Anda di halaman 1dari 7

Seminars in Cancer Biology 14 (2004) 63–69

Targets for molecular therapy of skin cancer


Cheryl L. Green b , Paul A. Khavari a,b,∗
a VA Palo Alto Healthcare System, Palo Alto, CA 94304, USA
b Program in Epithelial Biology, Stanford University School of Medicine, 269 Campus Drive, Room 2145, Stanford, CA 94305, USA

Abstract

Cancers of the skin encompass the first and second most common neoplasms in the United States, epidermal basal cell carcinoma (BCC)
and squamous cell carcinoma (SCC), respectively, as well as the melanocytic malignancy, malignant melanoma (MM). Recently identified
alterations in the function of specific genes in these cancers provide new potential therapeutic targets. These alterations affect conserved
regulators of cellular proliferation and viability, including the Sonic Hedgehog, Ras/Raf, ARF/p53, p16INK4A /CDK4/Rb and NF-␬B
pathways. New modalities designed to target these specific proteins may represent promising approaches to therapy of human skin cancers.
Published by Elsevier Ltd.

Keywords: Skin; Cancer; Melanoma; Squamous cell carcinoma; Basal cell carcinoma

1. Introduction over a third of the cancers occur on the face. Histologi-


cally, BCCs lack precursor lesions and can be subdivided
Cancers of the skin arise predominantly from cells located into a number of sub-types, including superficial, nodu-
within the outermost compartment of the tissue, the epi- lar and aggressive-growth or morpheaform [3,4]. Classic
dermis. The majority of epidermal cells are keratinocytes, histologic features include palisaded nuclei at the tumor
epithelial cells that generate the cutaneous barrier to the periphery, clefting at tumor–tissue interfaces and formation
external environment. These cutaneous epithelial cells pop- of peri-tumor stromal tissue [5]. Based on morphologic
ulate both the follicular and interfollicular regions of the observations in tissue sections, it is believed that a sub-
skin and give rise to the structure of the epidermis as well as stantial proportion of all BCCs may arise from hair follicle
its appendages, namely hair and sweat glands. A number of keratinocytes. Clinically, BCCs are characterized by local
neoplasms originate from cutaneous epithelial cells [1], the invasion and contiguous spread. While reports of metastatic
most common of which are basal cell carcinoma (BCC) and BCC exist in the literature, it is widely recognized that BCC
squamous cell carcinoma (SCC). Interspersed among epithe- metastasis is an extremely rare event, in contrast to SCC
lial cells are pigment-producing melanocytes, which give [3–5]. Thus, BCC exerts its major damage through destruc-
rise to malignant melanoma (MM). Although widespread tion of local tissue and, when arising on the head as it com-
and increasing in incidence, BCC, SCC and MM have been monly does, BCC growth can lead to loss of vital structures
poorly understood at the level of molecular pathogenesis and mutilating disfigurement. As noted below, BCC occurs
until recently. Progress over the past decade has identified both sporadically and as part of heritable syndromes. Cur-
an array of molecular alterations in these neoplasias that rent therapies for BCC are non-specific and the majority of
may provide opportunities for new molecular therapeutics. approaches rely primarily on surgical removal of the tumor.

1.1. Basal cell carcinoma 1.2. Squamous cell carcinoma

BCC is the most common cancer in fair-skinned pop- SCC is the second most common cancer in the US
ulations, with an estimated yearly US incidence of ap- with approximately 250,000 new cases diagnosed annually.
proximately 1 million [2]. Lightly pigmented individuals In contrast to BCC, SCC is a biologically aggressive tu-
are most commonly affected on sun exposed surfaces and mor and may metastasize at frequencies reported between
1 and 12.5% [6–8]. Following local invasion and tissue
∗ Corresponding author. Tel.: +1-650-725-5266; fax: +1-650-723-8762. destruction, SCC commonly metasizes to lymph nodes.
E-mail address: khavari@CMGM.stanford.edu (P.A. Khavari). Unlike BCC, there is no compelling evidence for a hair

1044-579X/$ – see front matter. Published by Elsevier Ltd.


doi:10.1016/j.semcancer.2003.11.007
64 C.L. Green, P.A. Khavari / Seminars in Cancer Biology 14 (2004) 63–69

follicle-derived origin for SCC, and SCC is thus believed 2. Molecular defects in skin cancer
to largely represent a neoplasm of interfollicular epidermis.
Histologically, SCC displays a number of differences from Over the past decade, a number of genetic alterations
BCC, including a series of well-defined precursors, a lack have been characterized in BCC, SCC and MM (Fig. 1).
of well-demarcated tumor periphery and the presence of These alterations involve both gain-of-function changes in
features of epidermal differentiation [8]. SCC precursors oncogenes as well as loss-of-function of genes that act as
include actinic keratosis, which represents a focal dysplasia suppressors of tumor formation. The spectrum of changes
of epidermal keratinocytes, and SCC in situ, or Bowen’s differs among these tumor types, with BCC and MM per-
disease. The incidence of SCC is greatly increased in haps best understood currently.
patients receiving immunosuppressive therapy, a process
that may be enhanced by concomitant human papilloma 2.1. Sonic Hedgehog pathway
virus (HPV) infection. In individuals with intact immune
systems, however, clear evidence for a widespread role In addition to its sporadic occurrence, BCC is a feature
for HPV in SCC pathogenesis is lacking and ultraviolet of a number of inherited syndromes, including the nevoid
(UV) radiation-mediated mutagenesis is believed to be the basal cell carcinoma syndrome (NBCCS, or Gorlin’s syn-
major environmental factor promoting this cancer. A num- drome), Bazex syndrome and Rombo syndrome [5]. The
ber of heritable conditions are associated with increased former was instrumental in the identification of mutations
incidences of SCC, including xeroderma pigmentosum, in the PATCHED1 (PTCH1) gene, an important component
recessive dystrophic epidermolysis bullosa, dyskeratosis of the Sonic Hedgehog (SHH) signal transduction pathway,
congenital, oculocutaneous albinism and epidermodysplasia in both NBCCS and sporadic BCC [12,13]. The PTCH1
verruciformis [8]. In contrast to BCC, however, the genetic gene encodes a transmembrane protein that binds SHH and
defects in these syndromes are not specific for SCC. Thus, inhibits SHH pathway action by blocking the function of
beyond underscoring the roles of genomic integrity and another transmembrane protein, Smoothened (SMO), a G
chronic wounding injury in carcinogenesis, their underlying protein-coupled receptor. SMO signals to GLI family tran-
defects are believed to shed little light on the precise medi- scription factors, which regulate genes involved in tissue
ators of its pathogenesis. As is the case for BCC, there are morphogenesis. Subsequent studies have shown that alter-
no specifically targeted therapies for SCC and the primary ations at a number of points in the SHH pathway in addition
therapeutic modality is surgery. to PTCH1 loss-of-function are relevant to BCC, including
gain-of-function in SHH, SMO and GLI proteins. In spo-
1.3. Melanoma radic BCC, pathogenic mutations have been found in genes
encoding SHH, SMO and PTCH1 [12–15]. In epidermis,
In addition to keratinocytes, the epidermis contains a SHH pathway activation promotes proliferation and renders
number of other cell types, among which are melanocytes. cells resistant to stimuli promoting their cell cycle exit and
Derived embryologically from the neural crest, melanocytes terminal differentiation [16]. In mice, overexpression of
synthesize the melanin pigment that provides cells of the SHH, active SMO, GLI1 or GLI2 promotes development
skin with photoprotection from mutagenic UV rays. MM is of BCC-like tumors [14,15,17,18]. The potency of SHH
less common than either BCC or SCC, with 87,000 cases pathway activation in inducing hallmark BCC features in
estimated in the US for 2002 [9]. Like BCC and SCC, the human tissue was confirmed using human skin regener-
incidence of MM appears to be increasing, with predicted ated on immune deficient mice, where SHH overexpression
lifetime risk in the US rising to 1:75 for those born in 2000 alone triggered cardinal features of BCC within 3 weeks
[9]. Although less common, MM accounts for a greater [19]. Thus, compelling genetic and functional data points
number of deaths than BCC and SCC combined. The risk to induction of the SHH pathway as a major factor in BCC
factors for MM include light pigmentation, UV exposure, a and suggests that its inhibition may represent a promising
tendency to sunburn and multiple nevi. Regarding the latter, therapeutic opportunity (Fig. 1 and Table 1).
atypical nevi may represent both a marker and a precursor
for MM. Although MM may arise in a pre-existing nevus, it 2.2. p16INK4A , p14ARF and CDK4
may also arise de novo. While there are a number of variants
of MM, tumor depth appears to be among the most potent The CDKN2A locus on 9p21 is widely mutated in a variety
prognostic factors, with deeper primary tumors associated of human cancers [20,21]. This locus is commonly mutated
with a less favorable prognosis [10]. Families with increased in both sporadic and familial melanoma and encodes both
incidence of MM exist and often display large numbers a cyclin-dependent kinase (CDK) inhibitor, p16INK4A , and
of atypical nevi in affected members. Because of the poor a p53 regulator that is translated via an alternative reading
prognosis of metastatic MM and the curative nature of MM frame (ARF) [22,23]. p16INK4A acts to inhibit cell cycle pro-
removal when the cancer is caught prior to metastasis, clini- gression in the G1 phase by binding and inhibiting CDK4/6
cal efforts have been focused on early detection and surgical kinases. CDK4/6 phosphorylate Rb proteins to remove the
excision [11]. mid-G1 Rb block to cell cycle progression [24]. Loss of
C.L. Green, P.A. Khavari / Seminars in Cancer Biology 14 (2004) 63–69 65

Fig. 1. Schema of a selected of proteins altered in skin carcinogensis. Color denotes proteins for which defined mutations have been confirmed in specific
skin cancer types: gain-of-function is shown in green, loss in red. The approximate percentage of primary tumors with alterations indicated are noted
in parentheses, however, in some cases these figures are either controversial (i.e., RAS in SCC) or not clearly quantitated (i.e., CDK4 in MM). For
presentation, proteins have been diagrammed in pathways but this simplification does not reflect either confirmed epistatic relationships in these cancers or
the likely complexity in actual tumor tissues. CyD: D-type cyclins, MM: malignant melanoma, SCC: squamous cell carcinoma, BCC: basal cell carcinoma.

p16INK4A leads to deregulated CDK4/6 action and promo- in MM commonly abrogate function of both p16INK4A and
tion of cell division. An additional subset of patients with p14ARF , lesions that only affect one or the other have also
familial MM display mutations in CDK4 that render the ki- been reported, consistent with a role for both proteins in in-
nase insensitive to p16INK4A inhibition [25], indicating that hibiting metanocytic neoplasia [29–32]. Extensive work in
multiple mechanisms to bypass the Rb block are operative in animal models of MM has demonstrated that impairing the
MM. In addition to MM, CDK4 has been implicated in SCC function of p16INK4A and ARF along with activating Ras sig-
development by the recent demonstration of its overexpres- naling can facilitate the development of this tumor [33–36].
sion in this tumor and the fact that it is sufficient to induce Data on the sufficiency of altering these genes in human tis-
this neoplasm in human epidermis when it is co-expressed sue to induce MM, however, has not yet been published. Ef-
with oncogenic Ras [26]. ARF (p14ARF in humans, p19ARF forts to preserve the function of both the Rb (p16INK4A and
in mice) acts to stabilize p53 by inhibiting MDM2-mediated CDK4) and p53 (ARF) pathways, then, may be of utility in
degradation and also appears to induce anti-proliferative the prevention and treatment of MM and, possibly, of SCC.
genes on its own [27,28]. While CDKN2A mutations found
2.3. Ras and Raf
Table 1 Ras GTPases regulate proliferation, angiogenesis, apop-
Potential molecular targets in skin cancer
tosis and cellular morphology and may represent the most
Protein Pathway Skin cancer widely altered oncoproteins in human neoplasia [37]. Ras
SHH SHH BCC exerts its effects via three main downstream pathways initi-
PTCH1 SHH BCC ated by binding of GTP-bound Ras to families of pathway
SMO SHH BCC effectors including the Raf kinases, type I phosphatidyl-
PKA SHH BCC inositol-3 kinases (PI3Ks) and Ral guanine nucleotide
GLI1/2 SHH BCC
Ras Ras SCC, BCC
exchange factors (RalGEF) [38]. The best-characterized
Raf Ras MM, SCC, BCC Ras effector pathway proceeds via a kinase cascade that
ERK Ras MM, SCC, BCC includes Raf, two mitogen activated protein kinase kinases
p53 p53 SCC, BCC (MAPKKs or MAP/ERK kinases (MEKs): MEK1 and
p14ARF p53 MM, SCC, BCC MEK2, also called MKK1 and MKK2), and two MAPKs
p16INK4A Rb MM, SCC
CDK4 Rb MM, SCC
(ERK1 and ERK1) [39]. ERK has been observed to be
IKK, I␬B␣ NF-␬B SCC constitutively active in MM cells and sequencing of RAS
NF-␬B subunits NF-␬B SCC and RAF genes in melanoma tissue and cell lines has iden-
SHH: Sonic Hedgehog, PTCH1: PATCHED1, SMO: smoothened, PKA: tified activating RAS mutations in approximately 10% and
protein kinase A, ERK: extracellular; IKK: I␬B kinase, CDK: cyclin- kinase-activating BRAF mutations in approximately 70%
dependent kinase. of MM [40–42]. Thus, Ras/Raf gain-of-function appears
66 C.L. Green, P.A. Khavari / Seminars in Cancer Biology 14 (2004) 63–69

characteristic of MM, however, the presence of mutations 2.5. NF-κB/Rel


that activate B-Raf in a majority of benign nevi indicates
that induction of the latter alone is not sufficient for tumori- NF-␬B/Rel proteins are conserved transcription factors
genesis of melanocytes. In addition, to melanoma, activation implicated in regulating inflammation, morphogenesis,
of Ras/Raf signaling has been implicated in both BCC and apoptosis, differentiation and proliferation. These proteins
SCC. In non-transformed epidermis, Ras and Raf inhibit contain a characteristic 300 amino acid Rel homology do-
differentiation, stimulate cell division and augment inte- main (RHD) involved in DNA binding and protein–protein
grin expression, changes all characteristic of SCC [43–45]. interactions [55,56]. NF-␬B can be activated by an upstream
The PDGFR␣ has been shown to be upregulated in BCC, cascade involving I␬B kinases (IKKs) and inhibited by I␬B
with associated activation of the Ras/MAPK pathway [46]. proteins [57,58]. In contrast to its oncogenic role in other
While activating mutations and amplification of RAS genes settings, NF-␬B may act as an important anti-proliferative
have been reported in a minority of SCC [47], such genetic safeguard in stratified epithelium because its blockade facil-
changes are not required for the increased levels of active itates hyper-proliferation in both murine [59,60] and human
GTP-bound Ras observed in the majority of spontaneous epidermis [60]. Consistent with this, mice with inhibited
human SCC tissue [48]. This suggests the possible exis- epidermal NF-␬B function display increased susceptibility
tence of an analogous situation to that seen in breast cancer to induction of SCC [59]. Moreover, NF-␬B blockade in
where RAS mutations are rarely seen but where Ras is acti- concert with oncogenic Ras can transform human epidermis
vated due to a number of factors, including overexpression into a highly aggressive invasive neoplasia indistinguishable
of upstream receptor tyrosine kinases [49]. Inhibition of from SCC [48]. Recent analysis of a series of spontaneous
Ras/Raf cascade signaling, then, may constitute a poten- SCCs demonstrated evidence of NF-␬B blockade with I␬B␣
tially promising approach in the management of MM, BCC upregulation and cytoplasmic sequestration of NF-␬B RelA
and SCC. [48]. Therefore, while less well established than other al-
terations in common skin cancers, NF-␬B dysfunction may
2.4. p53 constitute a significant feature of SCC and thus represent a
potential target for therapy.
While p53 dysfunction has been reported in a wide array
of cancers, including BCC and MM, a number of studies sug-
3. Potential molecular therapeutics approaches for
gest a significant role for it in the pathogenesis of SCC. For
skin cancer
example, p53 plays a central role in the induction of epider-
mal cell apoptosis following UV radiation-mediated DNA The expanding knowledge of the pathogenesis of com-
damage [50]. Since UV serves as both a genotoxic initiator mon cutaneous malignancies offers the opportunities to
as well as tumor promoter, p53-dependent death of these develop new therapies directed at the specific molecules
cells is believed to function as a protective mechanism pre- altered in these cancers (Table 1). In addition to surgery,
venting SCC development. Over 90% of SCOs display p53 current therapeutic approaches include topical measures
mutations [51]. Consistent with a significant role of p53 in such as cryosurgery, immunomodulation with imiquimod,
SCC development, SCC precursors display p53 mutations in 5-Fluorouracil, photodynamic therapy and radiation [5].
over 70% of actinic keratoses [52]. While these data strongly While surgical excision remains the practical modality of
implicate p53 abnormalities in SCC development, the degree choice due to its definitive nature, the increasing incidence
of p53 impact in this setting is not yet firmly established. of skin cancers and the enormous resources involved in
For example, TP53 knockout mice display a surprising re- their diagnosis and treatment has propelled efforts to de-
sistance to cutaneous carcinogenesis induced by overexpres- velop more specific new strategies for their prevention and
sion of certain oncogenes [53]. However, these mice appear therapy. Because early treatment, especially in the case
more susceptible to SCC induction by UV, a likely more of MM and SCC, is essential to reducing disease mortal-
biologically relevant stimulus [54]. In short-term studies ity and morbidity, potent topical agents for skin-localized
in human skin tissue, in contrast, mutant p53 protein itself cancers and their precursors are of especial interest. Cur-
fails to promote invasive neoplasia when co-expressed with rent challenges facing efforts towards this goal include
oncogenic Ras, suggesting that its anti-neoplastic effects (a) development of effective, specific agents targeted at
operate through protecting genomic integrity rather through molecules of interest and (b) transcutaneous delivery of
direct promotion of oncogenesis. These findings regarding these therapeutics across the formidable skin permeability
the functional actions of p53 in SCC are complicated by the barrier.
experimental approaches to alter p53 function, including
differences between genetic deletion versus overexpression 3.1. Gene transfer
of well-characterized mutant p53 proteins. Further studies
are required to address these issues but preserving p53 The field of cutaneous gene transfer has seen significant
function remains an attractive possibility for the prevention recent progress in the field of monogenic skin disorders and
of SCC. an array of genetic approaches may be envisioned to either
C.L. Green, P.A. Khavari / Seminars in Cancer Biology 14 (2004) 63–69 67

interfere with aberrant signaling or restore defective tumor molecules into the skin. Among such strategies are protein
suppressor genes [61]. Approaches of interest could restore transduction [72–74], which relies on arginine-rich peptides
expression of proteins, such as p16INK4A , or disrupt pathway or peptidomimetics that replicate them to deliver macro-
function with dominant-negative interference, such as by molecules across cell and tissue barriers. This technology
expression of transdominant ERKs. Applying gene transfer has recently been used to reversibly link and deliver both
to the treatment of skin cancer, however, is hindered by peptides and previously non-penetrating drugs across hu-
the difficulty in delivering therapeutic genes to every cell man and mouse skin [75,76]. Combined with highly specific
within a tumor. To date, no approach supports such efficient new compounds designed to target proteins dysfunctional in
delivery to skin or any other tissue. Without such efficiency, human skin cancer, such new delivery approaches provide
even the most potent genetic sequences would be of limited promise for new molecular therapies for these diseases.
value in this setting.

3.2. Small molecule drugs 4. Conclusion

Development of specific pharmacotherapeutics for skin Insight into the pathogenesis of human skin cancers has
cancer is still in early stages, however, a number of poten- provided potential opportunities for preventive and thera-
tially useful proteins are being targeted in broader screening peutic intervention. These targets include components of
efforts. Small molecule regulators have been identified for evolutionarily conserved signaling cascades importance
a number of pathways relevant to skin cancer, including el- in tissue morphogenesis and cell fate, namely the SHH,
ements of the SHH, Ras/Raf, NF-␬B, p16INK4A /CDK4/Rb Ras/Raf, NF-␬B, p16INK4A /CDK4/Rb and ARF/p53 path-
and ARF/p53 pathways. For SHH, both agonists and antag- ways. To represent a significant advance in the treatment
onists have been identified that extend original observations of skin-localized non-metastatic tumors, however, new tar-
made with the original antagonist, cyclopamine [62]. Ef- geted therapies will have to display superior features to the
forts at Ras/Raf inhibition involve an array of molecules, current gold standard, surgical excision. Given the definitive
including inhibitors of enzymes that mediate Ras activation nature of the latter approach and its widespread availability,
as well as Raf and MEK kinase inhibitors [63,64]. Compo- this will not prove easy to accomplish in the short-term.
nents of the IKK1/NF-␬B cascade are also an active focus The evolution of new approaches to deliver agents that alter
of pharmacologic efforts directed at cancer and inflamma- the function of specific molecules involved in skin neo-
tory diseases [65,66]. A variety of CDK kinase inhibitors plasms, however, may provide new future strategies to the
have also been developed to support maintenance of the Rb management of these common human malignancies.
block [67–69]. In the case of p53, novel molecules such
as CP-31398 have been developed that stabilize p53 pro-
tein even in the absence of ARF [70]. A key problem with Acknowledgements
many of these efforts to block pleiotropic master regulator
proteins, such as Ras and NF-␬B, has been the undesired We thank N. Griffiths and P. Bernstein for administrative
complexity of altering their function throughout the body support. This work was supported by the USVA office of
when these agents are administered systemically. For exam- Research and Development and by NIH AR44012, AR43799
ple, the majority of efforts directed at NF-␬B are designed to P.A.K.
to block its function in malignancies of lymphoid and other
tissue [66,71]. Unfortunately, this may end up promoting
cutaneous carcinogenesis [48], a major problem in immune References
suppressed cancer patients. By virtue of its tractability to
[1] Owens DM, Watt FM. Contribution of stem cells and differentiated
local topical therapy that could avoid unwanted paradoxical
cells to epidermal tumours. Nat Rev Cancer 2003;3:444–51.
effects in differing tissues, the skin offers unique advantages [2] Miller DL, Weinstock MA. Nonmelanoma skin cancer in the United
in this regard. Nonetheless, significant obstacles confront States: incidence. J Am Acad Dermatol 1994;30:774–8.
current efforts to capitalize on recent mechanistic insight [3] Miller SJ. Biology of basal cell carcinoma (Part I). J Am Acad
into skin neoplasia by developing new molecular therapies. Dermatol 1991;24:1–13.
[4] Miller SJ. Biology of basal cell carcinoma (Part II). J Am Acad
Dermatol 1991;24:161–75.
3.3. Transcutaneous transporters [5] Carucci JA, Lefell DJ. Basal cell carcinoma, in Freedberg IM, et al.,
editors. Dermatology in General Medicine. McGraw-Hill Press, New
Topical delivery of new therapeutics across the epidermal York, 2003: 747–54.
permeability barrier to prevent or treat localized pre-cancers [6] Cherpelis BS, Marcusen C, Lang PG. Prognostic factors for metas-
tasis in squamous cell carcinoma of the skin. Dermatol Surg
or cancers of the skin presents a significant challenge given
2002;28:268–73.
the skin’s well known capacity to exclude foreign molecules. [7] Czarnecki D, Staples M, Mar A, Giles G, Meehan C. Metastases
The development of new strategies to accomplish this, how- from squamous cell carcinoma of the skin in southern Australia.
ever, may permit the transport of a variety of therapeutic Dermatology 1994;189:52–4.
68 C.L. Green, P.A. Khavari / Seminars in Cancer Biology 14 (2004) 63–69

[8] Grossman D, Lefell DJ. Squamous cell carcinoma, in Freedberg but not CDKN2A in a melanoma-neural system tumour syndrome
IM, et al., editors. Dermatology in General Medicine. McGraw-Hill family. Hum Mol Genet 2001;10:55–62.
Press, New York, 2003: 737–47 [32] Rizos H, Puig S, Badenas C, Malvehy J, Darmanian AP, Jimenez
[9] Langley RGB, Barnhill RL, Mihm MC, Fitzpatrick TB, Sober AJ. L, et al. A melanoma-associated germlme mutation in exon 1 beta
Cutaneous melanoma, in Freedberg IM, et al., editors. Dermatology inactivates p14ARF. Oncogene 2001;20:5543–7.
in General Medicine. McGraw-Hill Press, New York, 2003: 917–47. [33] Chin L, Pomerantz J, Polsky D, Jacobson M, Cohen C, Cordon-Carde
[10] Kanzler MH, Swetter SM. Malignant melanoma. J Am Acad Der- C, et al. Cooperative effects of INK4a and ras in melanoma suscep-
matol 2003;48:780–3. tibility in vivo. Genes Dev 1997;11:2822–34.
[11] Swetter SM. Dermatological perspectives of malignant melanoma. [34] Chin L, Tam A, Pomerantz J, Wong M, Holash J, Bardeesy N, et
Surg Clin North Am 2003;83:77–95. al. Essential role for oncogenic Ras in tumour maintenance. Nature
[12] Hahn H, Wicking C, Zaphiropoulous PG, Gailani MR, Shanley 1999;400:468–72.
S, Chidambaram A, et al. Mutations of the human homolog of [35] Bardeesy N, Wong KK, DePinho RA, Chin L. Animal models of
Drosophila patched in the nevoid basal cell carcinoma syndrome. melanoma: recent advances and future prospects. Adv Cancer Res
Cell 1996;85:841–51. 2000;79:123–56.
[13] Johnson RL, Rothman AL, Xie J, Goodrich LV, Bare JW, Bonifas [36] Kannan K, Sharpless NE, Xu J, O’Hagan RC, Bosenberg M, Chin
JM, et al. Human homolog of patched, a candidate gene for the basal L. Components of the Rb pathway are critical targets of UV muta-
cell nevus syndrome. Science 1996;272:1668–71. genesis in a murine melanoma model. Proc Natl Acad Sci U S A
[14] Xie J, Murone M, Luoh SM, Ryan A, Gu Q, Zhang C, et al. 2003;100:1221–5.
Activating Smoothened mutations in sporadic basal-cell carcinoma. [37] Frame S, Balmain A. Integration of positive and negative growth
Nature 1998;391:90–2. signals during ras pathway activation in vivo. Curr Opin Genet Dev
[15] Oro AE, Higgins KM, Hu Z, Bonifas JM, Epstein EH, Scott MP. 2000;10:106–13.
Basal cell carcinomas in mice overexpressing sonic hedgehog. Sci- [38] Shields JM, Pruitt K, McFall A, Shaub A, Der CJ. Understanding
ence 1997;276:817–21. Ras: ‘it ain’t over ‘til it’s over’. Trends Cell Biol 2000;10:147–54.
[16] Fan H, Khavari PA. Sonic hedgehog opposes epithelial cell cycle [39] Chang L, Karin M. Mammalian MAP kinase signalling cascades.
arrest. J Cell Biol 1999;147:71–6. Nature 2001;410:37–40.
[17] Nilsson M, Unden AB, Krause D, Malmqwist U, Raza K, Zaphi- [40] Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, et al.
ropoulos PG, et al. Induction of basal cell carcinomas and trichoep- Mutations of the BRAF gene in human cancer. Nature 2002;417:949–
itheliomas in mice overexpressing GLI-1. Proc Natl Acad Sci U S 54.
A 2000;97:3438–43. [41] Albino AP, Nanus DM, Mentle IR, Cordon-Cardo C, McNutt NS,
[18] Grachtchouk M, Mo R, Yu S, Zhang X, Sasaki H, Hui CC, et al. Bressler J, et al. Analysis of ras oncogenes in malignant melanoma
Basal cell carcinomas in mice overexpressing Gli2 in skin. Nat Genet and precursor lesions: correlation of point mutations with differen-
2000;24:216–7. tiation phenotype. Oncogene 1989;4:1363–74.
[19] Fan H, Oro AE, Scott MP, Khavari PA. Induction of basal cell carci- [42] Demunter A, Stas M, Degreef H, De WoIf-Peeters C, van den Oord
noma features in transgenic human skin expressing Sonic Hedgehog. JJ. Analysis of N- and K-ras mutations in the distinctive tumor pro-
Nat Med 1997;3:788–92. gression phases of melanoma. J Invest Dermatol 2001;117:1483–9.
[20] Sasaki S, Kitagawa Y, Sekido Y, Minna JD, Kuwano H, Yokota J, [43] Dajee M, Cai MT, Khavari PA. Epidermal Ras blockade demonstrates
et al. Molecular processes of chromosome 9p21 deletions in human spatially localized Ras promotion of proliferation and inhibition of
cancers. Oncogene 2003;22:3792–8. differentiation. Oncogene 2002;21:1527–38.
[21] Kamb A, Gruis NA, Weaver-Feldhaus J, Liu Q, Harshman K, Tavti- [44] Cal T, Nishida K, Hirano T, Khavari PA. Gabi and SHP-2 promote
gian SV, et al. A cell cycle regulator potentially involved in genesis Ras/MAPK regulation of epidermal growth and differentiation. J Cell
of many tumor types. Science 1994;264:436–40. Biol 2002;159:103–12.
[22] Sharpless E, Chin L. The lNK4a/ARF locus and melanoma. Onco- [45] Tarutani M, Cal T, Dajee M, Khavari PA. Inducible activation of ras
gene 2003;22:3092–8. and raf in adult epidermis. Cancer Res 2003;63:319–23.
[23] Gibbs P, Brady BM, Robinson WA. The genes and genetics of [46] Xie J, Aszterbaum M, Zhang X, Bonifas JM, Zachary C, Epstein E,
malignant melanoma. J Cutan Med Surg 2002;6:229–35. et al. A role of PDGFRaIpha in basal cell carcinoma proliferation.
[24] Sherr CJ, Roberts JM. CDK inhibitors: positive and negative regu- Proc Natl Acad Sci U S A 2001;98:9255–9.
lators of Gi-phase progression. Genes Dev 1999;13:1501–12. [47] Pierceall WE, Goldberg LH, Tainsky MA, Mukhopadhyay T, Anan-
[25] Zuo L, Weger J, Yang Q, Goldstein AM, Tucker MA, Walker GJ, et thaswamy HN. Ras gene mutation and amplification in human non-
al. Germlme mutations in the p1 6INK4a binding domain of CDK4 melanoma skin cancers. Mol Carcinog 1991;4:196–202.
in familial melanoma. Nat Genet 1996;12:97–9. [48] Dajee M, Lazarov M, Zhang JY, Cai T, Green CL, Russell AJ, et
[26] Lazarov M, Kubo Y, Cai T, Dajee M, Tarutani M, Lin Q, et al. al. NF-kappaB blockade and oncogenic Ras trigger invasive human
CDK4 coexpression with Ras generates malignant human epidermal epidermal neoplasia. Nature 2003;421:639–43.
tumorigenesis. Nat Med 2002;8:1105–14. [49] von Lintig FC, Dreilinger AD, Varki NM, Wallace AM, Casteel DE,
[27] Sherr CJ. The INK4a/ARF network in tumour suppression. Nat Rev Boss GR. Ras activation in human breast cancer. Breast Cancer Res
Mol Cell Biol 2001;2:731–7. Treat 2000;62:51–62.
[28] Kuo ML, Duncavage EJ, Mathew R, den Besten W, Pei D, Naeve D, [50] Ziegler A, Jonason AS, Leffell DJ, Simon JA, Sharma HW, Kim-
et al. Arf induces p53-dependent and -independent antiproliferative melman J, et al. Sunburn and p53 in the onset of skin cancer. Nature
genes. Cancer Res 2003;63:1046–53. 1994;372:773–6.
[29] Hussussian CJ, Struewing JP, Goldstein AM, Higgins PA, Ally DS, [51] Leffell DJ. The scientific basis of skin cancer. J Am Acad Dermatol
Sheahan MD, et al. Germlme p16 mutations in familial melanoma. 2000;42:18–22.
Nat Genet 1994;8:15–21. [52] Campbell C, Quinn AG, Ro YS, Angus B, Rees JL. p53 mutations are
[30] Gruis NA, van der Velden PA, Sandkuijil LA, Prins DE, common and early events that precede tumor invasion in squamous
Weaver-Feldhaus J, Kamb A, et al. Homozygotes for CDKN2 (p16) cell neoplasia of the skin. J Invest Dermatol 1993;100:746–8.
germlme mutation in Dutch familial melanoma kindreds. Nat Genet [53] Greenhalgh DA, Wang XJ, Donehower LA, Roop DR. Paradoxical
1995;10:351–3. tumor inhibitory effect of p53 loss in transgenic mice expressing
[31] Randerson-Moor JA, Harland M, Williams S, Cuthbert-Heavens D, epidermal targeted v-rasHa, v-fos, or human transforming growth
Sheridan E, Aveyard J, et al. A germlme deletion of p14(ARF) factor alpha. Cancer Res 1996;56:4413–23.
C.L. Green, P.A. Khavari / Seminars in Cancer Biology 14 (2004) 63–69 69

[54] Jiang W, Ananthaswamy HN, Muller HK, Kripke ML. p53 pro- [66] Orlowski RZ, Baldwin Jr AS. NF-kappaB as a therapeutic target in
tects against skin cancer induction by UV-B radiation. Oncogene cancer. Trends Mol Med 2002;8:385–9.
1999;18:4247–53. [67] Engler TA, Furness K, Malhotra S, Sanchez-Martinez C, Shih C,
[55] Jia S, Flores-Saaib RD, Courey AJ. The Dorsal Rel homology domain Xie W, et al. Novel, potent and selective cyclin D1ICDK4 in-
plays an active role in transcriptional regulation. Mol Cell Biol hibitors: [indolo[6,7-a]pyrrolo(3,4 c]carbazoles. Bioorg Med Chem
2002;22:5089–99. Lett 2003;13:2261–7.
[56] Lin L, DeMartino GN, Greene WC. Cotranslational dimerization [68] Honma T, Hayashi K, Aoyama T, Hashimoto N, Machida T, Fuka-
of the Rel homology domain of NF-kappaBl generates p50-pi OS sawa K, et al. Structure-based generation of a new class of potent
heterodimers and is required for effective p50 production. Embo J Cdk4 inhibitors: new de novo design strategy and library design. J
2000;19:4712–22. Med Chem 2001;44:4615–27.
[57] Ghosh S, Karin M. 2002. Missing pieces in the NF-kappaB puzzle. [69] Fry DW, Bedford DC, Harvey PH, Fritsch A, Keller PR, Wu Z,
Cell 2002;109(Suppl):881–96. et al. Cell cycle and biochemical effects of PD 0183812. A potent
[58] Dixit V, Mak TW. NF-kappaB signaling. Many roads lead to madrid. inhibitor of the cyclin D-dependent kinases CDK4 and CDK6. J Biol
Cell 2002;111:615–9. Chem 2001;276:16617–23.
[59] van Hogerlinden M, Rozell BL, Ahrlund-Richter L, Toftgard R. Squa- [70] Wang W, Takimoto R, Rastinejad F, EI-Deiry WS. Stabilization of
mous cell carcinomas and increased apoptosis in skin with inhibited p53 by CP-31 398 inhibits ubiquitination without altering phos-
ReVnuclear factor-kappaB signaling. Cancer Res 1999;59:3299–303. phorylation at serine 15 or 20 or MDM2 binding. Mol Cell Biol
[60] Seitz CS, Lin Q, Deng H, Khavari PA. Alterations in NF-␬B function 2003;23:2171–81.
in transgenic epithelial tissue demonstrate a growth inhibitory role [71] Waddick KG, Uckun FM. Innovative treatment programs against
for NF-␬B. Proc Natl Acad Sci U S A 1998;95:2307–12. cancer: II. Nuclear factor-kappaB (NF-kappaB) as a molecular target.
[61] Khavari PA, Rollman O, Vahlquist A. Cutaneous gene transfer for Biochem Pharmacol 1999;57:9–17.
skin and systemic diseases. J Intern Med 2002;252:1–10. [72] Prochiantz A. Messenger proteins: homeoproteins, TAT and others.
[62] Frank-Kamenetsky M, Zhang XM, Bottega S, Guicherit O, Wichterle Curr Opin Cell Biol 2000;12:400–6.
H, Dudek H, et al. Small-molecule modulators of Hedgehog signal- [73] Ford KG, Souberbielle BE, Darling D, Farzaneh F. Protein transduc-
ing: identification and characterization of Smoothened agonists and tion: an alternative to genetic intervention? Gene Ther 2001;8:1–4.
antagonists. J Biol 2002;1:10. [74] Wadia JS, Dowdy SF. Protein transduction technology. Curr Opin
[63] Cox AD, Der CJ. Ras family signaling: therapeutic targeting. Cancer Biotechnol 2002;13:52–6.
Biol Ther 2002;1:599–606. [75] Rothbard JB, Garlington S, Lin Q, Kirschberg T, Kreider E, Mc-
[64] Downward J. Targeting RAS signalling pathways in cancer therapy. Grane PL, et al. Conjugation of arginine oligomers to cyclosporin A
Nat Rev Cancer 2003;3:11–22. facilitates topical delivery and inhibition of inflammation. Nat Med
[65] Karin M, Cao Y, Greten FR, Li ZW. NF-kappaB in cancer: from 2000;6:1253–7.
innocent bystander to major culprit. Nature Rev Cancer 2002;2: [76] Robbins PB. Peptide delivery to tissues via reversibly linked protein
301–10. transduction sequences. Biotechniques 2002;33:190–4.

Anda mungkin juga menyukai