15
The new england journal of medicine n engl j med 355;1 www.nejm.org july 6, 2006 51 review article mechanisms of disease Melanoma Arlo J. Miller, M.D., Ph.D., and Martin C. Mihm, Jr., M.D. From the Dermatopathology Unit, Massa- chusetts General Hospital, and Harvard Medical School — both in Boston. Ad- dress reprint requests to Dr. Mihm at the Department of Dermatopathology, Massa- chusetts General Hospital, 55 Fruit St., Warren 827, Boston, MA 02114. N Engl J Med 2006;355:51-65. Copyright © 2006 Massachusetts Medical Society. A lthough melanoma accounts for only 4 percent of all derma- tologic cancers, it is responsible for 80 percent of deaths from skin cancer; only 14 percent of patients with metastatic melanoma survive for five years. 1 The intractability of advanced melanoma shows how much we have to learn about the changes that facilitate the vertical growth and deep invasion of melanoma and about the mechanisms that block the effectiveness of chemotherapy. The Clark model of the progression of melanoma emphasizes the stepwise trans- formation of melanocytes to melanoma (Fig. 1). The model depicts the proliferation 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 genomic 3 and proteomic 4 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). Environmental and Genetic Interactions 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, The New England Journal of Medicine Downloaded from nejm.org on March 17, 2015. For personal use only. No other uses without permission. Copyright © 2006 Massachusetts Medical Society. All rights reserved.

Melanoma

Embed Size (px)

Citation preview

  • T h e n e w e ng l a nd j o u r na l o f m e dic i n e

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

    review article

    mechanisms of disease

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

    From the Dermatopathology Unit, Massa-chusetts General Hospital, and Harvard Medical School both in Boston. Ad-dress reprint requests to Dr. Mihm at the Department of Dermatopathology, Massa-chusetts General Hospital, 55 Fruit St., Warren 827, Boston, MA 02114.

    N Engl J Med 2006;355:51-65.Copyright 2006 Massachusetts Medical Society.

    A lthough melanoma accounts for only 4 percent of all derma-tologic cancers, it is responsible for 80 percent of deaths from skin cancer; only 14 percent of patients with metastatic melanoma survive for five years.1 The intractability of advanced melanoma shows how much we have to learn about the changes that facilitate the vertical growth and deep invasion of melanoma and about the mechanisms that block the effectiveness of chemotherapy.

    The Clark model of the progression of melanoma emphasizes the stepwise trans-formation of melanocytes to melanoma (Fig. 1). The model depicts the proliferation 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 tic In ter ac tions

    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 geneticenvironmental 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,

    The New England Journal of Medicine Downloaded from nejm.org on March 17, 2015. For personal use only. No other uses without permission.

    Copyright 2006 Massachusetts Medical Society. All rights reserved.

  • T h e n e w e ng l a nd j o u r na l o f m e dic i n e

    n engl j med 355;1 www.nejm.org july 6, 200652

    where it absorbs and dissipates ultraviolet energy.7 Clinically, variations in pigmentation and the tan-ning response to ultraviolet light are associated with variations in susceptibility to melanoma.8,9

    At the molecular level, exposure to ultraviolet light increases skin pigmentation, in part through the action of -melanocytestimulating hormone (-MSH) on its receptor, the melanocortin recep-tor 1 (MC1R) (Fig. 3). Binding of the hormone to the receptor stimulates intracellular signaling in melanocytes, and this signaling increases the ex-pression of enzymes involved in the production of melanin. Light-skinned and redheaded people often carry germ-line polymorphisms in the MC1R gene10,11 that reduce the activity of the receptor.12 Such polymorphisms increase the risk of mel-anoma considerably.13 In light-skinned people, therefore, the basis of increased susceptibility to melanoma is a genetic impairment in the produc-tion of melanin, the main defense of melanocytes against ultraviolet radiation.

    Although the tanning response to ultraviolet radiation appears dose-dependent, the nature of the exposure is also a factor. Melanoma occurs most frequently after intermittent exposure to the sun and in people with frequent sunburns. Epi-demiologic observations suggest that chronic or low-grade exposures to ultraviolet light induce protection against DNA damage, whereas intense, intermittent exposures cause genetic damage.7

    A Molecul a r Model of Mel a nom a Pro gr ession

    The Clark model (Fig. 1) describes the histologic changes that accompany the progression from normal melanocytes to malignant melanoma.2 We will relate these histologic changes to particu-lar gene mutations (Table 1) in melanoma and dis-cuss how these mutations affect molecular sig-naling to contribute to the progression from normal melanocytes to melanoma (Fig. 2).

    Hyperplasia and Nevus Formation

    In the Clark model, the first phenotypic change in melanocytes is the development of benign nevi, which are composed of neval melanocytes (Fig. 1). The control of growth in these cells is disrupted, yet the growth of a nevus is limited a nevus rarely progresses to cancer.2 The absence of pro-gression is probably due to oncogene-induced cell senescence, in which growth that is stimulat-

    ed by the activation of oncogenic pathways is lim-ited.14 At a molecular level, abnormal activation of the mitogen-activated protein kinase (MAPK) signaling pathway (also called extracellular-related kinase [ERK]) stimulates growth in melanoma cells (Fig. 4A).15-17 Activation of this pathway is the result of somatic mutations of N-RAS, which are associated with about 15 percent of melanomas, or BRAF, which are associated with about 50 per-cent of melanomas. These mutations, which occur exclusively of each other, cause constitutive acti-vation of the serinethreonine kinases in the ERKMAPK pathway.18-20

    BRAF mutations occur at a similar frequency in benign nevi and in primary and metastatic melanomas.21 Since most nevi cease proliferation and remain static for decades, these similar fre-quencies suggest that nevi must acquire addition-al molecular lesions to free themselves of growth restraints and become malignant. Experiments in model systems support this hypothesis. In zebra-fish, melanocyte-specific expression of a mutant BRAF protein causes an ectopic proliferation of melanocytes, analogous to human nevi.22 In these fish, the combination of a BRAF mutation and inactivation of the tumor-suppressor gene p53 causes melanocytes to become malignant.22 In human melanocytes, mutant BRAF protein induc-es cell senescence by increasing the expression of the cell-cycle inhibitor of kinase 4A (INK4A).23 INK4A limits hyperplastic growth caused by a BRAF mutation. The arrest of the cell cycle caused by INK4A can, however, be overcome by mutations in INK4A itself, as well as other cell-cycle factors.

    In vitro, depletion of BRAF and N-RAS from melanoma cells suppresses their growth.24-26 Small molecules that inhibit BRAF are being tested clinically (BAY 43-9006) but have had only limited success as single agents.27 In mice, the

    Figure 1 (facing page). The Clark Model (Hematoxylin and Eosin).

    Melanocytes progress through a series of steps toward malignant transformation. The frequency of both the progression of nevi toward becoming malignant and the regression of nevi is unknown. The model empha-sizes the histopathological changes that occur in the progression of melanoma. Normal melanocytes pro-gressively develop a malignant phenotype through the acquisition of various phenotypic features. The partic-ular histologic features characterizing each step of pro-gression are the visible manifestations of underlying genetic changes.

    The New England Journal of Medicine Downloaded from nejm.org on March 17, 2015. 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 on March 17, 2015. For personal use only. No other uses without permission.

    Copyright 2006 Massachusetts Medical Society. All rights reserved.

  • T h e n e w e ng l a nd j o u r na l o f m e dic i n e

    n engl j med 355;1 www.nejm.org july 6, 200654

    growth of melanomas with BRAF mutations can be suppressed by the inhibition of the down-stream MEK enzymes, providing a possible target for treatment.28

    Cytologic Atypia and Tumor-Suppressor Genes

    The Clark model suggests that the next step toward melanoma is the development of cytologic atypia in dysplastic nevi, which may arise from preex-isting benign nevi or as new lesions. The molecu-lar abnormalities at this stage of progression affect cell growth, DNA repair, and the suscepti-bility to cell death. In 25 to 40 percent of cases of familial melanoma,6 a genetic defect inactivates CDKN2A, a single gene that encodes two tumor-suppressor proteins, p16INK4A and p19ARF29,30; in 25 to 50 percent of nonfamilial melanoma,31,32 a different tumor-suppressor gene, phosphatase and tensin homologue (PTEN) (Fig. 3), is inactivated by mutation.33,34 In murine models of melanoma, mutation of either CDKN2A or PTEN alone fails to cause melanoma, but when combined with each other or with mutations in other genes,35 mela-nomas do arise. Mutation of CDKN2A or PTEN is only one molecular step on the path to the devel-opment of melanoma, but it is unclear precisely when such mutations occur. The increased sus-ceptibility to melanoma that is associated with loss of the germ-line CDKN2A gene suggests that this genetic lesion increases the probability that dysplastic nevi will become malignant or increas-es the rate of the development of new melanoma without a precursor.

    CDKN2AThe G1S checkpoint that governs the commitment of a cell to DNA replication during the S phase (synthesis of DNA) is a site where many pathways that control cell division converge36,37 (Fig. 4B). In some familial and sporadic cases of melano-ma,36,37 the CDKN2A locus is lost by homozygous deletion of a portion of chromosome 9.36-38 One of the genes in this locus encodes INK4A, (p16INK4A), a protein that blocks the cell cycle at the G1S checkpoint by inhibiting cyclin-depen-dent kinases. INK4A (an inhibitor of CDK4) sup-presses the proliferation of cells with damaged DNA or activated oncogenes and also acts when cells are old or crowded.39 Mice lacking INK4A appear normal but are abnormally sensitive to carcinogens and prone to the development of

    tumors.40 The development of melanoma in such mice requires mutations in other genes, such as an activating mutation in H-RAS, an upstream component in MAPK signaling, which triggers MEK signaling.41 Genes that encode CDK4 and cyclin D1 (CCND1) encode proteins that act down-stream of INK4A, and they are also mutated in some melanomas. These targets of INK4A func-tion together as part of a complex that promotes the progression of the cell cycle by phosphorylat-ing retinoblastoma (Rb) protein, a cell-cycle reg-ulator. Rare melanoma kindreds carry germ-line mutations in CDK4 that disrupt cell-cycle control by preventing the molecular interaction that allows INK4A to repress CDK4.42 Mice that carry the hu-man CDK4 mutation are prone to melanoma when exposed to various carcinogens.43

    The D-type cyclin CD1 may have an oncogenic role in acral melanoma, in which amplification of the CCND1 gene and overexpression of cyclin CD1 protein occur more frequently than in melanoma at other sites.44 Inhibition of CCND1 (with anti-sense CCND1) causes apoptosis of human mela-noma xenografts implanted in immunodeficient mice, without an apparent effect on normal mela-nocytes.

    Alternative splicing of various exons within CDKN2A yields two distinct tumor-suppressor pro-teins, INK4A and alternate reading frame (ARF) (Fig. 3).39 The ARF gene (also called p14ARF) de-rives its name from the use of an alternative reading frame of the exons it shares with INK4A. ARF functions as a tumor suppressor by arrest-ing the cell cycle or promoting cell death after DNA damage or when various oncogenes or loss of Rb stimulate aberrant cell proliferation. ARF participates in the core regulatory process that controls levels of the p53 protein. It acts through the mouse double minute 2 (MDM2) protein, which triggers the ubiquination of p53, thereby instigating its destruction in the proteosome. ARF binds to MDM2, sequestering it from p53 and in this way causes p53 to accumulate; p53 then arrests the cell cycle at the G2M site, allow-ing for repair of damaged DNA or the induction of apoptosis.45,46 In cells, ARF deficiency abro-gates oncogene-induced senescence and increases susceptibility to transformation.47 In vitro, im-mortalization of cells often occurs with the loss of either ARF or p53.48 In animals, ARF deficiency shortens the time required for the development of melanoma after exposure to ultraviolet light;

    The New England Journal of Medicine Downloaded from nejm.org on March 17, 2015. 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 55

    when both gene products of CDKN2A (INK4A and ARF) are deficient, the latent period is even short-er.49 These data suggest how ARF facilitates the progression of melanoma and indicate that the low frequency of p53 mutations in melanoma is partly related to loss of ARF, which renders the p53 pathway inactive.39

    PTEN, AKT, and Cell Death

    A second chromosomal region that is frequently affected by homozygous deletion in melanoma and other cancers is the PTEN locus on chromo-

    some 10.33,34,50 PTEN encodes a phosphatase that attenuates signaling by a variety of growth factors that use phosphatidylinositol phosphate (PIP

    3) as

    an intracellular signal. In the presence of such growth factors, intracellular levels of PIP

    3 rapidly

    increase. This increase triggers the activation of protein kinase B (PKB, also called AKT) by phos-phorylation (Fig. 3). Activated AKT phosphory-lates and inactivates proteins that suppress the cell cycle or stimulate apoptosis, thereby facilitat-ing the proliferation and survival of cells. PTEN normally keeps PIP

    3 levels low; in its absence,

    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, V3 integrin, and matrix metalloproteinase 2 (MMP-2).

    The New England Journal of Medicine Downloaded from nejm.org on March 17, 2015. For personal use only. No other uses without permission.

    Copyright 2006 Massachusetts Medical Society. All rights reserved.

  • T h e n e w e ng l a nd j o u r na l o f m e dic i n e

    n engl j med 355;1 www.nejm.org july 6, 200656

    Tabl

    e 1.

    Impo

    rtan

    t Gen

    es in

    Mel

    anom

    a.

    Path

    way

    Gen

    e or

    Pro

    tein

    *Fu

    nctio

    nC

    hang

    es in

    Mel

    anom

    a

    RA

    S an

    d M

    APK

    N-R

    AS

    Onc

    ogen

    eSp

    orad

    ic a

    ctiv

    atin

    g m

    utat

    ion

    at G

    13R

    BR

    AF

    Onc

    ogen

    eSp

    orad

    ic a

    ctiv

    atin

    g m

    utat

    ion

    at c

    odon

    V60

    0E in

    ne

    vi a

    nd m

    elan

    oma

    Mito

    gen-

    activ

    ated

    pro

    tein

    kin

    ase

    extr

    acel

    lula

    r-re

    late

    d ki

    nase

    (M

    EK)

    Sign

    al tr

    ansd

    uctio

    nU

    p-re

    gula

    ted

    in r

    adia

    l-gro

    wth

    and

    ver

    tical

    -gro

    wth

    ph

    ases

    Extr

    acel

    lula

    r-re

    late

    d ki

    nase

    1 o

    r 2

    (ER

    K1

    or E

    RK

    2)

    or m

    itoge

    n-ac

    tivat

    ed p

    rote

    in k

    inas

    e (M

    APK

    )Si

    gnal

    tran

    sduc

    tion

    Incr

    ease

    d ac

    tivity

    INK

    4A, C

    DK

    , and

    Rb

    Cyc

    lin-d

    epen

    dent

    kin

    ase

    inhi

    bito

    r 2A

    or

    inhi

    bito

    r of

    kin

    ase

    4A (

    CD

    KN

    2A o

    r IN

    K4A

    )Tu

    mor

    sup

    pres

    sor

    nega

    tive

    regu

    lato

    r of

    cel

    l pro

    lif-

    erat

    ion

    Ger

    m-li

    ne m

    utat

    ions

    in s

    ome

    fam

    ilial

    mel

    anom

    as;

    spor

    adic

    del

    etio

    ns, p

    rom

    oter

    inac

    tivat

    ion,

    loss

    of

    het

    eroz

    ygos

    ity in

    man

    y m

    elan

    omas

    Cyc

    lin-d

    epen

    dent

    kin

    ase

    4 (C

    DK

    4)Pr

    omot

    er o

    f cel

    l pro

    lifer

    atio

    nPr

    otei

    n in

    sens

    itive

    to in

    hibi

    tion

    by IN

    K4A

    due

    to

    rare

    fam

    ilial

    ger

    m-li

    ne m

    utat

    ions

    at R

    24C

    Cyc

    lin D

    1 (C

    CN

    D1)

    Prom

    oter

    of c

    ell p

    rolif

    erat

    ion

    Spor

    adic

    am

    plifi

    catio

    n in

    acr

    al m

    elan

    oma

    Ret

    inob

    last

    oma

    (Rb)

    Tum

    or s

    uppr

    esso

    rne

    gativ

    e re

    gula

    tor

    of c

    ell p

    rolif

    -er

    atio

    nPh

    osph

    oryl

    atio

    n le

    ads

    to p

    rogr

    essi

    on fr

    om

    G1

    to S

    AR

    F an

    d p5

    3

    Alte

    rnat

    e re

    adin

    g fr

    ame

    (AR

    F)Tu

    mor

    sup

    pres

    sor,

    deg

    rade

    s M

    DM

    2G

    erm

    -line

    mut

    atio

    ns in

    som

    e fa

    mili

    al m

    elan

    omas

    ; sp

    orad

    ic d

    elet

    ions

    , pro

    mot

    er in

    activ

    atio

    n, in

    m

    any

    mel

    anom

    as

    Tum

    or p

    rote

    in 5

    3 (p

    53)

    Tum

    or s

    uppr

    esso

    r th

    at in

    duce

    s ap

    opto

    sis

    and

    sup-

    pres

    sed

    prol

    ifera

    tion

    afte

    r D

    NA

    dam

    age

    Expr

    essi

    on u

    sual

    ly p

    rese

    nt in

    mel

    anom

    a

    Mou

    se d

    oubl

    e m

    inut

    e 2

    (MD

    M2)

    Targ

    eter

    of p

    53 fo

    r ub

    iqui

    natio

    n an

    d de

    stru

    ctio

    nU

    p-re

    gula

    ted

    in p

    rese

    nce

    of A

    RF

    mut

    atio

    n

    BC

    L-2

    asso

    ciat

    ed X

    pro

    tein

    (B

    AX

    )In

    duce

    r of

    cel

    l dea

    thV

    aria

    ble

    but u

    sual

    ly d

    own-

    regu

    late

    d

    PTEN

    and

    AK

    T

    Phos

    phat

    ase

    and

    tens

    in h

    omol

    ogue

    (PT

    EN)

    Tum

    or s

    uppr

    esso

    r, r

    epre

    sses

    PI3

    KSp

    orad

    ic d

    elet

    ion

    of c

    hrom

    osom

    al r

    egio

    n

    Phos

    phat

    idyl

    inos

    itol 3

    kin

    ase

    (PI3

    K)

    Sign

    alin

    g m

    olec

    ule

    for

    man

    y gr

    owth

    fact

    ors

    Act

    ive

    in p

    rese

    nce

    of P

    TEN

    mut

    atio

    n

    Prot

    ein

    kina

    se B

    (A

    KT

    or P

    KB

    )O

    ncog

    ene

    that

    is a

    ctiv

    ated

    by

    PI3K

    , lea

    ding

    to

    incr

    ease

    d ce

    ll su

    rviv

    alA

    mpl

    ified

    in s

    ome

    mel

    anom

    as

    BC

    L-2

    anta

    goni

    st o

    f cel

    l dea

    th (

    BA

    D)

    Indu

    cer

    of c

    ell d

    eath

    Var

    iabl

    e bu

    t oft

    en d

    own-

    regu

    late

    d

    Fork

    head

    rec

    epto

    r (F

    KH

    R)

    Gro

    wth

    sup

    ress

    ion

    Act

    ivat

    ed in

    res

    pons

    e to

    PI3

    pat

    hway

    The New England Journal of Medicine Downloaded from nejm.org on March 17, 2015. 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 57

    MSH

    and

    MIT

    F

    Pro-

    opio

    mel

    anoc

    ortin

    or

    -m

    elan

    ocyt

    est

    imul

    atin

    g ho

    rmon

    e (P

    OM

    C o

    r

    -MSH

    )Si

    gnal

    ing

    mol

    ecul

    e im

    port

    ant i

    n pi

    gmen

    tatio

    nIn

    crea

    sed

    mel

    anom

    a ve

    rtic

    al-g

    row

    th p

    hase

    Mel

    anoc

    ortin

    rec

    epto

    r 1

    (MC

    1R)

    Rec

    epto

    r fo

    r

    -MSH

    Poly

    mor

    phic

    gen

    e af

    fect

    ing

    hair

    and

    ski

    n co

    lor

    and

    resp

    onse

    to u

    ltrav

    iole

    t rad

    iatio

    n

    Ade

    nyla

    te c

    ycla

    se (

    AC

    )Pr

    oduc

    er o

    f cyc

    lic A

    MP

    Up-

    regu

    late

    d

    cAM

    P re

    spon

    se e

    lem

    ent

    bind

    ing

    prot

    ein

    (CR

    EB)

    Tran

    scri

    ptio

    n fa

    ctor

    Up-

    regu

    late

    d; a

    ffect

    s M

    ITF

    and

    mel

    anoc

    yte

    diffe

    rent

    iatio

    n

    Mic

    roph

    thal

    mia

    -ass

    ocia

    ted

    tran

    scri

    ptio

    n fa

    ctor

    (M

    ITF)

    Tran

    scri

    ptio

    n fa

    ctor

    Spor

    adic

    am

    plifi

    catio

    n of

    chr

    omos

    omal

    reg

    ion

    Tyro

    sina

    se (

    TYR

    )Pi

    gmen

    t syn

    thes

    isD

    ecre

    ased

    exp

    ress

    ion

    Tyro

    sina

    se-r

    elat

    ed p

    rote

    in 1

    (TY

    RP1

    )Pi

    gmen

    t syn

    thes

    isD

    ecre

    ased

    exp

    ress

    ion

    Dop

    achr

    ome

    taut

    omer

    ase

    (DC

    T)Pi

    gmen

    t syn

    thes

    isD

    ecre

    ased

    exp

    ress

    ion

    Mel

    an-A

    (M

    LAN

    A)

    Ant

    igen

    rec

    ogni

    zed

    by m

    elan

    -A a

    nd m

    elan

    oma

    anti-

    gen

    reco

    gniz

    ed b

    y T-

    cells

    1 (

    MA

    RT1

    ) an

    tibod

    ies

    Dec

    reas

    ed e

    xpre

    ssio

    n

    Silv

    er h

    omol

    ogue

    (SI

    LV)

    Ant

    igen

    rec

    ogni

    zed

    by H

    MB

    -45

    antib

    ody

    Dec

    reas

    ed e

    xpre

    ssio

    n

    Mel

    asta

    tin 1

    (TR

    PM1)

    Unk

    now

    nD

    ecre

    ased

    exp

    ress

    ion

    in m

    etas

    tatic

    mel

    anom

    a

    BC

    L-2

    Cel

    l sur

    viva

    lV

    aria

    ble

    up-r

    egul

    atio

    n in

    var

    ious

    pha

    ses

    of

    mel

    anom

    a

    Cel

    l adh

    esio

    n

    Win

    gles

    s-ty

    pe m

    amm

    ary

    tum

    or v

    irus

    inte

    grat

    ion-

    site

    fam

    ily (

    WN

    T)Pr

    otoo

    ncog

    ene,

    sec

    rete

    d gr

    owth

    fact

    or th

    at in

    acti-

    vate

    sG

    SK3-

    B

    Path

    way

    up-

    regu

    late

    d

    Gly

    coge

    n sy

    ntha

    se k

    inas

    e 3

    (G

    SK3

    )Se

    rine

    thr

    eoni

    ne k

    inas

    e th

    at ta

    rget

    s

    -cat

    enin

    for

    degr

    adat

    ion

    Var

    iabl

    e; a

    ffect

    ed b

    y W

    NT

    path

    way

    -C

    aten

    inA

    dher

    ens

    junc

    tion

    prot

    ein,

    tran

    scri

    ptio

    nal c

    o-ac

    tivat

    orSp

    orad

    ic s

    tabi

    lizin

    g m

    utat

    ions

    T-ce

    ll fa

    ctor

    lym

    phoi

    d-en

    hanc

    ing

    fact

    or (

    TCF

    LEF)

    Tran

    scri

    ptio

    n fa

    ctor

    Up-

    regu

    late

    d

    E-ca

    dher

    inC

    ell-a

    dhes

    ion

    mol

    ecul

    eD

    ecre

    ased

    exp

    ress

    ion

    in v

    ertic

    al-g

    row

    th p

    hase

    N-c

    adhe

    rin

    Cel

    l-adh

    esio

    n m

    olec

    ule

    Abe

    rran

    t exp

    ress

    ion

    in v

    ertic

    al-g

    row

    th p

    hase

    V

    3

    inte

    grin

    Dim

    er th

    at fo

    rms

    cell-

    adhe

    sion

    mol

    ecul

    eA

    berr

    ant e

    xpre

    ssio

    n in

    ver

    tical

    -gro

    wth

    pha

    se

    * A

    bbre

    viat

    ed fo

    rms

    of t

    he g

    ene

    are

    give

    n in

    par

    enth

    eses

    .

    The New England Journal of Medicine Downloaded from nejm.org on March 17, 2015. For personal use only. No other uses without permission.

    Copyright 2006 Massachusetts Medical Society. All rights reserved.

  • T h e n e w e ng l a nd j o u r na l o f m e dic i n e

    n engl j med 355;1 www.nejm.org july 6, 200658

    levels of PIP3 and active (phosphorylated) AKT in-

    crease. Increased AKT activity prolongs cell sur-vival through the inactivation of BCL-2 antago-nist of cell death (BAD) protein and increases cell proliferation by increasing CCND1 expres-sion, and affects many other cell-survival and cell-cycle genes through the activation of the forkhead (FKHR) transcription factor.32,51 AKT activity can

    also be increased in cells by mutations that cause the amplification and overexpression of the pro-tein. Restoration of PTEN in cultured mouse me-lanocytes decreases the ability of the cells to form tumors.52 In model systems, suppression of AKT3, a member of the AKT family, reduces the survival of melanoma cells and the growth of human mela-nomas implanted in immunodeficient nude mice.53

    Figure 3. Microphthalmia-Associated Transcription Factor (MITF) and -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 -catenin pathway, -catenin plays a central role in cell adhesion and cell signaling. Signals from WNT ligands block the breakdown of -catenin. When WNT proteins bind the G-proteincoupled receptor (called frizzled), they inacti-vate the kinase GSK3, an enzyme that phosphorylates -catenin and targets it for destruction in the proteosome. Then -catenin accu-mulates in the cytoplasm and translocates to the nucleus, where it binds to LEFTCF transcription factors and increases the expression of several genes, including MITF, the cell-cycle mediator cyclin D1 (CCND1), and matrix metalloproteinase 7 (MMP-7).

    The New England Journal of Medicine Downloaded from nejm.org on March 17, 2015. 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 59

    As compared with normal melanocytes, increased levels of the active form of AKT were found in the radial-growth phase.53

    MI TF a nd Mel a no c y te Differ en ti ation

    Clark proposed that many nevi regress through differentiation and that the failure of differentia-tion is necessary for dysplasia.2 The normal pro-cess of melanocyte differentiation requires exit from the cell cycle and the expression of genes that encode proteins necessary for the produc-tion of pigment two processes that are de-regulated in melanoma. The microphthalmia-associated transcription factor (MITF) regulates the development and differentiation of melano-cytes54 and maintains melanocyte progenitor cells in adults.55,56

    MITF in Development

    Mice lacking functional MITF are albino because they lack melanocytes, whereas those with par-tial MITF function have premature graying owing to the death of melanocytes. These experiments show that MITF is important in the differentiation and maintenance of melanocytes.57,58 MITF ap-pears to contribute to melanocyte survival by in-creasing the expression of the BCL-2 gene, a key antiapoptotic factor.59 In mice, deficiencies of both MITF and BCL-2 cause gray hair due to a loss of differentiated melanocytes. The loss of melano-cytes is due to the apoptosis of melanocyte pro-genitor cells in the hair follicle.55 In melanoma cell lines, a reduction in BCL-2 protein also causes cell death, suggesting that the survival of malig-nant melanocytes depends on BCL-2.60

    MITF in Differentiation

    MITF functions in a key pathway leading to me-lanocyte pigmentation (Fig. 3). Intracellular sig-naling induced by -MSH acting on MC1R in-creases MITF expression, which in turn increases the transcription of genes underlying melanin synthesis: tyrosinase, tyrosinase-related-protein 1, and dopachrome tautomerase.61 MITF also regu-lates the transcription of the melanocyte-specific genes silver homologue (SILV)62,63 and melan-A (MLANA),62 whose immunohistochemical detec-tion points to the diagnosis of melanoma. In addi-tion, MITF causes cell-cycle arrest by the induction of INK4A.64

    MITF in Melanoma

    Decreased or absent pigmentation and decreased or absent expression of SILV and MLANA accom-pany the progression from nevus to melanoma. Tumors that are deficient in these proteins have a poor prognosis.65-68 Expression of the mela-statin 1 (TRPM1) gene, whose function is unknown, is also controlled by MITF.69 Melanomas that are deficient in melastatin have a poor prognosis.70 The mechanism of decreased expression of these genes is a puzzle because MITF is present in nearly all melanomas.71-73

    Although MITF causes differentiation and cell-cycle arrest in normal melanocytes, melanoma cells do not have these characteristics. Recently, a large-scale search for genomic changes in mela-noma with the use of high-density single-nucleo-tide polymorphisms (SNPs) found an increased copy number (4 to 119 copies per cell) of a region of chromosome 3 that includes the MITF locus.74 This increase was accompanied by the increased expression of MITF protein. The overexpression of both MITF and BRAF could transform primary cultures of human melanocytes, implicating MITF as an oncogene. Notably, MITF amplification oc-curs most frequently in tumors that have a poor prognosis and is associated with resistance to chemotherapy.74 Interference with MITF function increased the chemosensitivity of a melanoma cell line, making MITF a potential target for treatment.

    Cel l A dhesion a nd In va sion

    Local invasion and metastatic spread are respon-sible for the morbidity and mortality in melano-ma. In the Clark model, invasive characteristics appear in the vertical-growth phase, when mela-noma cells not only penetrate the basement mem-brane but also grow intradermally as an expand-ing nodule (Fig. 2). Metastatic melanoma develops when tumor cells dissociate from the primary lesion, migrate through the surrounding stroma, and invade blood vessels and lymphatics to form a tumor at a distant site.75 Clinically, the absolute depth of local invasion, measured directly by histo-pathologic analysis (the Breslow index), is the principal prognostic factor and primary criterion in melanoma staging.76 Invasion and spread of melanoma are related to alterations in cell adhe-sion. Normally, cell adhesion controls cell migra-tion, tissue organization, and organogenesis,77

    The New England Journal of Medicine Downloaded from nejm.org on March 17, 2015. For personal use only. No other uses without permission.

    Copyright 2006 Massachusetts Medical Society. All rights reserved.

  • T h e n e w e ng l a nd j o u r na l o f m e dic i n e

    n engl j med 355;1 www.nejm.org july 6, 200660

    but disturbances in cell adhesion contribute to tumor invasion, tumorstroma interactions, and tumor-cell signaling.

    Cadherins

    Cadherins are multifunctional transmembrane proteins that sustain cell-to-cell contacts, form connections with the actin cytoskeleton, and in-fluence intracellular signaling. The extracellular domain of cadherins binds to like cadherins on other cells in regions of cell contacts called adhe-rens junctions. Cadherins are divided into three subtypes: E (epithelial), present in polarized epi-thelial cells in the epidermis, including melano-cytes and keratinocytes; P (placental); and N (neu-ral), found in mesenchymal cells in the dermis. The intracellular domain is associated with a large protein complex that includes -catenin and forms structural links with bundles of actin filaments.

    Several signaling pathways cause -catenin to dissociate from the cell adhesion complex and transduce signals to the nucleus (Fig. 3). One of these pathways is called the wingless-type mam-mary tumor virus integration-site family (WNT) pathway. WNTs are secreted proteins with impor-tant functions in development, especially in neu-ral crest cells like melanocytes. When WNT pro-teins bind their receptors, they inactivate the kinase GSK3, an enzyme that phosphorylates -catenin and targets it for destruction in the pro-teosome.78,79 Tyrosine phosphorylation of -catenin disrupts the association between E-cadherin and -catenin,80 allowing -catenin to translocate to the nucleus, where it binds to lymphoid enhancer factorT-cell factor (LEFTCF). Mutations in the -catenin gene can stabilize the -catenin pro-tein81 or increase its nuclear localization.82-84 In-creased levels of nuclear -catenin increase the expression of MITF85 and CCND1,86 and these in turn increase the survival and proliferation of melanoma cells. Alterations in cadherin expres-sion affect the interaction of melanoma cells with the environment and alter -catenin signaling. E-cadherin expression occurs in melanocytes and keratinocytes in the epidermis and causes mela-nocytes to associate with keratinocytes.87 In turn, contacts with undifferentiated keratinocytes from the basal-cell layer inhibit melanocyte prolif-eration, suppress the expression of melanoma

    Figure 4 (facing page). MAPK and PTEN Pathways and the CDKN2A Tumor-Suppressor Locus.

    Panel A shows the pathway associated with N-RAS, BRAF, and mitogen-activated protein kinase (MAPK). MAPKs are involved in signaling from numerous growth factors and cell-surface receptors. There are many vari-ations in the components of particular cascades from various cell-surface receptors. Typically, adapter proteins (not shown) link the growth-factor receptor to RAS proteins, including N-RAS. When activated, RAS proteins phosphorylate (P) the mitogen-activated protein kinase (MEK) kinases, which then act on extracellular-related kinase (ERK) kinases. ERK kinases phosphorylate many targets in the cytoplasm and interact with other path-ways, including phosphatidylinositol 3 kinase (PI3K) and MITF. ERK kinases translocate to the nucleus, where they activate transcription factors that promote cell-cycle progression and proliferation by increasing the transcrip-tion of many genes, including CD1. In survival signaling associated with phosphatase and tensin homologue (PTEN) and AKT, also known as pro-tein kinase B, PTEN inhibits growth-factor signaling by inactivating phos-phatidylinositol triphosphate (PIP3) generated by PI3K. A variety of growth factors (PDGF, NGF, and IGF-1) bind to their respective receptor tyrosine kinases and activate PI3K. The activated molecule converts the plasma membrane lipid phosphatidylinositol 4,5-bisphosphonate to PIP3. PIP3 acts as a second messenger, leading to the phosphorylation and activation of AKT. AKT is itself a kinase that phosphorylates protein substrates that af-fect the cell cycle, growth, and survival. Often, these AKT targets are inacti-vated by phosphorylation. PTEN attenuates this pathway through dephos-phorylation and inactivation of PIP3, suppressing signaling from growth factors by blocking the activation of AKT. In Panel B, CDKN2A encodes two distinct tumor-suppressor genes; separate first exons that are spliced into alternate reading frames (ARF) of the second and third exons permit the expression of two different proteins from the same genetic locus. The gene has 4 exons. Transcription of messenger RNA (mRNA) can be initiated at either E1B or E1A, and the initiation site determines which gene the locus will express. RNA that is transcribed from either exon is spliced with the re-maining two exons, E2 and E3, to produce mRNA for either INK4A or ARF. However, ARF uses a different reading frame of the exon 2 and 3 codons. In the cell-cycle progression involving INK4A, ARF, and retinoblastoma pro-tein (Rb), a family of cyclins and cyclin-dependent kinases (CDKs) regulate progression through the cell cycle, and a family of CDK inhibitors opposes this action. In particular, the two phases of the G1S checkpoint are gov-erned primarily by cyclin D associated with cyclin-dependent kinases 4 and 6 (CDK4 and CDK6) at its early phase and cyclin A or E associated with CDK2 at the later restriction phase. INK4A encodes a cyclin-dependent ki-nase inhibitor that inhibits CDK4 and CDK6. Ordinarily, these two kinases associate with D-type cyclins and drive the cell cycle by phosphorylating Rb, releasing it from its inhibitory interaction with the E2F transcription factor, thereby allowing the expression of E2F-related genes and progres-sion from G1 to S. The absence of INK4A leads to unopposed CDK4 or 6 activity and increased cell-cycle activity. In response to DNA damage, mouse double minute 2 (MDM2) protein binds to the transcriptional acti-vation domain of protein 53 (p53), blocking p53-mediated gene regulation while simultaneously leading to p53 ubiquination, nuclear export, and pro-teosomal degradation. ARF opposes this action by sequestering MDM2. This disruption of the MDM2p53 interaction stabilizes p53 and increases p53 activity. Depending on other events, p53 either activates DNA repair and cell-cycle arrest or causes apoptosis and the formation of BCL-2asso-ciated X protein (BAX). In the absence of ARF, p53 levels are decreased and the response to DNA damage is blunted.

    The New England Journal of Medicine Downloaded from nejm.org on March 17, 2015. 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 on March 17, 2015. For personal use only. No other uses without permission.

    Copyright 2006 Massachusetts Medical Society. All rights reserved.

  • T h e n e w e ng l a nd j o u r na l o f m e dic i n e

    n engl j med 355;1 www.nejm.org july 6, 200662

    markers, and cause melanocytes to become den-dritic.88

    Progression from the radial-growth phase to the vertical-growth phase of melanoma is marked by the loss of E-cadherin and the expression of N-cadherin89-91 (Fig. 2). N-cadherin is a character-istic of invasive carcinomas and enables metastatic spread by permitting melanoma cells to interact with other N-cadherinexpressing cells, such as dermal fibroblasts and the vascular endothelium.87 Besides these changes in cell adhesion, decreased E-cadherin expression92 and aberrant N-cadherin expression increase the survival of melanoma cells by stimulating -catenin signaling.93,94

    Integrins

    The integrins mediate cell contacts with fibro-nectin, collagens, and laminin, components of the extracellular matrix.95 Transition from radial to vertical growth of melanoma is associated with the expression of V3 integrin.96 This integrin induces expression of matrix metalloproteinase 2, an enzyme that degrades the collagen in basement membrane.97-99 In addition, V3 integrin increas-es expression of the prosurvival gene BCL-2100 and stimulates the motility of melanoma cells through the reorganization of melanoma cytoskeleton.101

    These observations form a rationale for the de-velopment of integrin antagonists to treat mela-noma.102

    Patterns of Genetic Alteration

    The genetic changes in melanoma can be seen as particular combinations of molecular lesions that interrupt a precise set of pathways, each with a crucial role in the development of melanoma. The MEK pathway can be activated by a mutation in either NRAS or BRAF, and an NRAS mutation can activate both the MEK and PTEN pathways. Similarly, INK4A, CDK4, and CCND1 function in a unique pathway that affects the cell cycle; a mu-tation of INK4A has similar consequences as a mutation of CCND1 or CDK4.103-105

    There are particular genetic changes in mela-nomas in different sites, consistent differences related to ultraviolet exposure on sites that are

    chronically exposed (head and neck) or intermit-tently exposed (chest and back) and in acral and mucosal skin. For example, CCND1 amplification occurs predominantly in acral regions,44 whereas activating mutations in BRAF occur most frequent-ly in skin sites of intermittent sun exposure.106

    Model ing Mel a nom a Pro gr ession

    For many of the molecular lesions we have de-scribed, animal models have provided validation. A surprising new model is the zebrafish, in which premalignant and malignant lesions can be cre-ated by the expression of mutant BRAF with or without p53 mutation.22 This model is the only currently tractable system in which genetic screens can be performed for modifiers of mela-noma.

    Human melanomas that are grafted onto or injected into nude mice allow measures of the tumors metastatic potential and have allowed for the testing of therapeutic interventions. Genetic manipulation of mice has validated the contribu-tion of many genetic alterations in melanoma, but there are fundamental differences between mouse and human skin. Mouse melanocytes occur in hair follicles and the dermis, rather than in the epidermis, as in humans. To circumvent this problem, human melanocytes can be altered in cell culture and combined with keratinocytes to produce graft material. Using this system, the inactivation of p53 and the simultaneous intro-duction of activated N-RAS, CDK4, and telomer-ase led to darkly pigmented grafts that became grossly ulcerated and displayed histologic features of melanoma, including vertical invasion.107 This experimental system provides a novel model to test invasion and metastases of transformed hu-man melanocytes in a host organism.

    Supported by a grant (MCM202534) from the Cancer Research Institute of New York and a grant (T32-GM07753, to Dr. Miller) from the National Institute of General Medical Science. No other potential conflict of interest relevant to this article was reported.

    We are indebted to Drs. David E. Fisher, Adriano Piris, Jenni-fer Y. Lin, and Jennifer C. Broder for their critical reading of the manuscript, and to Dr. Claudio Clemente for contributing im-ages for Figure 1.

    References

    Cancer facts & figures, 2003. Atlanta: American Cancer Society, 2003.

    Clark WH Jr, Elder DE, Guerry D IV, Epstein MN, Greene MH, Van Horn M. A study of tumor progression: the precur-

    1.

    2.

    sor lesions of superficial spreading and nodular melanoma. Hum Pathol 1984;15:1147-65.

    Curtin JA, Fridlyand J, Kageshita T, et al. Distinct sets of genetic alterations 3.

    in melanoma. N Engl J Med 2005;353:2135-47.

    Alonso SR, Ortiz P, Pollan M, et al. Progression in cutaneous malignant mel-anoma is associated with distinct expres-

    4.

    The New England Journal of Medicine Downloaded from nejm.org on March 17, 2015. 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 63

    sion profiles: a tissue microarray-based study. Am J Pathol 2004;164:193-203.

    Aitken J, Welch J, Duffy D, et al. CD-KN2A variants in a population-based sam-ple of Queensland families with melano-ma. J Natl Cancer Inst 1999;91:446-52.

    Thompson JF, Scolyer RA, Kefford RF. Cutaneous melanoma. Lancet 2005;365:687-701.

    Gilchrest BA, Eller MS, Geller AC, Yaar M. The pathogenesis of melanoma induced by ultraviolet radiation. N Engl J Med 1999;340:1341-8.

    MacKie RM. Risk factors for the de-velopment of primary cutaneous malig-nant melanoma. Dermatol Clin 2002;20:597-600.

    Marks R. Epidemiology of melanoma. Clin Exp Dermatol 2000;25:459-63.

    Naysmith L, Waterston K, Ha T, et al. Quantitative measures of the effect of the melanocortin 1 receptor on human pig-mentary status. J Invest Dermatol 2004;122:423-8.

    Valverde P, Healy E, Jackson I, Rees JL, Thody AJ. Variants of the melanocyte-stimulating hormone receptor gene are associated with red hair and fair skin in humans. Nat Genet 1995;11:328-30.

    Frandberg PA, Doufexis M, Kapas S, Chhajlani V. Human pigmentation pheno-type: a point mutation generates non-functional MSH receptor. Biochem Biophys Res Commun 1998;245:490-2.

    Kennedy C, ter Huurne J, Berkhout M, et al. Melanocortin 1 receptor (MC1R) gene variants are associated with an in-creased risk for cutaneous melanoma which is largely independent of skin type and hair color. J Invest Dermatol 2001;117:294-300.

    Braig M, Schmitt CA. Oncogene-induced senescence: putting the brakes on tumor development. Cancer Res 2006;66:2881-4.

    Welsh CF, Roovers K, Villanueva J, Liu Y, Schwartz MA, Assoian RK. Timing of cyclin D1 expression within G1 phase is controlled by Rho. Nat Cell Biol 2001;3:950-7.

    Brunet A, Roux D, Lenormand P, Dowd S, Keyse S, Pouyssegur J. Nuclear translocation of p42/p44 mitogen-activat-ed protein kinase is required for growth factor-induced gene expression and cell cycle entry. EMBO J 1999;18:664-74.

    Lin AW, Barradas M, Stone JC, van Aelst L, Serrano M, Lowe SW. Premature senescence involving p53 and p16 is acti-vated in response to constitutive MEK/MAPK mitogenic signaling. Genes Dev 1998;12:3008-19.

    Albino AP, Nanus DM, Mentle IR, et al. Analysis of ras oncogenes in malignant melanoma and precursor lesions: correla-tion of point mutations with differentia-tion phenotype. Oncogene 1989;4:1363-74.

    Davies H, Bignell GR, Cox C, et al. Mutations of the BRAF gene in human cancer. Nature 2002;417:949-54.

    5.

    6.

    7.

    8.

    9.

    10.

    11.

    12.

    13.

    14.

    15.

    16.

    17.

    18.

    19.

    Omholt K, Platz A, Kanter L, Ring-borg U, Hansson J. NRAS and BRAF mutations arise early during melanoma pathogenesis and are preserved through-out tumor progression. Clin Cancer Res 2003;9:6483-8.

    Pollock PM, Harper UL, Hansen KS, et al. High frequency of BRAF mutations in nevi. Nat Genet 2003;33:19-20.

    Patton EE, Widlund HR, Kutok JL, et al. BRAF mutations are sufficient to pro-mote nevi formation and cooperate with p53 in the genesis of melanoma. Curr Biol 2005;15:249-54.

    Michaloglou C, Vredeveld LC, Soen-gas MS, et al. BRAFE600-associated se-nescence-like cell cycle arrest of human naevi. Nature 2005;436:720-4.

    Eskandarpour M, Kiaii S, Zhu C, Cas-tro J, Sakko AJ, Hansson J. Suppression of oncogenic NRAS by RNA interference in-duces apoptosis of human melanoma cells. Int J Cancer 2005;115:65-73.

    Hingorani SR, Jacobetz MA, Robert-son GP, Herlyn M, Tuveson DA. Suppres-sion of BRAF(V599E) in human melano-ma abrogates transformation. Cancer Res 2003;63:5198-202.

    Hoeflich KP, Gray DC, Eby MT, et al. Oncogenic BRAF is required for tumor growth and maintenance in melanoma models. Cancer Res 2006;66:999-1006.

    Lyons JF, Wilhelm S, Hibner B, Bollag G. Discovery of a novel Raf kinase inhibi-tor. Endocr Relat Cancer 2001;8:219-25.

    Solit DB, Garraway LA, Pratilas CA, et al. BRAF mutation predicts sensitivity to MEK inhibition. Nature 2006;439:358-62.

    Kamb A, Gruis NA, Weaver-Feldhaus J, et al. A cell cycle regulator potentially in-volved in genesis of many tumor types. Science 1994;264:436-40.

    Nobori T, Miura K, Wu DJ, Lois A, Takabayashi K, Carson DA. Deletions of the cyclin-dependent kinase-4 inhibitor gene in multiple human cancers. Nature 1994;368:753-6.

    Flores JF, Walker GJ, Glendening JM, et al. Loss of the p16INK4a and p15INK4b genes, as well as neighboring 9p21 mark-ers, in sporadic melanoma. Cancer Res 1996;56:5023-32.

    Wu H, Goel V, Haluska FG. PTEN sig-naling pathways in melanoma. Oncogene 2003;22:3113-22.

    Li J, Yen C, Liaw D, et al. PTEN, a pu-tative protein tyrosine phosphatase gene mutated in human brain, breast, and pros-tate cancer. Science 1997;275:1943-7.

    Steck PA, Pershouse MA, Jasser SA, et al. Identification of a candidate tumour suppressor gene, MMAC1, at chromosome 10q23.3 that is mutated in multiple ad-vanced cancers. Nat Genet 1997;15:356-62.

    You MJ, Castrillon DH, Bastian BC, et al. Genetic analysis of Pten and Ink4a/Arf interactions in the suppression of tumori-genesis in mice. Proc Natl Acad Sci U S A 2002;99:1455-60.

    20.

    21.

    22.

    23.

    24.

    25.

    26.

    27.

    28.

    29.

    30.

    31.

    32.

    33.

    34.

    35.

    Kamb A, Shattuck-Eidens D, Eeles R, et al. Analysis of the p16 gene (CDKN2) as a candidate for the chromosome 9p mela-noma susceptibility locus. Nat Genet 1994;8:23-6.

    Hussussian CJ, Struewing JP, Gold-stein AM, et al. Germline p16 mutations in familial melanoma. Nat Genet 1994;8:15-21.

    Pollock PM, Trent JM. The genetics of cutaneous melanoma. Clin Lab Med 2000;20:667-90.

    Sharpless E, Chin L. The INK4a/ARF locus and melanoma. Oncogene 2003;22:3092-8.

    Serrano M, Lee H, Chin L, Cordon-Cardo C, Beach D, DePinho RA. Role of the INK4a locus in tumor suppression and cell mortality. Cell 1996;85:27-37.

    Chin L, Pomerantz J, Polsky D, et al. Cooperative effects of INK4a and ras in melanoma susceptibility in vivo. Genes Dev 1997;11:2822-34.

    Zuo L, Weger J, Yang Q, et al. Germ-line mutations in the p16INK4a binding domain of CDK4 in familial melanoma. Nat Genet 1996;12:97-9.

    Sotillo R, Garcia JF, Ortega S, et al. Invasive melanoma in Cdk4-targeted mice. Proc Natl Acad Sci U S A 2001;98:13312-7.

    Sauter ER, Yeo UC, von Stemm A, et al. Cyclin D1 is a candidate oncogene in cutaneous melanoma. Cancer Res 2002;62:3200-6.

    Pomerantz J, Schreiber-Agus N, Liege-ois NJ, et al. The Ink4a tumor suppressor gene product, p19Arf, interacts with MDM2 and neutralizes MDM2s inhibition of p53. Cell 1998;92:713-23.

    Harris SL, Levine AJ. The p53 path-way: positive and negative feedback loops. Oncogene 2005;24:2899-908.

    Sharpless NE, Ramsey MR, Balasub-ramanian P, Castrillon DH, DePinho RA. The differential impact of p16(INK4a) or p19(ARF) deficiency on cell growth and tumorigenesis. Oncogene 2004;23:379-85.

    Kamijo T, Zindy F, Roussel MF, et al. Tumor suppression at the mouse INK4a locus mediated by the alternative reading frame product p19ARF. Cell 1997;91:649-59.

    Recio JA, Noonan FP, Takayama H, et al. Ink4a/arf deficiency promotes ultravi-olet radiation-induced melanomagenesis. Cancer Res 2002;62:6724-30.

    Guldberg P, Thor Straten P, Birck A, Ahrenkiel V, Kirkin AF, Zeuthen J. Disrup-tion of the MMAC1/PTEN gene by dele-tion or mutation is a frequent event in malignant melanoma. Cancer Res 1997;57:3660-3.

    Cantley LC, Neel BG. New insights into tumor suppression: PTEN suppresses tumor formation by restraining the phos-phoinositide 3-kinase/AKT pathway. Proc Natl Acad Sci U S A 1999;96:4240-5.

    Stahl JM, Cheung M, Sharma A, Trive-di NR, Shanmugam S, Robertson GP.

    36.

    37.

    38.

    39.

    40.

    41.

    42.

    43.

    44.

    45.

    46.

    47.

    48.

    49.

    50.

    51.

    52.

    The New England Journal of Medicine Downloaded from nejm.org on March 17, 2015. For personal use only. No other uses without permission.

    Copyright 2006 Massachusetts Medical Society. All rights reserved.

  • T h e n e w e ng l a nd j o u r na l o f m e dic i n e

    n engl j med 355;1 www.nejm.org july 6, 200664

    Loss of PTEN promotes tumor develop-ment in malignant melanoma. Cancer Res 2003;63:2881-90.

    Stahl JM, Sharma A, Cheung M, et al. Deregulated Akt3 activity promotes de-velopment of malignant melanoma. Can-cer Res 2004;64:7002-10.

    Hodgkinson CA, Moore KJ, Nakaya-ma A, et al. Mutations at the mouse mi-crophthalmia locus are associated with defects in a gene encoding a novel basic-helix-loop-helix-zipper protein. Cell 1993;74:395-404.

    Nishimura EK, Granter SR, Fisher DE. Mechanisms of hair graying: incomplete melanocyte stem cell maintenance in the niche. Science 2005;307:720-4.

    Widlund HR, Fisher DE. Microphtha-lamia-associated transcription factor: a critical regulator of pigment cell devel-opment and survival. Oncogene 2003;22:3035-41.

    Lerner AB, Shiohara T, Boissy RE, Ja-cobson KA, Lamoreux ML, Moellmann GE. A mouse model for vitiligo. J Invest Der-matol 1986;87:299-304.

    Steingrimsson E, Moore KJ, Lamor-eux ML, et al. Molecular basis of mouse microphthalmia (mi) mutations helps ex-plain their developmental and phenotypic consequences. Nat Genet 1994;8:256-63.

    McGill GG, Horstmann M, Widlund HR, et al. Bcl2 regulation by the melano-cyte master regulator Mitf modulates lin-eage survival and melanoma cell viability. Cell 2002;109:707-18.

    Banerjee D. Genasense (Genta Inc). Curr Opin Investig Drugs 2001;2:574-80.

    Goding CR. Mitf from neural crest to melanoma: signal transduction and tran-scription in the melanocyte lineage. Genes Dev 2000;14:1712-28.

    Du J, Miller AJ, Widlund HR, Horst-mann MA, Ramaswamy S, Fisher DE. MLANA/MART1 and SILV/PMEL17/GP100 are transcriptionally regulated by MITF in melanocytes and melanoma. Am J Pathol 2003;163:333-43.

    Baxter LL, Pavan WJ. Pmel17 expres-sion is Mitf-dependent and reveals cranial melanoblast migration during murine de-velopment. Gene Expr Patterns 2003;3:703-7.

    Loercher AE, Tank EMH, Delston RB, Harbour JW. MITF links differentiation with cell cycle arrest in melanocytes by transcriptional activation of INK4A. J Cell Biol 2005;168:35-40.

    Hofbauer GF, Kamarashev J, Geertsen R, Boni R, Dummer R. Melan A/MART-1 immunoreactivity in formalin-fixed par-affin-embedded primary and metastatic melanoma: frequency and distribution. Melanoma Res 1998;8:337-43.

    Salti GI, Manougian T, Farolan M, Shilkaitis A, Majumdar D, Das Gupta TK. Micropthalmia transcription factor: a new prognostic marker in intermediate-thick-

    53.

    54.

    55.

    56.

    57.

    58.

    59.

    60.

    61.

    62.

    63.

    64.

    65.

    66.

    ness cutaneous malignant melanoma. Can-cer Res 2000;60:5012-6.

    Seiter S, Monsurro V, Nielsen MB, et al. Frequency of MART-1/MelanA and gp100/PMel17-specific T cells in tumor metasta-ses and cultured tumor-infiltrating lym-phocytes. J Immunother 2002;25:252-63.

    Takeuchi H, Kuo C, Morton DL, Wang HJ, Hoon DS. Expression of differentia-tion melanoma-associated antigen genes is associated with favorable disease out-come in advanced-stage melanomas. Can-cer Res 2003;63:441-8.

    Miller AJ, Du J, Rowan S, Hershey CL, Widlund HR, Fisher DE. Transcriptional regulation of the melanoma prognostic marker melastatin (TRPM1) by MITF in melanocytes and melanoma. Cancer Res 2004;64:509-16.

    Duncan LM, Deeds J, Hunter J, et al. Down-regulation of the novel gene mela-statin correlates with potential for mela-noma metastasis. Cancer Res 1998;58:1515-20.

    King R, Weilbaecher KN, McGill G, Cooley E, Mihm M, Fisher DE. Microph-thalmia transcription factor: a sensitive and specific melanocyte marker for mela-noma diagnosis. Am J Pathol 1999;155:731-8.

    Miettinen M, Fernandez M, Franssila K, Gatalica Z, Lasota J, Sarlomo-Rikala M. Microphthalmia transcription factor in the immunohistochemical diagnosis of meta-static melanoma: comparison with four other melanoma markers. Am J Surg Pathol 2001;25:205-11.

    Granter SR, Weilbaecher KN, Quigley C, Fisher DE. Role for microphthalmia transcription factor in the diagnosis of metastatic malignant melanoma. Appl Im-munohistochem Mol Morphol 2002;10:47-51.

    Garraway LA, Widlund HR, Rubin MA, et al. Integrative genomic analyses iden-tify MITF as a lineage survival oncogene amplified in malignant melanoma. Na-ture 2005;436:117-22.

    Haass NK, Smalley KS, Li L, Herlyn M. Adhesion, migration and communication in melanocytes and melanoma. Pigment Cell Res 2005;18:150-9.

    Balch CM, Soong SJ, Gershenwald JE, et al. Prognostic factors analysis of 17,600 melanoma patients: validation of the American Joint Committee on Cancer melanoma staging system. J Clin Oncol 2001;19:3622-34.

    Johnson JP. Cell adhesion molecules in the development and progression of malignant melanoma. Cancer Metastasis Rev 1999;18:345-57.

    Bienz M. Beta-catenin: a pivot between cell adhesion and Wnt signalling. Curr Biol 2005;15:R64-R67.

    Gottardi CJ, Gumbiner BM. Adhesion signaling: how beta-catenin interacts with its partners. Curr Biol 2001;11:R792-R794.

    67.

    68.

    69.

    70.

    71.

    72.

    73.

    74.

    75.

    76.

    77.

    78.

    79.

    Brembeck FH, Schwarz-Romond T, Bakkers J, Wilhelm S, Hammerschmidt M, Birchmeier W. Essential role of BCL9-2 in the switch between beta-catenins ad-hesive and transcriptional functions. Genes Dev 2004;18:2225-30.

    Rubinfeld B, Robbins P, El-Gamil M, Albert I, Porfiri E, Polakis P. Stabilization of beta-catenin by genetic defects in mel-anoma cell lines. Science 1997;275:1790-2.

    Cowley GP, Smith ME. Cadherin ex-pression in melanocytic naevi and malig-nant melanomas. J Pathol 1996;179:183-7.

    Rimm DL, Caca K, Hu G, Harrison FB, Fearon ER. Frequent nuclear/cytoplas-mic localization of beta-catenin without exon 3 mutations in malignant melano-ma. Am J Pathol 1999;154:325-9.

    Sanders DS, Blessing K, Hassan GA, Bruton R, Marsden JR, Jankowski J. Al-terations in cadherin and catenin expres-sion during the biological progression of melanocytic tumours. Mol Pathol 1999;52:151-7.

    Widlund HR, Horstmann MA, Price ER, et al. Beta-catenin-induced melanoma growth requires the downstream target Microphthalmia-associated transcription factor. J Cell Biol 2002;158:1079-87.

    Shtutman M, Zhurinsky J, Simcha I, et al. The cyclin D1 gene is a target of the beta-catenin/LEF-1 pathway. Proc Natl Acad Sci U S A 1999;96:5522-7.

    Hsu M, Andl T, Li G, Meinkoth JL, Herlyn M. Cadherin repertoire determines partner-specific gap junctional communi-cation during melanoma progression. J Cell Sci 2000;113:1535-42.

    Valyi-Nagy IT, Hirka G, Jensen PJ, Shih IM, Juhasz I, Herlyn M. Undifferenti-ated keratinocytes control growth, mor-phology, and antigen expression of normal melanocytes through cell-cell contact. Lab Invest 1993;69:152-9.

    Danen EH, de Vries TJ, Morandini R, Ghanem GG, Ruiter DJ, van Muijen GN. E-cadherin expression in human melano-ma. Melanoma Res 1996;6:127-31.

    Hsu MY, Wheelock MJ, Johnson KR, Herlyn M. Shifts in cadherin profiles be-tween human normal melanocytes and melanomas. J Investig Dermatol Symp Proc 1996;1:188-94.

    Scott RA, Lauweryns B, Snead DM, Haynes RJ, Mahida Y, Dua HS. E-cadherin distribution and epithelial basement mem-brane characteristics of the normal human conjunctiva and cornea. Eye 1997;11:607-12.

    Gottardi CJ, Wong E, Gumbiner BM. E-cadherin suppresses cellular transfor-mation by inhibiting beta-catenin signal-ing in an adhesion-independent manner. J Cell Biol 2001;153:1049-60.

    Qi J, Chen N, Wang J, Siu CH. Trans-endothelial migration of melanoma cells involves N-cadherin-mediated adhesion and

    80.

    81.

    82.

    83.

    84.

    85.

    86.

    87.

    88.

    89.

    90.

    91.

    92.

    93.

    The New England Journal of Medicine Downloaded from nejm.org on March 17, 2015. 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 65

    activation of the beta-catenin signaling pathway. Mol Biol Cell 2005;16:4386-97.

    Li G, Satyamoorthy K, Herlyn M. N-cadherin-mediated intercellular inter-actions promote survival and migration of melanoma cells. Cancer Res 2001;61:3819-25.

    Kuphal S, Bauer R, Bosserhoff AK. In-tegrin signaling in malignant melanoma. Cancer Metastasis Rev 2005;24:195-222.

    Danen EH, Ten Berge PJ, Van Muijen GN, Van t Hof-Grootenboer AE, Brocker EB, Ruiter DJ. Emergence of alpha 5 beta 1 fibronectin- and alpha v beta 3 vitronec-tin-receptor expression in melanocytic tu-mour progression. Histopathology 1994;24:249-56.

    Brooks PC, Stromblad S, Sanders LC, et al. Localization of matrix metallopro-teinase MMP-2 to the surface of invasive cells by interaction with integrin alpha v beta 3. Cell 1996;85:683-93.

    Felding-Habermann B, Fransvea E, OToole TE, Manzuk L, Faha B, Hensler M. Involvement of tumor cell integrin al-

    94.

    95.

    96.

    97.

    98.

    pha v beta 3 in hematogenous metastasis of human melanoma cells. Clin Exp Metas-tasis 2002;19:427-36.

    Hofmann UB, Westphal JR, Waas ET, Becker JC, Ruiter DJ, van Muijen GN. Co-expression of integrin alpha(v)beta3 and matrix metalloproteinase-2 (MMP-2) co-incides with MMP-2 activation: correla-tion with melanoma progression. J Invest Dermatol 2000;115:625-32.

    Petitclerc E, Stromblad S, von Schal-scha TL, et al. Integrin alpha(v)beta3 pro-motes M21 melanoma growth in human skin by regulating tumor cell survival. Cancer Res 1999;59:2724-30.

    Li X, Regezi J, Ross FP, et al. Integrin alphavbeta3 mediates K1735 murine mel-anoma cell motility in vivo and in vitro. J Cell Sci 2001;114:2665-72.

    Dechantsreiter MA, Planker E, Matha B, et al. N-methylated cyclic RGD peptides as highly active and selective alpha(V)beta(3) integrin antagonists. J Med Chem 1999;42:3033-40.

    Tsao H, Zhang X, Fowlkes K, Haluska

    99.

    100.

    101.

    102.

    103.

    FG. Relative reciprocity of NRAS and PTEN/MMAC1 alterations in cutaneous melanoma cell lines. Cancer Res 2000;60:1800-4.

    Daniotti M, Oggionni M, Ranzani T, et al. BRAF alterations are associated with complex mutational profiles in ma-lignant melanoma. Oncogene 2004;23:5968-77.

    Tsao H, Goel V, Wu H, Yang G, Haluska FG. Genetic interaction between NRAS and BRAF mutations and PTEN/MMAC1 inactivation in melanoma. J Invest Dermatol 2004;122:337-41.

    Maldonado JL, Fridlyand J, Patel H, et al. Determinants of BRAF mutations in primary melanomas. J Natl Cancer Inst 2003;95:1878-90.

    Chudnovsky Y, Adams AE, Robbins PB, Lin Q, Khavari PA. Use of human tis-sue to assess the oncogenic activity of melanoma-associated mutations. Nat Gen-et 2005;37:745-9.Copyright 2006 Massachusetts Medical Society.

    104.

    105.

    106.

    107.

    JOURNAL EDITORIAL FELLOWThe Journals 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.

    The New England Journal of Medicine Downloaded from nejm.org on March 17, 2015. For personal use only. No other uses without permission.

    Copyright 2006 Massachusetts Medical Society. All rights reserved.