of melanin, the main defense of melanocytes result of somatic mutations of N-RAS, which are
against ultraviolet radiation. associated with about 15 percent of melanomas,
Although the tanning response to ultraviolet or BRAF, which are associated with about 50 per-
radiation appears dose-dependent, the nature of cent of melanomas. These mutations, which occur
the exposure is also a factor. Melanoma occurs exclusively of each other, cause constitutive acti-
most frequently after intermittent exposure to the vation of the serine–threonine kinases in the
sun and in people with frequent sunburns. Epi- ERK–MAPK pathway.18-20
demiologic observations suggest that chronic or BRAF mutations occur at a similar frequency
low-grade exposures to ultraviolet light induce in benign nevi and in primary and metastatic
protection against DNA damage, whereas intense, melanomas.21 Since most nevi cease proliferation
intermittent exposures cause genetic damage.7 and remain static for decades, these similar fre-
quencies suggest that nevi must acquire addition-
al molecular lesions to free themselves of growth
A Mol e cul a r Model
of Mel a nom a Pro gr e s sion restraints and become malignant. Experiments in
model systems support this hypothesis. In zebra-
The Clark model (Fig. 1) describes the histologic fish, melanocyte-specific expression of a mutant
changes that accompany the progression from BRAF protein causes an ectopic proliferation of
normal melanocytes to malignant melanoma.2 melanocytes, analogous to human nevi.22 In these
We will relate these histologic changes to particu- fish, the combination of a BRAF mutation and
lar gene mutations (Table 1) in melanoma and dis- inactivation of the tumor-suppressor gene p53
cuss how these mutations affect molecular sig- causes melanocytes to become malignant.22 In
naling to contribute to the progression from human melanocytes, mutant BRAF protein induc-
normal melanocytes to melanoma (Fig. 2). es cell senescence by increasing the expression
of the cell-cycle inhibitor of kinase 4A (INK4A).23
Hyperplasia and Nevus Formation INK4A limits hyperplastic growth caused by a
In the Clark model, the first phenotypic change BRAF mutation. The arrest of the cell cycle caused
in melanocytes is the development of benign nevi, by INK4A can, however, be overcome by mutations
which are composed of neval melanocytes (Fig. 1). in INK4A itself, as well as other cell-cycle factors.
The control of growth in these cells is disrupted, In vitro, depletion of BRAF and N-RAS from
yet the growth of a nevus is limited — a nevus melanoma cells suppresses their growth.24-26
rarely progresses to cancer.2 The absence of pro- Small molecules that inhibit BRAF are being
gression is probably due to oncogene-induced tested clinically (BAY 43-9006) but have had only
cell senescence, in which growth that is stimulat- limited success as single agents.27 In mice, the

52 n engl j med 355;1 www.nejm.org july 6, 2006

The New England Journal of Medicine


Downloaded from nejm.org at GLASGOW UNIVERSITY LIBRARY on April 6, 2013. For personal use only. No other uses without permission.
Copyright © 2006 Massachusetts Medical Society. All rights reserved.
mechanisms of disease

n engl j med 355;1 www.nejm.org july 6, 2006 53

The New England Journal of Medicine


Downloaded from nejm.org at GLASGOW UNIVERSITY LIBRARY on April 6, 2013. For personal use only. No other uses without permission.
Copyright © 2006 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

growth of melanomas with BRAF mutations can tumors.40 The development of melanoma in such
be suppressed by the inhibition of the down- mice requires mutations in other genes, such as
stream MEK enzymes, providing a possible target an activating mutation in H-RAS, an upstream
for treatment.28 component in MAPK signaling, which triggers
MEK signaling.41 Genes that encode CDK4 and
Cytologic Atypia and Tumor-Suppressor cyclin D1 (CCND1) encode proteins that act down-
Genes stream of INK4A, and they are also mutated in
The Clark model suggests that the next step toward some melanomas. These targets of INK4A func-
melanoma is the development of cytologic atypia tion together as part of a complex that promotes
in dysplastic nevi, which may arise from preex- the progression of the cell cycle by phosphorylat-
isting benign nevi or as new lesions. The molecu- ing retinoblastoma (Rb) protein, a cell-cycle reg-
lar abnormalities at this stage of progression ulator. Rare melanoma kindreds carry germ-line
affect cell growth, DNA repair, and the suscepti- mutations in CDK4 that disrupt cell-cycle control
bility to cell death. In 25 to 40 percent of cases of by preventing the molecular interaction that allows
familial melanoma,6 a genetic defect inactivates INK4A to repress CDK4.42 Mice that carry the hu-
CDKN2A, a single gene that encodes two tumor- man CDK4 mutation are prone to melanoma when
suppressor proteins, p16INK4A and p19ARF29,30; in exposed to various carcinogens.43
25 to 50 percent of nonfamilial melanoma,31,32 a The D-type cyclin CD1 may have an oncogenic
different tumor-suppressor gene, phosphatase and role in acral melanoma, in which amplification of
tensin homologue (PTEN) (Fig. 3), is inactivated the CCND1 gene and overexpression of cyclin CD1
by mutation.33,34 In murine models of melanoma, protein occur more frequently than in melanoma
mutation of either CDKN2A or PTEN alone fails to at other sites.44 Inhibition of CCND1 (with anti-
cause melanoma, but when combined with each sense CCND1) causes apoptosis of human mela-
other or with mutations in other genes,35 mela- noma xenografts implanted in immunodeficient
nomas do arise. Mutation of CDKN2A or PTEN is mice, without an apparent effect on normal mela-
only one molecular step on the path to the devel- nocytes.
opment of melanoma, but it is unclear precisely Alternative splicing of various exons within
when such mutations occur. The increased sus- CDKN2A yields two distinct tumor-suppressor pro-
ceptibility to melanoma that is associated with teins, INK4A and alternate reading frame (ARF)
loss of the germ-line CDKN2A gene suggests that (Fig. 3).39 The ARF gene (also called p14ARF) de-
this genetic lesion increases the probability that rives its name from the use of an alternative
dysplastic nevi will become malignant or increas- reading frame of the exons it shares with INK4A.
es the rate of the development of new melanoma ARF functions as a tumor suppressor by arrest-
without a precursor. ing the cell cycle or promoting cell death after
DNA damage or when various oncogenes or loss
CDKN2A of Rb stimulate aberrant cell proliferation. ARF
The G1–S checkpoint that governs the commitment participates in the core regulatory process that
of a cell to DNA replication during the S phase controls levels of the p53 protein. It acts through
(synthesis of DNA) is a site where many pathways the mouse double minute 2 (MDM2) protein,
that control cell division converge36,37 (Fig. 4B). which triggers the ubiquination of p53, thereby
In some familial and sporadic cases of melano- instigating its destruction in the proteosome.
ma,36,37 the CDKN2A locus is lost by homozygous ARF binds to MDM2, sequestering it from p53
deletion of a portion of chromosome 9.36-38 One and in this way causes p53 to accumulate; p53
of the genes in this locus encodes INK4A, then arrests the cell cycle at the G2–M site, allow-
(p16INK4A), a protein that blocks the cell cycle at ing for repair of damaged DNA or the induction
the G1–S checkpoint by inhibiting cyclin-depen- of apoptosis.45,46 In cells, ARF deficiency abro-
dent kinases. INK4A (an inhibitor of CDK4) sup- gates oncogene-induced senescence and increases
presses the proliferation of cells with damaged susceptibility to transformation.47 In vitro, im-
DNA or activated oncogenes and also acts when mortalization of cells often occurs with the loss
cells are old or crowded.39 Mice lacking INK4A of either ARF or p53.48 In animals, ARF deficiency
appear normal but are abnormally sensitive to shortens the time required for the development
carcinogens and prone to the development of of melanoma after exposure to ultraviolet light;

54 n engl j med 355;1 www.nejm.org july 6, 2006

The New England Journal of Medicine


Downloaded from nejm.org at GLASGOW UNIVERSITY LIBRARY on April 6, 2013. For personal use only. No other uses without permission.
Copyright © 2006 Massachusetts Medical Society. All rights reserved.
mechanisms of disease

Figure 2. Biologic Events and Molecular Changes in the Progression of Melanoma.


At the stage of the benign nevus, BRAF mutation and activation of the mitogen-activated protein kinase (MAPK) pathway occur. The cy-
tologic atypia in dysplastic nevi reflect lesions within the cyclin-dependent kinase inhibitor 2A (CDKN2A) and phosphatase and tensin
homologue (PTEN) pathways. Further progression of melanoma is associated with decreased differentiation and the decreased expres-
sion of melanoma markers regulated by microphthalmia-associated transcription factor (MITF). The vertical-growth phase and meta-
static melanoma are notable for striking changes in the control of cell adhesion. Changes in the expression of the melanocyte-specific
gene melastatin 1 (TRPM1) correlate with metastatic propensity, but the function of this gene remains unknown. Other changes include
the loss of E-cadherin and increased expression of N-cadherin, αVβ3 integrin, and matrix metalloproteinase 2 (MMP-2).

when both gene products of CDKN2A (INK4A and some 10.33,34,50 PTEN encodes a phosphatase that
attenuates signaling by a variety of growth factors
ARF) are deficient, the latent period is even short-
that use phosphatidylinositol phosphate (PIP3) as
er.49 These data suggest how ARF facilitates the
progression of melanoma and indicate that the an intracellular signal. In the presence of such
low frequency of p53 mutations in melanoma is growth factors, intracellular levels of PIP3 rapidly
increase. This increase triggers the activation of
partly related to loss of ARF, which renders the
p53 pathway inactive.39 protein kinase B (PKB, also called AKT) by phos-
phorylation (Fig. 3). Activated AKT phosphory-
PTEN, AKT, and Cell Death lates and inactivates proteins that suppress the
A second chromosomal region that is frequently cell cycle or stimulate apoptosis, thereby facilitat-
affected by homozygous deletion in melanoma ing the proliferation and survival of cells. PTEN
and other cancers is the PTEN locus on chromo- normally keeps PIP3 levels low; in its absence,

n engl j med 355;1 www.nejm.org july 6, 2006 55

The New England Journal of Medicine


Downloaded from nejm.org at GLASGOW UNIVERSITY LIBRARY on April 6, 2013. For personal use only. No other uses without permission.
Copyright © 2006 Massachusetts Medical Society. All rights reserved.
56
Table 1. Important Genes in Melanoma.

Pathway Gene or Protein* Function Changes in Melanoma


RAS and MAPK
N-RAS Oncogene Sporadic activating mutation at G13R
BRAF Oncogene Sporadic activating mutation at codon V600E in
nevi and melanoma
Mitogen-activated protein kinase–extracellular- Signal transduction Up-regulated in radial-growth and vertical-growth
related kinase (MEK) phases
Extracellular-related kinase 1 or 2 (ERK1 or ERK2) Signal transduction Increased activity
or mitogen-activated protein kinase (MAPK)
INK4A, CDK, and Rb
The

Cyclin-dependent kinase inhibitor 2A or inhibitor Tumor suppressor–negative regulator of cell prolif- Germ-line mutations in some familial melanomas;
of kinase 4A (CDKN2A or INK4A) eration sporadic deletions, promoter inactivation, loss
of heterozygosity in many melanomas
Cyclin-dependent kinase 4 (CDK4) Promoter of cell proliferation Protein insensitive to inhibition by INK4A due to
rare familial germ-line mutations at R24C
Cyclin D1 (CCND1) Promoter of cell proliferation Sporadic amplification in acral melanoma

n engl j med 355;1


Retinoblastoma (Rb) Tumor suppressor–negative regulator of cell prolif- Phosphorylation leads to progression from
eration G1 to S
ARF and p53
Alternate reading frame (ARF) Tumor suppressor, degrades MDM2 Germ-line mutations in some familial melanomas;

www.nejm.org
n e w e ng l a n d j o u r na l

sporadic deletions, promoter inactivation, in

The New England Journal of Medicine


of

many melanomas
Tumor protein 53 (p53) Tumor suppressor that induces apoptosis and sup- Expression usually present in melanoma
pressed proliferation after DNA damage

july 6, 2006
Mouse double minute 2 (MDM2) Targeter of p53 for ubiquination and destruction Up-regulated in presence of ARF mutation

Copyright © 2006 Massachusetts Medical Society. All rights reserved.


BCL-2–associated X protein (BAX) Inducer of cell death Variable but usually down-regulated
m e dic i n e

PTEN and AKT


Phosphatase and tensin homologue (PTEN) Tumor suppressor, represses PI3K Sporadic deletion of chromosomal region
Phosphatidylinositol 3 kinase (PI3K) Signaling molecule for many growth factors Active in presence of PTEN mutation
Protein kinase B (AKT or PKB) Oncogene that is activated by PI3K, leading to Amplified in some melanomas
increased cell survival
BCL-2 antagonist of cell death (BAD) Inducer of cell death Variable but often down-regulated
Forkhead receptor (FKHR) Growth supression Activated in response to PI3 pathway

Downloaded from nejm.org at GLASGOW UNIVERSITY LIBRARY on April 6, 2013. For personal use only. No other uses without permission.
MSH and MITF
Pro-opiomelanocortin or α-melanocyte–stimulating Signaling molecule important in pigmentation Increased melanoma vertical-growth phase
hormone (POMC or α-MSH)
Melanocortin receptor 1 (MC1R) Receptor for α-MSH Polymorphic gene affecting hair and skin color
and response to ultraviolet radiation
Adenylate cyclase (AC) Producer of cyclic AMP Up-regulated
cAMP response element–binding protein (CREB) Transcription factor Up-regulated; affects MITF and melanocyte
differentiation
Microphthalmia-associated transcription factor Transcription factor Sporadic amplification of chromosomal region
(MITF)
Tyrosinase (TYR) Pigment synthesis Decreased expression
Tyrosinase-related protein 1 (TYRP1) Pigment synthesis Decreased expression
Dopachrome tautomerase (DCT) Pigment synthesis Decreased expression
Melan-A (MLANA) Antigen recognized by melan-A and melanoma anti- Decreased expression

n engl j med 355;1


gen recognized by T-cells 1 (MART1) antibodies
Silver homologue (SILV) Antigen recognized by HMB-45 antibody Decreased expression

Melastatin 1 (TRPM1) Unknown Decreased expression in metastatic melanoma


BCL-2 Cell survival Variable up-regulation in various phases of

www.nejm.org
melanoma
mechanisms of disease

Cell adhesion
Wingless-type mammary tumor virus integration- Protooncogene, secreted growth factor that inacti- Pathway up-regulated
site family (WNT) vates

The New England Journal of Medicine


july 6, 2006
GSK3-B
Glycogen synthase kinase 3β (GSK3β) Serine–threonine kinase that targets β-catenin for Variable; affected by WNT pathway
degradation
β-Catenin Adherens junction protein, transcriptional co- Sporadic stabilizing mutations
activator

Copyright © 2006 Massachusetts Medical Society. All rights reserved.


T-cell factor–lymphoid-enhancing factor (TCF–LEF) Transcription factor Up-regulated
E-cadherin Cell-adhesion molecule Decreased expression in vertical-growth phase
N-cadherin Cell-adhesion molecule Aberrant expression in vertical-growth phase
αVβ 3integrin Dimer that forms cell-adhesion molecule Aberrant expression in vertical-growth phase

* Abbreviated forms of the gene are given in parentheses.

Downloaded from nejm.org at GLASGOW UNIVERSITY LIBRARY on April 6, 2013. For personal use only. No other uses without permission.
57
The n e w e ng l a n d j o u r na l of m e dic i n e

Figure 3. Microphthalmia-Associated Transcription Factor (MITF) and β-Catenin Pathways.


In the MITF pathway, MITF is regulated at both transcriptional and post-translational levels. The post-translational activation can occur
through the ERK component of the MAPK pathway. The chief transcriptional pathways that are activated by extracellular signals are the
melanocortin and WNT pathways. The melanocortin pathway regulates pigmentation through the MC1R. MC1R activates the cyclic AMP
(cAMP) response-element binding protein (CREB). Increased expression of MITF and its activation by phosphorylation (P) stimulate the
transcription of tyrosinase (TYR), tyrosinase-related protein 1 (TYRP1), and dopachrome tautomerase (DCT), which produce melanin;
melan-A, silver homologue, and melastatin 1 (TRPM1) are melanoma markers; inhibitor of kinase 4A (INK4A) leads to cell-cycle arrest,
and BCL-2 suppresses apoptosis. In the β-catenin pathway, β-catenin plays a central role in cell adhesion and cell signaling. Signals from
WNT ligands block the breakdown of β-catenin. When WNT proteins bind the G-protein–coupled receptor (called frizzled), they inacti-
vate the kinase GSK3β, an enzyme that phosphorylates β-catenin and targets it for destruction in the proteosome. Then β-catenin accu-
mulates in the cytoplasm and translocates to the nucleus, where it binds to LEF–TCF transcription factors and increases the expression
of several genes, including MITF, the cell-cycle mediator cyclin D1 (CCND1), and matrix metalloproteinase 7 (MMP-7).

levels of PIP3 and active (phosphorylated) AKT in- also be increased in cells by mutations that cause
crease. Increased AKT activity prolongs cell sur- the amplification and overexpression of the pro-
vival through the inactivation of BCL-2 antago- tein. Restoration of PTEN in cultured mouse me-
nist of cell death (BAD) protein and increases lanocytes decreases the ability of the cells to form
cell proliferation by increasing CCND1 expres- tumors.52 In model systems, suppression of AKT3,
sion, and affects many other cell-survival and cell- a member of the AKT family, reduces the survival
cycle genes through the activation of the forkhead of melanoma cells and the growth of human mela-
(FKHR) transcription factor.32,51 AKT activity can nomas implanted in immunodeficient nude mice.53

58 n engl j med 355;1 www.nejm.org july 6, 2006

The New England Journal of Medicine


Downloaded from nejm.org at GLASGOW UNIVERSITY LIBRARY on April 6, 2013. For personal use only. No other uses without permission.
Copyright © 2006 Massachusetts Medical Society. All rights reserved.
mechanisms of disease

As compared with normal melanocytes, increased MITF in Melanoma


levels of the active form of AKT were found in the Decreased or absent pigmentation and decreased
radial-growth phase.53 or absent expression of SILV and MLANA accom-
pany the progression from nevus to melanoma.
MI TF a nd Mel a no c y te Tumors that are deficient in these proteins have
Differ en t i at ion a poor prognosis.65-68 Expression of the mela-
statin 1 (TRPM1) gene, whose function is unknown,
Clark proposed that many nevi regress through is also controlled by MITF.69 Melanomas that are
differentiation and that the failure of differentia- deficient in melastatin have a poor prognosis.70
tion is necessary for dysplasia.2 The normal pro- The mechanism of decreased expression of these
cess of melanocyte differentiation requires exit genes is a puzzle because MITF is present in nearly
from the cell cycle and the expression of genes all melanomas.71-73
that encode proteins necessary for the produc- Although MITF causes differentiation and cell-
tion of pigment — two processes that are de- cycle arrest in normal melanocytes, melanoma
regulated in melanoma. The microphthalmia- cells do not have these characteristics. Recently,
associated transcription factor (MITF) regulates a large-scale search for genomic changes in mela-
the development and differentiation of melano- noma with the use of high-density single-nucleo-
cytes54 and maintains melanocyte progenitor cells tide polymorphisms (SNPs) found an increased
in adults.55,56 copy number (4 to 119 copies per cell) of a region
of chromosome 3 that includes the MITF locus.74
MITF in Development This increase was accompanied by the increased
Mice lacking functional MITF are albino because expression of MITF protein. The overexpression
they lack melanocytes, whereas those with par- of both MITF and BRAF could transform primary
tial MITF function have premature graying owing cultures of human melanocytes, implicating MITF
to the death of melanocytes. These experiments as an oncogene. Notably, MITF amplification oc-
show that MITF is important in the differentiation curs most frequently in tumors that have a poor
and maintenance of melanocytes.57,58 MITF ap- prognosis and is associated with resistance to
pears to contribute to melanocyte survival by in- chemotherapy.74 Interference with MITF function
creasing the expression of the BCL-2 gene, a key increased the chemosensitivity of a melanoma
antiapoptotic factor.59 In mice, deficiencies of both cell line, making MITF a potential target for
MITF and BCL-2 cause gray hair due to a loss of treatment.
differentiated melanocytes. The loss of melano-
cytes is due to the apoptosis of melanocyte pro- Cel l A dhe sion a nd In va sion
genitor cells in the hair follicle.55 In melanoma
cell lines, a reduction in BCL-2 protein also causes Local invasion and metastatic spread are respon-
cell death, suggesting that the survival of malig- sible for the morbidity and mortality in melano-
nant melanocytes depends on BCL-2.60 ma. In the Clark model, invasive characteristics
appear in the vertical-growth phase, when mela-
MITF in Differentiation noma cells not only penetrate the basement mem-
MITF functions in a key pathway leading to me- brane but also grow intradermally as an expand-
lanocyte pigmentation (Fig. 3). Intracellular sig- ing nodule (Fig. 2). Metastatic melanoma develops
naling induced by α-MSH acting on MC1R in- when tumor cells dissociate from the primary
creases MITF expression, which in turn increases lesion, migrate through the surrounding stroma,
the transcription of genes underlying melanin and invade blood vessels and lymphatics to form
synthesis: tyrosinase, tyrosinase-related-protein 1, a tumor at a distant site.75 Clinically, the absolute
and dopachrome tautomerase.61 MITF also regu- depth of local invasion, measured directly by histo-
lates the transcription of the melanocyte-specific pathologic analysis (the Breslow index), is the
genes silver homologue (SILV)62,63 and melan-A principal prognostic factor and primary criterion
(MLANA),62 whose immunohistochemical detec- in melanoma staging.76 Invasion and spread of
tion points to the diagnosis of melanoma. In addi- melanoma are related to alterations in cell adhe-
tion, MITF causes cell-cycle arrest by the induction sion. Normally, cell adhesion controls cell migra-
of INK4A.64 tion, tissue organization, and organogenesis,77

n engl j med 355;1 www.nejm.org july 6, 2006 59

The New England Journal of Medicine


Downloaded from nejm.org at GLASGOW UNIVERSITY LIBRARY on April 6, 2013. For personal use only. No other uses without permission.
Copyright © 2006 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

but disturbances in cell adhesion contribute to


Figure 4 (facing page). MAPK and PTEN Pathways and the CDKN2A Tumor-
tumor invasion, tumor–stroma interactions, and Suppressor Locus.
tumor-cell signaling. Panel A shows the pathway associated with N-RAS, BRAF, and mitogen-
activated protein kinase (MAPK). MAPKs are involved in signaling from
Cadherins numerous growth factors and cell-surface receptors. There are many vari-
Cadherins are multifunctional transmembrane ations in the components of particular cascades from various cell-surface
receptors. Typically, adapter proteins (not shown) link the growth-factor
proteins that sustain cell-to-cell contacts, form
receptor to RAS proteins, including N-RAS. When activated, RAS proteins
connections with the actin cytoskeleton, and in- phosphorylate (P) the mitogen-activated protein kinase (MEK) kinases,
fluence intracellular signaling. The extracellular which then act on extracellular-related kinase (ERK) kinases. ERK kinases
domain of cadherins binds to like cadherins on phosphorylate many targets in the cytoplasm and interact with other path-
other cells in regions of cell contacts called adhe- ways, including phosphatidylinositol 3 kinase (PI3K) and MITF. ERK kinases
translocate to the nucleus, where they activate transcription factors that
rens junctions. Cadherins are divided into three
promote cell-cycle progression and proliferation by increasing the transcrip-
subtypes: E (epithelial), present in polarized epi- tion of many genes, including CD1. In survival signaling associated with
thelial cells in the epidermis, including melano- phosphatase and tensin homologue (PTEN) and AKT, also known as pro-
cytes and keratinocytes; P (placental); and N (neu- tein kinase B, PTEN inhibits growth-factor signaling by inactivating phos-
ral), found in mesenchymal cells in the dermis. phatidylinositol triphosphate (PIP3) generated by PI3K. A variety of growth
factors (PDGF, NGF, and IGF-1) bind to their respective receptor tyrosine
The intracellular domain is associated with a large
kinases and activate PI3K. The activated molecule converts the plasma
protein complex that includes β-catenin and forms membrane lipid phosphatidylinositol 4,5-bisphosphonate to PIP3 . PIP3 acts
structural links with bundles of actin filaments. as a second messenger, leading to the phosphorylation and activation of
Several signaling pathways cause β-catenin to AKT. AKT is itself a kinase that phosphorylates protein substrates that af-
dissociate from the cell adhesion complex and fect the cell cycle, growth, and survival. Often, these AKT targets are inacti-
vated by phosphorylation. PTEN attenuates this pathway through dephos-
transduce signals to the nucleus (Fig. 3). One of
phorylation and inactivation of PIP3 , suppressing signaling from growth
these pathways is called the wingless-type mam- factors by blocking the activation of AKT. In Panel B, CDKN2A encodes two
mary tumor virus integration-site family (WNT) distinct tumor-suppressor genes; separate first exons that are spliced into
pathway. WNTs are secreted proteins with impor- alternate reading frames (ARF) of the second and third exons permit the
tant functions in development, especially in neu- expression of two different proteins from the same genetic locus. The gene
has 4 exons. Transcription of messenger RNA (mRNA) can be initiated at
ral crest cells like melanocytes. When WNT pro-
either E1B or E1A, and the initiation site determines which gene the locus
teins bind their receptors, they inactivate the will express. RNA that is transcribed from either exon is spliced with the re-
kinase GSK3β, an enzyme that phosphorylates maining two exons, E2 and E3, to produce mRNA for either INK4A or ARF.
β-catenin and targets it for destruction in the pro- However, ARF uses a different reading frame of the exon 2 and 3 codons.
teosome.78,79 Tyrosine phosphorylation of β-catenin In the cell-cycle progression involving INK4A, ARF, and retinoblastoma pro-
tein (Rb), a family of cyclins and cyclin-dependent kinases (CDKs) regulate
disrupts the association between E-cadherin and
progression through the cell cycle, and a family of CDK inhibitors opposes
β-catenin,80 allowing β-catenin to translocate to this action. In particular, the two phases of the G1–S checkpoint are gov-
the nucleus, where it binds to lymphoid enhancer erned primarily by cyclin D associated with cyclin-dependent kinases 4 and
factor–T-cell factor (LEF–TCF). Mutations in the 6 (CDK4 and CDK6) at its early phase and cyclin A or E associated with
β-catenin gene can stabilize the β-catenin pro- CDK2 at the later restriction phase. INK4A encodes a cyclin-dependent ki-
nase inhibitor that inhibits CDK4 and CDK6. Ordinarily, these two kinases
tein81 or increase its nuclear localization.82-84 In-
associate with D-type cyclins and drive the cell cycle by phosphorylating
creased levels of nuclear β-catenin increase the Rb, releasing it from its inhibitory interaction with the E2F transcription
expression of MITF85 and CCND1,86 and these in factor, thereby allowing the expression of E2F-related genes and progres-
turn increase the survival and proliferation of sion from G1 to S. The absence of INK4A leads to unopposed CDK4 or
melanoma cells. Alterations in cadherin expres- 6 activity and increased cell-cycle activity. In response to DNA damage,
mouse double minute 2 (MDM2) protein binds to the transcriptional acti-
sion affect the interaction of melanoma cells with
vation domain of protein 53 (p53), blocking p53-mediated gene regulation
the environment and alter β-catenin signaling. while simultaneously leading to p53 ubiquination, nuclear export, and pro-
E-cadherin expression occurs in melanocytes and teosomal degradation. ARF opposes this action by sequestering MDM2.
keratinocytes in the epidermis and causes mela- This disruption of the MDM2–p53 interaction stabilizes p53 and increases
nocytes to associate with keratinocytes.87 In turn, p53 activity. Depending on other events, p53 either activates DNA repair
and cell-cycle arrest or causes apoptosis and the formation of BCL-2–asso-
contacts with undifferentiated keratinocytes from
ciated X protein (BAX). In the absence of ARF, p53 levels are decreased and
the basal-cell layer inhibit melanocyte prolif- the response to DNA damage is blunted.
eration, suppress the expression of melanoma

60 n engl j med 355;1 www.nejm.org july 6, 2006

The New England Journal of Medicine


Downloaded from nejm.org at GLASGOW UNIVERSITY LIBRARY on April 6, 2013. For personal use only. No other uses without permission.
Copyright © 2006 Massachusetts Medical Society. All rights reserved.
mechanisms of disease

n engl j med 355;1 www.nejm.org july 6, 2006 61

The New England Journal of Medicine


Downloaded from nejm.org at GLASGOW UNIVERSITY LIBRARY on April 6, 2013. For personal use only. No other uses without permission.
Copyright © 2006 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

markers, and cause melanocytes to become den- chronically exposed (head and neck) or intermit-
dritic.88 tently exposed (chest and back) and in acral and
Progression from the radial-growth phase to mucosal skin. For example, CCND1 amplification
the vertical-growth phase of melanoma is marked occurs predominantly in acral regions,44 whereas
by the loss of E-cadherin and the expression of activating mutations in BRAF occur most frequent-
N-cadherin89-91 (Fig. 2). N-cadherin is a character- ly in skin sites of intermittent sun exposure.106
istic of invasive carcinomas and enables metastatic
spread by permitting melanoma cells to interact Model ing Mel a nom a
with other N-cadherin–expressing cells, such as Pro gr e s sion
dermal fibroblasts and the vascular endothelium.87
Besides these changes in cell adhesion, decreased For many of the molecular lesions we have de-
E-cadherin expression92 and aberrant N-cadherin scribed, animal models have provided validation.
expression increase the survival of melanoma A surprising new model is the zebrafish, in which
cells by stimulating β-catenin signaling.93,94 premalignant and malignant lesions can be cre-
ated by the expression of mutant BRAF with or
Integrins without p53 mutation.22 This model is the only
The integrins mediate cell contacts with fibro- currently tractable system in which genetic
nectin, collagens, and laminin, components of the screens can be performed for modifiers of mela-
extracellular matrix.95 Transition from radial to noma.
vertical growth of melanoma is associated with Human melanomas that are grafted onto or
the expression of αVβ3 integrin.96 This integrin injected into nude mice allow measures of the
induces expression of matrix metalloproteinase 2, tumors’ metastatic potential and have allowed for
an enzyme that degrades the collagen in basement the testing of therapeutic interventions. Genetic
membrane.97-99 In addition, αVβ3 integrin increas- manipulation of mice has validated the contribu-
es expression of the prosurvival gene BCL-2100 and tion of many genetic alterations in melanoma, but
stimulates the motility of melanoma cells through there are fundamental differences between mouse
the reorganization of melanoma cytoskeleton.101 and human skin. Mouse melanocytes occur in
These observations form a rationale for the de- hair follicles and the dermis, rather than in the
velopment of integrin antagonists to treat mela- epidermis, as in humans. To circumvent this
noma.102 problem, human melanocytes can be altered in
cell culture and combined with keratinocytes to
Patterns of Genetic Alteration produce graft material. Using this system, the
The genetic changes in melanoma can be seen as inactivation of p53 and the simultaneous intro-
particular combinations of molecular lesions that duction of activated N-RAS, CDK4, and telomer-
interrupt a precise set of pathways, each with a ase led to darkly pigmented grafts that became
crucial role in the development of melanoma. grossly ulcerated and displayed histologic features
The MEK pathway can be activated by a mutation of melanoma, including vertical invasion.107 This
in either NRAS or BRAF, and an NRAS mutation experimental system provides a novel model to
can activate both the MEK and PTEN pathways. test invasion and metastases of transformed hu-
Similarly, INK4A, CDK4, and CCND1 function in man melanocytes in a host organism.
a unique pathway that affects the cell cycle; a mu- Supported by a grant (MCM202534) from the Cancer Research
Institute of New York and a grant (T32-GM07753, to Dr. Miller)
tation of INK4A has similar consequences as a from the National Institute of General Medical Science. No other
mutation of CCND1 or CDK4.103-105 potential conflict of interest relevant to this article was reported.
There are particular genetic changes in mela- We are indebted to Drs. David E. Fisher, Adriano Piris, Jenni-
fer Y. Lin, and Jennifer C. Broder for their critical reading of the
nomas in different sites, consistent differences manuscript, and to Dr. Claudio Clemente for contributing im-
related to ultraviolet exposure on sites that are ages for Figure 1.

References
1. Cancer facts & figures, 2003. Atlanta: sor lesions of superficial spreading and in melanoma. N Engl J Med 2005;353:
American Cancer Society, 2003. nodular melanoma. Hum Pathol 1984;15: 2135-47.
2. Clark WH Jr, Elder DE, Guerry D IV, 1147-65. 4. Alonso SR, Ortiz P, Pollan M, et al.
Epstein MN, Greene MH, Van Horn M. 3. Curtin JA, Fridlyand J, Kageshita T, Progression in cutaneous malignant mel-
A study of tumor progression: the precur- et al. Distinct sets of genetic alterations anoma is associated with distinct expres-

62 n engl j med 355;1 www.nejm.org july 6, 2006

The New England Journal of Medicine


Downloaded from nejm.org at GLASGOW UNIVERSITY LIBRARY on April 6, 2013. For personal use only. No other uses without permission.
Copyright © 2006 Massachusetts Medical Society. All rights reserved.
mechanisms of disease

sion profiles: a tissue microarray-based 20. Omholt K, Platz A, Kanter L, Ring- 36. Kamb A, Shattuck-Eidens D, Eeles R,
study. Am J Pathol 2004;164:193-203. borg U, Hansson J. NRAS and BRAF et al. Analysis of the p16 gene (CDKN2) as
5. Aitken J, Welch J, Duffy D, et al. CD- mutations arise early during melanoma a candidate for the chromosome 9p mela-
KN2A variants in a population-based sam- pathogenesis and are preserved through- noma susceptibility locus. Nat Genet 1994;
ple of Queensland families with melano- out tumor progression. Clin Cancer Res 8:23-6.
ma. J Natl Cancer Inst 1999;91:446-52. 2003;9:6483-8. 37. Hussussian CJ, Struewing JP, Gold-
6. Thompson JF, Scolyer RA, Kefford RF. 21. Pollock PM, Harper UL, Hansen KS, stein AM, et al. Germline p16 mutations
Cutaneous melanoma. Lancet 2005;365: et al. High frequency of BRAF mutations in familial melanoma. Nat Genet 1994;8:
687-701. in nevi. Nat Genet 2003;33:19-20. 15-21.
7. Gilchrest BA, Eller MS, Geller AC, 22. Patton EE, Widlund HR, Kutok JL, et 38. Pollock PM, Trent JM. The genetics of
Yaar M. The pathogenesis of melanoma al. BRAF mutations are sufficient to pro- cutaneous melanoma. Clin Lab Med 2000;
induced by ultraviolet radiation. N Engl J mote nevi formation and cooperate with 20:667-90.
Med 1999;340:1341-8. p53 in the genesis of melanoma. Curr Biol 39. Sharpless E, Chin L. The INK4a/ARF
8. MacKie RM. Risk factors for the de- 2005;15:249-54. locus and melanoma. Oncogene 2003;22:
velopment of primary cutaneous malig- 23. Michaloglou C, Vredeveld LC, Soen- 3092-8.
nant melanoma. Dermatol Clin 2002;20: gas MS, et al. BRAFE600-associated se- 40. Serrano M, Lee H, Chin L, Cordon-
597-600. nescence-like cell cycle arrest of human Cardo C, Beach D, DePinho RA. Role of
9. Marks R. Epidemiology of melanoma. naevi. Nature 2005;436:720-4. the INK4a locus in tumor suppression
Clin Exp Dermatol 2000;25:459-63. 24. Eskandarpour M, Kiaii S, Zhu C, Cas- and cell mortality. Cell 1996;85:27-37.
10. Naysmith L, Waterston K, Ha T, et al. tro J, Sakko AJ, Hansson J. Suppression of 41. Chin L, Pomerantz J, Polsky D, et al.
Quantitative measures of the effect of the oncogenic NRAS by RNA interference in- Cooperative effects of INK4a and ras in
melanocortin 1 receptor on human pig- duces apoptosis of human melanoma melanoma susceptibility in vivo. Genes
mentary status. J Invest Dermatol 2004; cells. Int J Cancer 2005;115:65-73. Dev 1997;11:2822-34.
122:423-8. 25. Hingorani SR, Jacobetz MA, Robert- 42. Zuo L, Weger J, Yang Q, et al. Germ-
11. Valverde P, Healy E, Jackson I, Rees JL, son GP, Herlyn M, Tuveson DA. Suppres- line mutations in the p16INK4a binding
Thody AJ. Variants of the melanocyte- sion of BRAF(V599E) in human melano- domain of CDK4 in familial melanoma.
stimulating hormone receptor gene are ma abrogates transformation. Cancer Res Nat Genet 1996;12:97-9.
associated with red hair and fair skin in 2003;63:5198-202. 43. Sotillo R, Garcia JF, Ortega S, et al.
humans. Nat Genet 1995;11:328-30. 26. Hoeflich KP, Gray DC, Eby MT, et al. Invasive melanoma in Cdk4-targeted mice.
12. Frandberg PA, Doufexis M, Kapas S, Oncogenic BRAF is required for tumor Proc Natl Acad Sci U S A 2001;98:13312-
Chhajlani V. Human pigmentation pheno- growth and maintenance in melanoma 7.
type: a point mutation generates non- models. Cancer Res 2006;66:999-1006. 44. Sauter ER, Yeo UC, von Stemm A, et
functional MSH receptor. Biochem Biophys 27. Lyons JF, Wilhelm S, Hibner B, Bollag al. Cyclin D1 is a candidate oncogene in
Res Commun 1998;245:490-2. G. Discovery of a novel Raf kinase inhibi- cutaneous melanoma. Cancer Res 2002;
13. Kennedy C, ter Huurne J, Berkhout M, tor. Endocr Relat Cancer 2001;8:219-25. 62:3200-6.
et al. Melanocortin 1 receptor (MC1R) 28. Solit DB, Garraway LA, Pratilas CA, et 45. Pomerantz J, Schreiber-Agus N, Liege-
gene variants are associated with an in- al. BRAF mutation predicts sensitivity to ois NJ, et al. The Ink4a tumor suppressor
creased risk for cutaneous melanoma MEK inhibition. Nature 2006;439:358-62. gene product, p19Arf, interacts with MDM2
which is largely independent of skin type 29. Kamb A, Gruis NA, Weaver-Feldhaus J, and neutralizes MDM2’s inhibition of
and hair color. J Invest Dermatol 2001; et al. A cell cycle regulator potentially in- p53. Cell 1998;92:713-23.
117:294-300. volved in genesis of many tumor types. 46. Harris SL, Levine AJ. The p53 path-
14. Braig M, Schmitt CA. Oncogene- Science 1994;264:436-40. way: positive and negative feedback loops.
induced senescence: putting the brakes on 30. Nobori T, Miura K, Wu DJ, Lois A, Oncogene 2005;24:2899-908.
tumor development. Cancer Res 2006;66: Takabayashi K, Carson DA. Deletions of 47. Sharpless NE, Ramsey MR, Balasub-
2881-4. the cyclin-dependent kinase-4 inhibitor ramanian P, Castrillon DH, DePinho RA.
15. Welsh CF, Roovers K, Villanueva J, Liu gene in multiple human cancers. Nature The differential impact of p16(INK4a) or
Y, Schwartz MA, Assoian RK. Timing of 1994;368:753-6. p19(ARF) deficiency on cell growth and
cyclin D1 expression within G1 phase is 31. Flores JF, Walker GJ, Glendening JM, tumorigenesis. Oncogene 2004;23:379-85.
controlled by Rho. Nat Cell Biol 2001;3: et al. Loss of the p16INK4a and p15INK4b 48. Kamijo T, Zindy F, Roussel MF, et al.
950-7. genes, as well as neighboring 9p21 mark- Tumor suppression at the mouse INK4a
16. Brunet A, Roux D, Lenormand P, ers, in sporadic melanoma. Cancer Res locus mediated by the alternative reading
Dowd S, Keyse S, Pouyssegur J. Nuclear 1996;56:5023-32. frame product p19ARF. Cell 1997;91:649-
translocation of p42/p44 mitogen-activat- 32. Wu H, Goel V, Haluska FG. PTEN sig- 59.
ed protein kinase is required for growth naling pathways in melanoma. Oncogene 49. Recio JA, Noonan FP, Takayama H, et
factor-induced gene expression and cell 2003;22:3113-22. al. Ink4a/arf deficiency promotes ultravi-
cycle entry. EMBO J 1999;18:664-74. 33. Li J, Yen C, Liaw D, et al. PTEN, a pu- olet radiation-induced melanomagenesis.
17. Lin AW, Barradas M, Stone JC, van tative protein tyrosine phosphatase gene Cancer Res 2002;62:6724-30.
Aelst L, Serrano M, Lowe SW. Premature mutated in human brain, breast, and pros- 50. Guldberg P, Thor Straten P, Birck A,
senescence involving p53 and p16 is acti- tate cancer. Science 1997;275:1943-7. Ahrenkiel V, Kirkin AF, Zeuthen J. Disrup-
vated in response to constitutive MEK/ 34. Steck PA, Pershouse MA, Jasser SA, et tion of the MMAC1/PTEN gene by dele-
MAPK mitogenic signaling. Genes Dev al. Identification of a candidate tumour tion or mutation is a frequent event in
1998;12:3008-19. suppressor gene, MMAC1, at chromosome malignant melanoma. Cancer Res 1997;57:
18. Albino AP, Nanus DM, Mentle IR, et al. 10q23.3 that is mutated in multiple ad- 3660-3.
Analysis of ras oncogenes in malignant vanced cancers. Nat Genet 1997;15:356- 51. Cantley LC, Neel BG. New insights
melanoma and precursor lesions: correla- 62. into tumor suppression: PTEN suppresses
tion of point mutations with differentia- 35. You MJ, Castrillon DH, Bastian BC, et tumor formation by restraining the phos-
tion phenotype. Oncogene 1989;4:1363-74. al. Genetic analysis of Pten and Ink4a/Arf phoinositide 3-kinase/AKT pathway. Proc
19. Davies H, Bignell GR, Cox C, et al. interactions in the suppression of tumori- Natl Acad Sci U S A 1999;96:4240-5.
Mutations of the BRAF gene in human genesis in mice. Proc Natl Acad Sci U S A 52. Stahl JM, Cheung M, Sharma A, Trive-
cancer. Nature 2002;417:949-54. 2002;99:1455-60. di NR, Shanmugam S, Robertson GP.

n engl j med 355;1 www.nejm.org july 6, 2006 63

The New England Journal of Medicine


Downloaded from nejm.org at GLASGOW UNIVERSITY LIBRARY on April 6, 2013. For personal use only. No other uses without permission.
Copyright © 2006 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

Loss of PTEN promotes tumor develop- ness cutaneous malignant melanoma. Can- 80. Brembeck FH, Schwarz-Romond T,
ment in malignant melanoma. Cancer cer Res 2000;60:5012-6. Bakkers J, Wilhelm S, Hammerschmidt
Res 2003;63:2881-90. 67. Seiter S, Monsurro V, Nielsen MB, et al. M, Birchmeier W. Essential role of BCL9-2
53. Stahl JM, Sharma A, Cheung M, et al. Frequency of MART-1/MelanA and gp100/ in the switch between beta-catenin’s ad-
Deregulated Akt3 activity promotes de- PMel17-specific T cells in tumor metasta- hesive and transcriptional functions. Genes
velopment of malignant melanoma. Can- ses and cultured tumor-infiltrating lym- Dev 2004;18:2225-30.
cer Res 2004;64:7002-10. phocytes. J Immunother 2002;25:252-63. 81. Rubinfeld B, Robbins P, El-Gamil M,
54. Hodgkinson CA, Moore KJ, Nakaya- 68. Takeuchi H, Kuo C, Morton DL, Wang Albert I, Porfiri E, Polakis P. Stabilization
ma A, et al. Mutations at the mouse mi- HJ, Hoon DS. Expression of differentia- of beta-catenin by genetic defects in mel-
crophthalmia locus are associated with tion melanoma-associated antigen genes anoma cell lines. Science 1997;275:1790-
defects in a gene encoding a novel basic- is associated with favorable disease out- 2.
helix-loop-helix-zipper protein. Cell 1993; come in advanced-stage melanomas. Can- 82. Cowley GP, Smith ME. Cadherin ex-
74:395-404. cer Res 2003;63:441-8. pression in melanocytic naevi and malig-
55. Nishimura EK, Granter SR, Fisher DE. 69. Miller AJ, Du J, Rowan S, Hershey CL, nant melanomas. J Pathol 1996;179:183-7.
Mechanisms of hair graying: incomplete Widlund HR, Fisher DE. Transcriptional 83. Rimm DL, Caca K, Hu G, Harrison
melanocyte stem cell maintenance in the regulation of the melanoma prognostic FB, Fearon ER. Frequent nuclear/cytoplas-
niche. Science 2005;307:720-4. marker melastatin (TRPM1) by MITF in mic localization of beta-catenin without
56. Widlund HR, Fisher DE. Microphtha- melanocytes and melanoma. Cancer Res exon 3 mutations in malignant melano-
lamia-associated transcription factor: a 2004;64:509-16. ma. Am J Pathol 1999;154:325-9.
critical regulator of pigment cell devel- 70. Duncan LM, Deeds J, Hunter J, et al. 84. Sanders DS, Blessing K, Hassan GA,
opment and survival. Oncogene 2003;22: Down-regulation of the novel gene mela- Bruton R, Marsden JR, Jankowski J. Al-
3035-41. statin correlates with potential for mela- terations in cadherin and catenin expres-
57. Lerner AB, Shiohara T, Boissy RE, Ja- noma metastasis. Cancer Res 1998;58: sion during the biological progression of
cobson KA, Lamoreux ML, Moellmann GE. 1515-20. melanocytic tumours. Mol Pathol 1999;52:
A mouse model for vitiligo. J Invest Der- 71. King R, Weilbaecher KN, McGill G, 151-7.
matol 1986;87:299-304. Cooley E, Mihm M, Fisher DE. Microph- 85. Widlund HR, Horstmann MA, Price
58. Steingrimsson E, Moore KJ, Lamor- thalmia transcription factor: a sensitive ER, et al. Beta-catenin-induced melanoma
eux ML, et al. Molecular basis of mouse and specific melanocyte marker for mela- growth requires the downstream target
microphthalmia (mi) mutations helps ex- noma diagnosis. Am J Pathol 1999;155:731- Microphthalmia-associated transcription
plain their developmental and phenotypic 8. factor. J Cell Biol 2002;158:1079-87.
consequences. Nat Genet 1994;8:256-63. 72. Miettinen M, Fernandez M, Franssila 86. Shtutman M, Zhurinsky J, Simcha I, et
59. McGill GG, Horstmann M, Widlund K, Gatalica Z, Lasota J, Sarlomo-Rikala M. al. The cyclin D1 gene is a target of the
HR, et al. Bcl2 regulation by the melano- Microphthalmia transcription factor in the beta-catenin/LEF-1 pathway. Proc Natl Acad
cyte master regulator Mitf modulates lin- immunohistochemical diagnosis of meta- Sci U S A 1999;96:5522-7.
eage survival and melanoma cell viability. static melanoma: comparison with four 87. Hsu M, Andl T, Li G, Meinkoth JL,
Cell 2002;109:707-18. other melanoma markers. Am J Surg Herlyn M. Cadherin repertoire determines
60. Banerjee D. Genasense (Genta Inc). Pathol 2001;25:205-11. partner-specific gap junctional communi-
Curr Opin Investig Drugs 2001;2:574-80. 73. Granter SR, Weilbaecher KN, Quigley cation during melanoma progression.
61. Goding CR. Mitf from neural crest to C, Fisher DE. Role for microphthalmia J Cell Sci 2000;113:1535-42.
melanoma: signal transduction and tran- transcription factor in the diagnosis of 88. Valyi-Nagy IT, Hirka G, Jensen PJ,
scription in the melanocyte lineage. metastatic malignant melanoma. Appl Im- Shih IM, Juhasz I, Herlyn M. Undifferenti-
Genes Dev 2000;14:1712-28. munohistochem Mol Morphol 2002;10:47- ated keratinocytes control growth, mor-
62. Du J, Miller AJ, Widlund HR, Horst- 51. phology, and antigen expression of normal
mann MA, Ramaswamy S, Fisher DE. 74. Garraway LA, Widlund HR, Rubin MA, melanocytes through cell-cell contact. Lab
MLANA/MART1 and SILV/PMEL17/GP100 et al. Integrative genomic analyses iden- Invest 1993;69:152-9.
are transcriptionally regulated by MITF in tify MITF as a lineage survival oncogene 89. Danen EH, de Vries TJ, Morandini R,
melanocytes and melanoma. Am J Pathol amplified in malignant melanoma. Na- Ghanem GG, Ruiter DJ, van Muijen GN.
2003;163:333-43. ture 2005;436:117-22. E-cadherin expression in human melano-
63. Baxter LL, Pavan WJ. Pmel17 expres- 75. Haass NK, Smalley KS, Li L, Herlyn M. ma. Melanoma Res 1996;6:127-31.
sion is Mitf-dependent and reveals cranial Adhesion, migration and communication 90. Hsu MY, Wheelock MJ, Johnson KR,
melanoblast migration during murine de- in melanocytes and melanoma. Pigment Herlyn M. Shifts in cadherin profiles be-
velopment. Gene Expr Patterns 2003;3:703- Cell Res 2005;18:150-9. tween human normal melanocytes and
7. 76. Balch CM, Soong SJ, Gershenwald JE, melanomas. J Investig Dermatol Symp
64. Loercher AE, Tank EMH, Delston RB, et al. Prognostic factors analysis of 17,600 Proc 1996;1:188-94.
Harbour JW. MITF links differentiation melanoma patients: validation of the 91. Scott RA, Lauweryns B, Snead DM,
with cell cycle arrest in melanocytes by American Joint Committee on Cancer Haynes RJ, Mahida Y, Dua HS. E-cadherin
transcriptional activation of INK4A. J Cell melanoma staging system. J Clin Oncol distribution and epithelial basement mem-
Biol 2005;168:35-40. 2001;19:3622-34. brane characteristics of the normal human
65. Hofbauer GF, Kamarashev J, Geertsen 77. Johnson JP. Cell adhesion molecules conjunctiva and cornea. Eye 1997;11:607-
R, Boni R, Dummer R. Melan A/MART-1 in the development and progression of 12.
immunoreactivity in formalin-fixed par- malignant melanoma. Cancer Metastasis 92. Gottardi CJ, Wong E, Gumbiner BM.
affin-embedded primary and metastatic Rev 1999;18:345-57. E-cadherin suppresses cellular transfor-
melanoma: frequency and distribution. 78. Bienz M. Beta-catenin: a pivot between mation by inhibiting beta-catenin signal-
Melanoma Res 1998;8:337-43. cell adhesion and Wnt signalling. Curr ing in an adhesion-independent manner.
66. Salti GI, Manougian T, Farolan M, Biol 2005;15:R64-R67. J Cell Biol 2001;153:1049-60.
Shilkaitis A, Majumdar D, Das Gupta TK. 79. Gottardi CJ, Gumbiner BM. Adhesion 93. Qi J, Chen N, Wang J, Siu CH. Trans-
Micropthalmia transcription factor: a new signaling: how beta-catenin interacts with endothelial migration of melanoma cells
prognostic marker in intermediate-thick- its partners. Curr Biol 2001;11:R792-R794. involves N-cadherin-mediated adhesion and

64 n engl j med 355;1 www.nejm.org july 6, 2006

The New England Journal of Medicine


Downloaded from nejm.org at GLASGOW UNIVERSITY LIBRARY on April 6, 2013. For personal use only. No other uses without permission.
Copyright © 2006 Massachusetts Medical Society. All rights reserved.
mechanisms of disease

activation of the beta-catenin signaling pha v beta 3 in hematogenous metastasis FG. Relative reciprocity of NRAS and
pathway. Mol Biol Cell 2005;16:4386-97. of human melanoma cells. Clin Exp Metas- PTEN/MMAC1 alterations in cutaneous
94. Li G, Satyamoorthy K, Herlyn M. tasis 2002;19:427-36. melanoma cell lines. Cancer Res 2000;60:
N-cadherin-mediated intercellular inter- 99. Hofmann UB, Westphal JR, Waas ET, 1800-4.
actions promote survival and migration Becker JC, Ruiter DJ, van Muijen GN. Co- 104. Daniotti M, Oggionni M, Ranzani T,
of melanoma cells. Cancer Res 2001;61: expression of integrin alpha(v)beta3 and et al. BRAF alterations are associated
3819-25. matrix metalloproteinase-2 (MMP-2) co- with complex mutational profiles in ma-
95. Kuphal S, Bauer R, Bosserhoff AK. In- incides with MMP-2 activation: correla- lignant melanoma. Oncogene 2004;23:
tegrin signaling in malignant melanoma. tion with melanoma progression. J Invest 5968-77.
Cancer Metastasis Rev 2005;24:195-222. Dermatol 2000;115:625-32. 105. Tsao H, Goel V, Wu H, Yang G,
96. Danen EH, Ten Berge PJ, Van Muijen 100. Petitclerc E, Stromblad S, von Schal- Haluska FG. Genetic interaction between
GN, Van ’t Hof-Grootenboer AE, Brocker scha TL, et al. Integrin alpha(v)beta3 pro- NRAS and BRAF mutations and PTEN/
EB, Ruiter DJ. Emergence of alpha 5 beta motes M21 melanoma growth in human MMAC1 inactivation in melanoma. J Invest
1 fibronectin- and alpha v beta 3 vitronec- skin by regulating tumor cell survival. Dermatol 2004;122:337-41.
tin-receptor expression in melanocytic tu- Cancer Res 1999;59:2724-30. 106. Maldonado JL, Fridlyand J, Patel H,
mour progression. Histopathology 1994;24: 101. Li X, Regezi J, Ross FP, et al. Integrin et al. Determinants of BRAF mutations in
249-56. alphavbeta3 mediates K1735 murine mel- primary melanomas. J Natl Cancer Inst
97. Brooks PC, Stromblad S, Sanders LC, anoma cell motility in vivo and in vitro. 2003;95:1878-90.
et al. Localization of matrix metallopro- J Cell Sci 2001;114:2665-72. 107. Chudnovsky Y, Adams AE, Robbins
teinase MMP-2 to the surface of invasive 102. Dechantsreiter MA, Planker E, Matha PB, Lin Q, Khavari PA. Use of human tis-
cells by interaction with integrin alpha v B, et al. N-methylated cyclic RGD peptides sue to assess the oncogenic activity of
beta 3. Cell 1996;85:683-93. as highly active and selective alpha(V)beta(3) melanoma-associated mutations. Nat Gen-
98. Felding-Habermann B, Fransvea E, integrin antagonists. J Med Chem 1999; et 2005;37:745-9.
O’Toole TE, Manzuk L, Faha B, Hensler 42:3033-40. Copyright © 2006 Massachusetts Medical Society.
M. Involvement of tumor cell integrin al- 103. Tsao H, Zhang X, Fowlkes K, Haluska

JOURNAL EDITORIAL FELLOW


The Journal’s editorial office invites applications for a one-year
research fellowship beginning in July 2007 from individuals at any
stage of training. The editorial fellow will work on Journal projects
and will participate in the day-to-day editorial activities of the Journal
but is expected in addition to have his or her own independent
projects. Please send curriculum vitae and research interests
to the Editor-in-Chief, 10 Shattuck St., Boston, MA 02115
(fax, 617-739-9864), by October 1, 2006.

n engl j med 355;1 www.nejm.org july 6, 2006 65

The New England Journal of Medicine


Downloaded from nejm.org at GLASGOW UNIVERSITY LIBRARY on April 6, 2013. For personal use only. No other uses without permission.
Copyright © 2006 Massachusetts Medical Society. All rights reserved.

You might also like