Current Cancer Therapy Reviews, 2009, 5, 37-44
37
Squamous Cell Carcinoma of the Skin: Current Strategies for Treatment and Prevention Michael A. Bachelora and David M. Owensa,b,* a
Departments of Dermatology and Pathology, College of Physicians & Surgeons, bColumbia University, New York, NY 10032, USA Abstract: Non-melanoma skin cancer (NMSC) is the most frequent malignancy in the United States, with over one million new cases reported annually. Approximately 80% of NMSC are basal cell carcinomas (BCC), while the remaining 20% of NMSC are squamous cell carcinomas (SCC). BCC and SCC commonly arise in regions of the skin subjected to chronic sun exposure implicating ultraviolet radiation (UV)-induced cellular damage as the primary causative agent. While surgical excision is an effective treatment of NMSC, strategies aimed at NMSC prevention have not been exploited clinically. The application of sunscreens has been the primary means of prevention by physically blocking the absorption of UV radiation. However, sunscreens have had limited success in decreasing NMSC incidence overall, necessitating more targeted strategies against UV damage. In this review we will discuss the relevance of recently identified UVinduced cellular changes, including induction of reactive oxygen species, immune modulation, and damage to mitochondrial DNA to serve as potential targets for the chemoprevention of NMSC. Finally, we will discuss the potential of genes required for the maintenance of epithelial stem cells in the skin as therapeutic targets for cutaneous cancer.
Key Words: Non melanoma skin cancer, ultraviolet radiation, reactive oxygen species, inflammation, stem cell, keratinocyte. INTRODUCTION Nonmelanoma skin cancer (NMSC) is the most common malignancy in fair-skinned populations. In the United States alone, it is estimated that 1.2 million new cases of NMSC are diagnosed each year, accounting for 40% of all new cancer cases [1,2]. In addition, another one million cases of NMSC are estimated to go unreported each year 1. The vast majority of NMSC are comprised of basal cell carcinoma (BCC) and squamous cell carcinoma (SCC), which arise primarily on sun-exposed regions of the body implicating ultraviolet (UV) radiation as the main etiological agent [3-5]. BCC and SCC comprise 80% and 16% of all skin cancers, respectively, with malignant melanomas comprising 4%. Based on their distinct histopathological features and expression patterns of differentiation lineage markers, SCC are thought to originate from the epidermis and BCC from the hair follicle [6] (Fig. 1). Skin cancer incidence has been increasing in recent years as the population ages and higher levels of UV radiation reach the surface of the Earth as a result of ozone depletion. Even minor changes in the ozone are thought to have a substantial impact on UV carcinogenesis as a 1% decrease in column ozone has been calculated to result in a 1.56% increase in carcinogenic UV radiation penetrance and a concomitant increase in NMSC by 2.7% [7]. Current prevention of NMSC involves limiting excessive exposure to UV, wearing protective clothing and the use of sunscreens. However,
*Address correspondence to this author at the Columbia University Medical Center, Herbert Irving Comprehensive Cancer Center, 1130 St. Nicholas Ave, Room 321A, New York, New York 10032, USA; Tel: (212) 851-4544; Fax: (212) 851-4540; E-mail:
[email protected]
1
American Cancer Society, Cancer Facts & Figures, 2008 1573-3947/09 $55.00+.00
these approaches have remained unsuccessful in impacting the overall incidence of NMSC. In light of the inability of primary prevention strategies to remain effective, and the high frequency of recurrence in patients diagnosed with BCC and SCC, more customized therapies may be necessary to adequately target these lesions. BCC are slow growing lesions that rarely metastasize, although these tumors can have significant morbidity due to local invasiveness. Contrastingly, SCC are highly invasive lesions with a much greater propensity for metastasis. [8,9]. The reported incidence of metastatic SCC varies ranging from 0.5 to 16%, and metastatic SCC account for 1200 deaths annually [10]. Metastatic SCC are difficult to control in spite of aggressive therapeutic intervention, including surgery, and/or radiation. Moreover, metastatic SCC patients also have a greater likelihood of developing additional skin lesions within five years of initial diagnosis [11,12]. Compounding this problem is the lack of reliable diagnostic predictors of SCC metastatic potential, which currently includes tumors size (larger than 2 cm), poor differentiation, and degree of invasiveness [13,14]. In addition to exposure to damaging sunlight, other factors also increase the risk for developing NMSC. For example, a high incidence of cutaneous SCC is associated with patients receiving post-operative immunosuppressive drugs following organ transplant. These transplant recipient patients also have a rate of metastasis incidence twice that of SCC patients that are not immunosuppressed [15]. An 8-10 fold increased risk for developing skin cancer and SCC, in particular, has also been reported in patients with chronic lymphocytic leukemia (CLL) where individuals lack competent innate and humoral immunity [16,17]. Finally, individuals with the autosomal-dominant disorder nevoid basal cell © 2009 Bentham Science Publishers Ltd.
38 Current Cancer Therapy Reviews, 2009, Vol. 5, No. 1
Bachelor and Owens
Fig. (1). UV-induced pathways associated with SCC formation in skin epithelium. Schematic of adult skin showing the multiple epidermal layers, pilosebaceous unit and dermis. SCC lesion is designated within the epidermis as mutant (red) cells. UVA and UVB have differential effects on the skin due in part to their penetration into various layers of the skin (depth indicated by red arrows). (A) UVA induced cellular changes arises from increases in ROS production. (B) UVB induced cellular changes implicated in SCC pathogenesis include: increases in COX-2 activity, production of inflammatory cytokines, mutations in mtDNA and signature mutations in the tumor suppressor TP53. Abbreviations: DP, dermal papilla; SG, sebaceous gland; HF, hair follicle, GL, granular layer; SC, spinous layer; BL, basal layer.
carcinoma syndrome (Gorlin-Goltz syndrome) present with large numbers of BCC that recur throughout life [18]. While typical surgical intervention may be effective and in some cases curative for these patients, these procedures are highly distressful when performed frequently. Recent interest in targeted-based therapy for the treatment of skin cancer has shed light on numerous signaling pathways that might provide new therapeutic targets. Cell signaling pathways involved in proliferation and viability are discussed in a recent review which highlights potential molecular targets specific to SCC and BCC or that are common in both tumor types [19]. The focus of this review will be to highlight UV-mediated cellular changes, particularly events mediated by reactive oxygen species (ROS) and proinflammatory molecules that could potentially be exploited therapeutically through a targeted approach to ameliorate the deleterious effects of UV exposure. We will also highlight recent work regarding epithelial stem cells in the skin and discuss their potential role as chemopreventive and therapeutic targets. CURRENT NMSC
THERAPEUTIC
APPROACHES
FOR
Surgery is the most common form of treatment for both BCC and SCC. The goal of surgical treatment is complete removal of the tumor while preserving normal surrounding
tissue [20]. Nonsurgical options are available and include radiation therapy, topical chemotherapeutics (5-fluorouracil), retinoids and photodynamic therapy. While nonsurgical options are used less frequently, they may be beneficial when surgery would produce disfiguring results or for patients unable to tolerate surgery [21,22]. Several agents have been used in chemoprevention clinical trials including retinoids (retinol or Vitamin A, isotretinoin or 13-cis-retinoic acid), -carotene and selenium. Retinoids are derivatives of Vitamin A and affect keratinocyte growth, differentiation, adhesion and immune modulation mediated through the binding of the nuclear retinoic acid receptors and retinoid C receptors [23]. -carotene and selenium both act as antioxidants to reduce UV-induced reactive oxygen species [24,25]. Chemoprevention trials involving selenium, -carotene, and isotretinoin showed that these compounds have no effect on decreasing NMSC incidence in patients with a history of SCC or BCC whereas patients receiving retinol had a 26% less risk of being diagnosed with SCC [26-28]. Treatment of moderate risk patients, having a history of more than 10 actinic keratoses (the premalignant precursor to SCC), but no more than two preexisting SCC or BCC, with retinol was effective in preventing new SCC but not BCC [28]. However, retinol and isotretinoin were ineffective in blocking the development of new NMSC in highrisk patients (history of four or more SCC or BCC) [29]. The therapeutic effects of retinoids in reducing NMSC in patients
Squamous Cell Carcinoma of the Skin
with actinic keratoses but not in patients with more progressed skin cancer suggest that these agents are more effective in the earlier stages of UV-induced skin carcinogenesis. Consistent with this idea, retinoids have proven effective in reducing the number of new actinic keratoses or NMSC in patients known to be at very high risk for developing skin cancer. Individuals with defects in DNA repair (Xeroderma Pigmentosa or XP patients) and organ transplant recipients receiving immunosuppressive had a 63% and 42% reduction in incidence, respectively [30,31]. While retinoids may be effective at treating these early lesions induced by UV light, no single agent has been identified that has had significant impact on more progressed stages of skin cancer. Definition of Ultraviolet Spectrum The UV component of sunlight is strongly implicated with both BCC and SCC pathogenesis and is capable of inducing pleiotropic cellular changes that can alter tissue homeostasis by impacting proliferation, differentiation, apoptosis and inflammation. The UV spectrum is divided into three regions based on wavelength, UVA (320-400nm), UVB (280-320nm) and UVC (200-280nm). Wavelengths up to 310nm are filtered by the ozone layer thereby preventing UVC and most of the UVB spectrum (~95%) from reaching the surface of the Earth. Therefore, 90-99% of the biologically relevant solar radiation reaching the surface of the Earth is comprised of UVA, with the remaining 1-10% composed of UVB [32]. When applied separately, both UVA and UVB can act as complete carcinogens in experimental mouse skin models; however, UVB is 1000-10,000 times more potent in inducing skin tumorigenesis than UVA [33,34]. Importantly, UVA and UVB induce different cellular changes including erythema, inflammation, signal transduction, and ROS production as a consequence of the ability of different energy wavelengths to penetrate distinct layers of the skin. While both UVA and UVB play a role in SCC formation, these unique properties may have clinical significance, as distinct chemopreventive strategies may be required for each component of UV light. At the tissue level, long-wave UVA penetrates deep into the dermis of the skin whereas short wave UVB only penetrates through the epidermis (Fig. 1). At the cellular level, UVB causes direct excitation of genomic DNA, inducing DNA adducts, strand breaks, DNA crosslinks and DNAprotein crosslinks [35]. If left unrepaired, these DNA adducts can lead to mutations upon DNA replication, the most common of which are detected in the tumor suppressor gene TP53 in sun-damaged skin [36-41]. In contrast, the deleterious effects due to chronic UVA exposure are thought to be a result of increased ROS [42]. UVA produces oxidative DNA damage more efficiently than UVB as ROS is generated upon absorption through cellular photosensitizers such as porphyrins and NADH [43-45]. Interestingly, it has recently been reported that UVA causes photoproducts similar to those induced by UVB albeit at much lower frequencies [46]. These adducts occur at unique sequences from that observed with UVB irradiation and suggest UVA-induced genetic damage may act independently from direct excitation of DNA.
Current Cancer Therapy Reviews, 2009, Vol. 5, No. 1
39
UV-Induced Signature Mutations Characteristic UV-induced genetic mutations have been detected in the TP53 tumor suppressor from chronically sundamaged skin, actinic keratoses and SCC, indicating that mutations in TP53 gene are an early event in UV-induced skin carcinogenesis [37,39,40,47]. TP53 is the most commonly mutated gene in human SCC with reports ranging from 35%-58% [36,48]. TP53 plays an important role in maintaining the genomic integrity of the cell by blocking DNA replication in response to UV-induced DNA damage. In both cultured cells and the epidermis of skin, exposure to UVB induces the expression of wild-type TP53. If the cell has sustained irreparable DNA damage due to UV radiation, the induction of p53 blocks cell cycle progression and causes cells to undergo programmed cell death, or apoptosis [49]. In response to high doses of UVB, sunburn cells can also be observed in the epidermis, initiated via a TP53-dependent pathway and resulting in the removal of damaged cells, thereby minimizing the risk of tumor development. This concept is illustrated in Trp53 -/- mice where increased sensitivity to UVB and fewer apoptotic cells are observed in the epidermis [47]. In addition, Trp53 -/- mice develop more aggressive tumors when compared to mice expressing functional p53 indicating the necessity for removal of damaged cells for the prevention of tumor development [50]. The use of sunscreens can be highly effective in the prevention of UV-induced TP53 mutations. A sunscreen absorbing in the UVB only or UVA and UVB spectra showed an 88% and 92% reduction, respectively in Trp53 mutation frequency [51]. The findings of this study were reproduced in a follow-up report using solar-simulated radiation (SSR), a combination of UVA and UVB mimicking normal environmental exposure, where an 80-100% decrease in the mutation frequency of Trp53 was reported using sunscreens that absorbed only UVB or both UVA and UVB [52]. More importantly, the reduction of Trp53 mutations correlated with a reduction in skin tumor indicating that p53 is a useful biological endpoint to indicate risk of UV-induced skin tumor formation. When non-sun exposed human skin was grafted onto immunocompromised mice and exposed to solarsimulated UV radiation, UV-induced TP53 mutations could be detected in all grafts while grafts treated with SPF-15 sunscreen blocked the induction of mutations in TP53 [49]. From a therapeutic perspective, these data underscore the value of primary prevention of UV-induced genetic damage in reducing the incidence of skin tumor development. Despite the ability of sunscreens in reducing TP53 mutations and decreasing SCC formation under experimental conditions, their effect in decreasing overall NMSC incidence has been limited. In fact, several clinical trials have reported no decrease in NMSC incidence with the use of sunscreens [53,54] and a substantial portion of diagnosed SCC do not contain TP53 mutations [36]. Therefore, it seems likely that UV radiation is producing additional responses in the cell that might serve as potential chemopreventive targets. Although sunscreens have not proven successful in significantly impacting NMSC incidence, new chemopreventive strategies in addition to sunscreen application may be a more effective means of preventing skin cancer.
40 Current Cancer Therapy Reviews, 2009, Vol. 5, No. 1
Bachelor and Owens
UV MEDIATED INFLAMMATION AND IMMUNE SUPPRESSION
to protect the body from tumor development both in the skin and in distant organs.
Inflammation is a key factor for tumorigenesis in numerous tissues. In UV-irradiated skin, inflammation is observed as a result of increased blood flow and infiltration by macrophages and neutrophils [55]. In addition, epithelial keratinocytes exposed to UV release pro-inflammatory cytokines, such as TNF-, IL-1, IL-1 and IL-6 [55]. TNF- secretion plays an important role in SCC development in mouse epidermis [56]. TNF- knockout (KO) mice treated in a twostage chemical carcinogenesis protocol were resistant to keratinocyte hyperproliferation, inflammation and the development of benign and malignant skin tumors, suggesting a role for inflammation in SCC development that can be mediated by UV-induced cytokines [56]. While TNF- therapies have not been tested as targets for the treatment of NMSC, they have been met with some success in other epithelial cancers. Patients with recurrent ovarian cancer receiving etanercept, a TNF- inhibitor, achieved prolonged disease stabilization although no tumor regression was observed [57]. In metastatic breast cancer patients, however etanercept did not achieve significant disease stabilization or tumor regression [58]. It is well established that keratinocytes secrete IL-1 and IL-1 in response to UV radiation; however, a direct link between induction of either IL-1 and IL-1 in skin tumor development in vivo remains to be determined. UVB irradiated IL-6 KO mice display delayed epidermal hyperplasia and greater overall damage to the epidermis suggesting IL-6 plays an important role in the repair of UVdamaged skin [59]; however, IL-6 protection from UV damage may not be critical in preventing tumor formation [60]. Recently, an IL-6 monoclonal antibody, tocilizumab was tested for its use in the treatment of rheumatoid arthritis; however its efficacy in the treatment of epithelial cancers has not yet been tested [61]. Interestingly, IL-6 may play a larger role in outside of inflammation by regulating levels of IL-10, a critical mediator of UV-induced immunosuppression. In addition to pro-inflammatory molecules produced by epidermal cells, the importance of the inflammatory response derived from the underlying dermis has recently been illustrated. Mice deficient in the D6 chemokine receptor demonstrate that sequestration of the inflammatory D6 chemokine, produced by dermal mast cells, is essential to suppress epidermal carcinogenesis by altering keratinocyte proliferation [62]. Whether UV can impact the induction of dermal chemokines and influence cutaneous tumor development is an interesting concept that requires further investigation.
UV induces lipid peroxidation in the cutaneous epithelium, which results in damage to the cell membrane and increases in ROS [65]. In response to lipid peroxidation caused by UVA and UVB irradiation, cells also increase production of prostaglandins as a consequence of arachidonic release from membrane-bound phospholipase A2. The production of nitric oxide and prostaglandins further contribute to the prooxidant state of UV-irradiated cells through production of additional ROS. PGE2, a product of cyclooxygenase-2 (COX-2) enzyme catalysis of arachidonic acid, contributes to UV-induced inflammation, edema, keratinocyte proliferation and inhibits UV-induced apoptosis [66]. Increases in PGE2 levels resulting from UV-induced oxidative damage also contribute to immunosuppression [67]. Basal COX-2 expression is maintained at low levels in the skin and is induced in response to UVB [68]. Increased COX enzymatic activity produces ROS, which in combination with ROS produced by inflammatory cells may contribute to oxidative DNA damage [35,65,69]. COX-2 gene expression is critical for UVinduced skin tumorigenesis as evidenced by reduction in the number of tumors and increase in latency in COX-2 KO mice. COX-2 +/- mice also exhibit reduced levels of PGE2 following UVB irradiation [70]. Transgenic mice overexpressing COX-2 in the basal layer of the epidermis had dramatically increased levels of PGE2 in the epidermis and displayed greater sensitivity to tumor formation [71]. In addition to altering levels of prostaglandins in the skin, it has been suggested that by inhibiting COX-2 activity and subsequent PGE2 levels in the skin, levels of apoptosis are enhanced, as skin tumors are dependent on PGE2 signaling for growth [72].
In conjunction with the pro-inflammatory response of the skin, UV radiation also has local and systemic immunosuppressive effects. Local immunosuppression primarily involves depletion in the number of Langerhans cells, the major antigen-presenting cell within the skin, which could be advantageous for the survival of mutant epidermal cells. Langerhans cells are also compromised in their ability to present antigens due to a reduction in major histocompatibility complex II surface molecules following UV exposure [63]. Keratinocytes produce IL-10 in response to UV radiation, which appears to be the critical mediator of UVinduced immunosuppression by inhibiting the function of antigen presenting cells [64]. Overall, UV exposure in the skin can impact normal immune surveillance that functions
Traditional non-steroidal anti-inflammatory drugs (NSAIDs), inhibitors of both COX-1 and COX-2 isoforms, as well as selective COX-2 inhibitors, such as celecoxib and indomethacin, can inhibit UV-induced tumorigenesis in mice [73,74]. The inhibition of COX-2 appears to be more effective as a chemoprevention strategy, as treatment with celecoxib had little or no therapeutic impact on regression of existing tumors [69,73,74]. The development of inhibitors of PGE2 signaling may prove more clinically viable in human trials as the side effects of COX inhibitors preclude them for use as chemopreventive agents. NSAIDs, such as aspirin, have harmful gastrointestinal complications resulting from the inhibition of both COX-1 and COX-2 isoforms. These side effects were the initial impetus for the development of selective COX-2 inhibitors, although these have also recently been shown to be associated with increased risk for cardiovascular complications [75]. Further studies will be necessary to develop a strategy to target PGE2 signaling as a chemopreventive strategy while minimizing side effects. ROS-INDUCED EFFECTS ON SKIN ROS are generated by epithelial keratinocytes and dermal fibroblasts in skin at low levels during the course of aerobic metabolism but are normally removed by nonenzymatic and enzymatic processes to prevent oxidative cellular damage. UVA and UVB irradiation also induce specific ROS species production. When the antioxidant defense mechanisms be-
Squamous Cell Carcinoma of the Skin
come overwhelmed in favor of pro-oxidants, the cell enters a state of oxidative stress, as occurs in UV-irradiated skin [76,77]. In addition, the effects of UV irradiation are compounded by the inability of cells to sufficiently scavenge ROS, it remains of interest to explore the effects of altering enzymatic oxidants within the cell. One of the primary enzymatic antioxidant defenses that metabolize ROS within the cell is catalase [78]. The importance of catalase in the breakdown of ROS has been demonstrated using malignant mouse keratinocytes [79]. In these malignant cells, overexpression of catalase can inhibit ROS and concomitantly establish a functional role for decreased catalase levels in augmenting the malignant phenotype [78,80-82]. The overexpression of catalase also reduces UVB-induced apoptosis in normal human keratinocytes preventing DNA damage induced by increases in ROS [83,84]. Collectively, these studies highlight catalase as a key player in mediating the effects of ROS on tumor progression. Manipulating catalase levels has also been utilized to alter levels of apoptosis in reconstructed epidermis from Xeroderma pigmentosum type C (XPC) patients. XPC is an autosomal recessive disease, resulting from inactivation of the XPC protein involved in DNA damage recognition and resulting in defective nucleotide excision repair. UVB irradiation of reconstructed XPC human epidermis overexpressing catalase resulted in a significant decrease in sunburn cell formation, caspase-3 activation and p53 accumulation [83]. These studies highlight the clinical impact that enzyme replacement or manipulation can have in ameliorating a genetic defect. In addition to targeting the XPC genetic defect itself, manipulation of enzymatic pathways may be a viable alternative in limiting photosensitivity in XPC patients. Resveratrol is a polyphenolic compound found in grapes and red wine, having anti-carcinogenic effects associated with its antioxidant activity. Resveratrol can inhibit a number of cellular proteins, including COX enzymes, hydroperoxidase, protein kinase C, Bcl-2 phosphorylation, AKT, focal adhesion kinase, NF-B, matrix metalloproteinase-9, and cell cycle regulators [85]. Resveratrol treatment decreases tumor incidence and delays the onset of tumor formation in mice exposed to UVB irradiation or treated with a two-stage chemical carcinogenesis protocol [86,87]. Pretreatment of UVB-irradiated mice mouse skin with resveratrol decreased bifold skin thickness, edema and reduced the number of infiltrating leukocytes [88]. In addition, resveratrol treatment reduced UVB-induced COX-2 expression and H2O2 production in the skin, confirming previous reports of resveratrol as a strong antioxidant [89,90]. The putative ability of resveratrol to target multiple UV-induced cellular changes makes it a strong candidate for use as a chemopreventive agent in NMSC. While nuclear DNA is the usually the focus of UVinduced damage, mitochondrial DNA (mtDNA) can also be damaged by oxidative stress [91]. Mutations in nuclear DNA are more likely to be repaired whereas mutations occurring in mtDNA are apt to accumulate, as repair mechanisms are less efficient [92]. Accordingly, sun-exposed samples of human skin contained mutated mtDNA in 27% of samples analyzed, compared to 1.1% in non sun-exposed areas [93]. Interestingly, 93% of the mutations were contained in the
Current Cancer Therapy Reviews, 2009, Vol. 5, No. 1
41
dermal compartment. Further studies demonstrated that the deletions in dermal cell mtDNA were attributable to singlet oxygen, the principle ROS species induced by UVA irradiation [94]. Reciprocal exchange experiments, which swap the mtDNA between tumor cells with low metastatic potential and cells with high metastatic potential, demonstrate that mutations in mtDNA can critically impact metastasis [95]. Defects in complex I mtDNA were shown to be responsible for acquiring metastatic potential and were due to a mutant NADH dehydrogenase subunit 6 gene which also leads to the overproduction of ROS [95-97]. The concomitant increases in ROS can stimulate metastasis through the upregulation of genes associated with tumor progression, including HIF-1, MCL-1 and VEGF. Interestingly, inappropriate expression of these genes was reversed when the mutated mtDNA was replaced with wild-type mtNDA [95]. Therapies targeting mtDNA defects remain largely unexplored and may offer significant clinical impact for UV-induced NMSC. EPIDERMAL STEM CELLS AS TARGETS FOR CHEMOPREVENTION Despite the vulnerability of epidermal keratinocytes to UV-induced genetic mutations, relatively few cancers develop as most of these mutant cells are shed from the skin during the normal process of terminal differentiation [6]. Given that the development of skin cancer takes 20-30 years to develop in humans, only cells that are long-term residents in the skin, i.e. stem cells, are capable of acquiring the necessary number of genetic hits necessary to result in tumor formation. The cutaneous epithelium is now thought to be composed of multiple progenitor cell niches. Epithelial progenitor cell populations have been identified in both the sebaceous gland (SG) and interfollicular epidermis (IFE) [98101]. In addition to the stem cell reservoir in the bulge region [102], additional progenitor cell populations have also recently been identified in the hair follicle [103-105]. These progenitor populations are long-term residents in the skin and can maintain distinct epithelial lineages including the epidermis, hair follicles and sebaceous glands [103-105]. Importantly, expression array studies identified genes that are unique or enriched in these progenitor populations and in some cases are responsible for maintaining the integrity of certain epithelial lineages [102,105]. These aforementioned characteristics of epithelial progenitor populations suggests that these cells may serve as target cells for UV-induced NMSC and raises the possibility that genes unique to these cells may play a role in propagating epithelial tumor stem cells. A better understanding of these regionalized compartments of phenotypically distinct epithelial progenitor cells may aid in the design for more effective strategies in preventing and treating UV-induced NMSC. Multiple lines of experimental evidence have provided support for the idea that genes responsible for the maintenance of stem cells may also be altered in skin cancer. Overexpression of genes involved in epidermal stem cell maintenance including c-myc or Lef1 lead to de novo skin tumor formation [106,107] whereas ablation of the small Rho GTPase, Rac1, leads to depletion of the entire epidermis and hair follicles [108]. Sox9, a gene essential for maintenance of the hair follicle bulge stem cell compartment is also constitutively expressed in BCC through upregulation of upstream
42 Current Cancer Therapy Reviews, 2009, Vol. 5, No. 1
signaling pathways [109,110]. Manipulation of stem cell niches in other tissue types including Wnt and Hedgehog signaling in the hematopoietic system and Bmi1 in brain indicate they have a role in both development and oncogenesis [111,112]. Observations made by augmenting expression of genes within the stem cell compartment underscore their capacity to regulate homeostasis and tumorigenesis and indicate that genes unique to epidermal stem cells are potentially lucrative targets for SCC treatment and prevention. A prime example supporting this concept has been observed in CD34+ cancer stem cells recently identified in chemically induced SCC [113]. This subpopulation of CD34+ keratinocytes, but not CD34- SCC cells, recapitulate the original tumor phenotype indicating that the CD34+ population of cells contains putative cancer stem cells [113]. Interestingly, CD34+ SCC cells were found to be dependent on -catenin signaling, a pathway required for normal hair follicle development, as deletion of -catenin in these tumors led to a reduced population of CD34+ cells, along with increased terminal differentiation. These findings were also applicable to human SCC development as knockdown of -catenin signaling in a human SCC cell line led to decreases in xenograft tumors. This study illustrates how a signaling pathway modulating stem cell maintenance under normal conditions affect tumorigenesis and offers an avenue for therapeutic intervention.
Bachelor and Owens [14] [15]
[16]
[17] [18]
[19] [20] [21] [22] [23]
[24] [25] [26]
ACKNOWLEDGEMENTS This work was supported by National Institutes of Health F32AR055007 (M.A.B.), RO3AR054071 (D.M.O.) and R01CA114014 (D.M.O.) research grants. REFERENCES [1]
[2] [3] [4] [5] [6] [7]
[8] [9] [10]
[11]
[12] [13]
Johnson TM, Dolan OM, Hamilton TA, Lu MC, Swanson NA, Lowe L. Clinical and histologic trends of melanoma. J Am Acad Dermatol 1998; 38(5 Pt 1): 681-6. Parker SL, Tong T, Bolden S, Wingo PA. Cancer statistics 1997. CA Cancer J Clin 1997; 47(1): 5-27. de Gruijl FR, Sterenborg HJ, Forbes PD, et al. Wavelength dependence of skin cancer induction by ultraviolet irradiation of albino hairless mice. Cancer Res 1993; 53(1): 53-60. Young AR. Cumulative effects of ultraviolet radiation on the skin: cancer and photoaging. Semin Dermatol 1990; 9(1): 25-31. Armstrong BK, Kricker A. The epidemiology of UV induced skin cancer. J Photochem Photobiol B 2001; 63(1-3): 8-18. Owens DM, Watt FM. Contribution of stem cells and differentiated cells to epidermal tumours. Nat Rev Cancer 2003; 3(6): 444-51. Kelfkens G, de Gruijl FR, van der Leun JC. Ozone depletion and increase in annual carcinogenic ultraviolet dose. Photochem Photobiol 1990; 52(4): 819-23. Marks R. An overview of skin cancers. Incidence and causation. Cancer 1995; 75(2 Suppl): 607-12. Moller R, Reymann F, Hou-Jensen K. Metastases in dermatological patients with squamous cell carcinoma. Arch Dermatol 1979; 115(6): 703-5. Dinehart SM, Pollack SV. Metastases from squamous cell carcinoma of the skin and lip. An analysis of twenty-seven cases. J Am Acad Dermatol 1989; 21(2 Pt 1): 241-8. Karagas MR, Stukel TA, Greenberg ER, Baron JA, Mott LA, Stern RS. Risk of subsequent basal cell carcinoma and squamous cell carcinoma of the skin among patients with prior skin cancer. Skin Cancer Prevention Study Group. JAMA 1992; 267(24): 3305-10. Preston DS, Stern RS. Nonmelanoma cancers of the skin. N Engl J Med 1992; 327(23): 1649-62. Cherpelis BS, Marcusen C, Lang PG. Prognostic factors for metastasis in squamous cell carcinoma of the skin. Dermatol Surg 2002; 28(3): 268-73.
[27]
[28]
[29]
[30]
[31]
[32] [33] [34] [35] [36]
[37]
[38]
Weinberg AS, Ogle CA, Shim EK. Metastatic cutaneous squamous cell carcinoma: an update. Dermatol Surg 2007; 33(8): 885-99. Rowe DE, Carroll RJ, Day CL, Jr. Prognostic factors for local recurrence, metastasis, and survival rates in squamous cell carcinoma of the skin, ear, and lip. Implications for treatment modality selection. J Am Acad Dermatol 1992; 26(6): 976-90. Mehrany K, Byrd DR, Roenigk RK, et al. Lymphocytic infiltrates and subclinical epithelial tumor extension in patients with chronic leukemia and solid-organ transplantation. Dermatol Surg 2003; 29(2): 129-34. Manusow D, Weinerman BH. Subsequent neoplasia in chronic lymphocytic leukemia. JAMA 1975; 232(3): 267-9. Mitropoulos P, Norman R. Nevoid basal cell carcinoma syndrome (Gorlin syndrome): updated review of minimally invasive treatments. Cutis 2008; 81(1): 53-60. Green CL, Khavari PA. Targets for molecular therapy of skin cancer. Semin Cancer Biol 2004; 14(1): 63-9. Neville JA, Welch E, Leffell DJ. Management of nonmelanoma skin cancer in 2007. Nat Clin Pract Oncol 2007; 4(8): 462-9. Martinez JC, Otley CC. The management of melanoma and nonmelanoma skin cancer: a review for the primary care physician. Mayo Clin Proc 2001; 76(12): 1253-65. Nguyen TH, Ho DQ. Nonmelanoma skin cancer. Curr Treat Options Oncol 2002; 3(3): 193-203. Campbell RM, DiGiovanna JJ. Skin cancer chemoprevention with systemic retinoids: an adjunct in the management of selected highrisk patients. Dermatol Ther 2006; 19(5): 306-14. McKenzie RC. Selenium, ultraviolet radiation and the skin. Clin Exp Dermatol 2000; 25(8): 631-6. Stahl W, Sies H. Bioactivity and protective effects of natural carotenoids. Biochim Biophys Acta 2005; 1740(2): 101-7. Clark LC, Combs GF, Jr., Turnbull BW et al. Effects of selenium supplementation for cancer prevention in patients with carcinoma of the skin. A randomized controlled trial. Nutritional Prevention of Cancer Study Group. JAMA 1996; 276(24): 1957-63. Greenberg ER, Baron JA, Stukel TA, et al. A clinical trial of beta carotene to prevent basal-cell and squamous-cell cancers of the skin. The Skin Cancer Prevention Study Group. N Engl J Med 1990; 323(12): 789-95. Moon TE, Levine N, Cartmel B, et al. Effect of retinol in preventing squamous cell skin cancer in moderate-risk subjects: a randomized, double-blind, controlled trial. Southwest Skin Cancer Prevention Study Group. Cancer Epidemiol Biomarkers Prev 1997; 6(11): 949-56. Levine N, Moon TE, Cartmel B, et al. Trial of retinol and isotretinoin in skin cancer prevention: a randomized, double-blind, controlled trial. Southwest Skin Cancer Prevention Study Group. Cancer Epidemiol Biomarkers Prev 1997; 6(11): 957-61. Bavinck JN, Tieben LM, Van der Woude FJ, et al. Prevention of skin cancer and reduction of keratotic skin lesions during acitretin therapy in renal transplant recipients: a double-blind, placebocontrolled study. J Clin Oncol 1995; 13(8): 1933-8. Kraemer KH, DiGiovanna JJ, Moshell AN, Tarone RE, Peck GL. Prevention of skin cancer in xeroderma pigmentosum with the use of oral isotretinoin. N Engl J Med 1988; 318(25): 1633-7. Matsui MS, DeLeo VA. Longwave ultraviolet radiation and promotion of skin cancer. Cancer Cells 1991; 3(1): 8-12. Berg RJ, de Gruijl FR, van der Leun JC. Interaction between ultraviolet A and ultraviolet B radiations in skin cancer induction in hairless mice. Cancer Res 1993; 53(18): 4212-7. Sterenborg HJ, van der Leun JC. Tumorigenesis by a long wavelength UV-A source. Photochem Photobiol 1990; 51(3): 325-30. Bowden GT. Prevention of non-melanoma skin cancer by targeting ultraviolet-B-light signalling. Nat Rev Cancer 2004; 4(1): 23-35. Brash DE, Rudolph JA, Simon JA, et al. A role for sunlight in skin cancer: UV-induced p53 mutations in squamous cell carcinoma. Proc Natl Acad Sci U S A 1991; 88(22): 10124-8. Campbell C, Quinn AG, Ro YS, Angus B, Rees JL. p53 mutations are common and early events that precede tumor invasion in squamous cell neoplasia of the skin. J Invest Dermatol 1993; 100(6): 746-8. Kanjilal S, Strom SS, Clayman GL, et al. p53 mutations in nonmelanoma skin cancer of the head and neck: molecular evidence for field cancerization. Cancer Res 1995; 55(16): 3604-9.
Squamous Cell Carcinoma of the Skin [39]
[40] [41]
[42] [43] [44]
[45] [46]
[47] [48]
[49]
[50]
[51]
[52]
[53]
[54] [55]
[56] [57]
[58]
[59]
[60] [61]
Nelson MA, Einspahr JG, Alberts DS, et al. Analysis of the p53 gene in human precancerous actinic keratosis lesions and squamous cell cancers. Cancer Lett 1994; 85(1): 23-9. Tornaletti S, Rozek D, Pfeifer GP. The distribution of UV photoproducts along the human p53 gene and its relation to mutations in skin cancer. Oncogene 1993; 8(8): 2051-7. Ziegler A, Leffell DJ, Kunala S, et al. Mutation hotspots due to sunlight in the p53 gene of nonmelanoma skin cancers. Proc Natl Acad Sci U S A 1993; 90(9): 4216-20. de Gruijl FR. Photocarcinogenesis: UVA vs UVB. Methods Enzymol 2000; 319: 359-66. Runger TM. Role of UVA in the pathogenesis of melanoma and non-melanoma skin cancer. A short review. Photodermatol Photoimmunol Photomed 1999; 15(6): 212-6. Silvers AL, Bowden GT. UVA irradiation-induced activation of activator protein-1 is correlated with induced expression of AP-1 family members in the human keratinocyte cell line HaCaT. Photochem Photobiol 2002; 75(3): 302-10. Grether-Beck S, Krutmann J. Gene regulation by ultraviolet A radiation and singlet oxygen. Methods Enzymol 2000; 319: 280-90. Ikehata H, Kawai K, Komura J, et al. UVA1 genotoxicity is mediated not by oxidative damage but by cyclobutane pyrimidine dimers in normal mouse skin. J Invest Dermatol 2008; 128(9): 228996. Ziegler A, Jonason AS, Leffell DJ, et al. Sunburn and p53 in the onset of skin cancer. Nature 1994; 372(6508): 773-6. Bolshakov S, Walker CM, Strom SS, et al. p53 mutations in human aggressive and nonaggressive basal and squamous cell carcinomas. Clin Cancer Res 2003; 9(1): 228-34. Benjamin CL, Ullrich SE, Kripke ML, Ananthaswamy HN. p53 tumor suppressor gene: a critical molecular target for UV induction and prevention of skin cancer. Photochem Photobiol 2008; 84(1): 55-62. Weinberg WC, Azzoli CG, Kadiwar N, Yuspa SH. p53 gene dosage modifies growth and malignant progression of keratinocytes expressing the v-rasHa oncogene. Cancer Res 1994; 54(21): 558492. Ananthaswamy HN, Loughlin SM, Cox P, Evans RL, Ullrich SE, Kripke ML. Sunlight and skin cancer: inhibition of p53 mutations in UV-irradiated mouse skin by sunscreens. Nat Med 1997; 3(5): 510-4. Ananthaswamy HN, Ullrich SE, Mascotto RE, et al. Inhibition of solar simulator-induced p53 mutations and protection against skin cancer development in mice by sunscreens. J Invest Dermatol 1999; 112(5): 763-8. Harvey I, Frankel S, Marks R, Shalom D, Nolan-Farrell M. Nonmelanoma skin cancer and solar keratoses II analytical results of the South Wales Skin Cancer Study. Br J Cancer 1996; 74(8): 1308-12. Naylor MF, Boyd A, Smith DW, Cameron GS, Hubbard D, Neldner KH. High sun protection factor sunscreens in the suppression of actinic neoplasia. Arch Dermatol 1995; 131(2): 170-5. Svobodova A, Walterova D, Vostalova J. Ultraviolet light induced alteration to the skin. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2006; 150(1): 25-38. Moore RJ, Owens DM, Stamp G, et al. Mice deficient in tumor necrosis factor-alpha are resistant to skin carcinogenesis. Nat Med 1999; 5(7): 828-31. Madhusudan S, Muthuramalingam SR, Braybrooke JP, et al. Study of etanercept, a tumor necrosis factor-alpha inhibitor, in recurrent ovarian cancer. J Clin Oncol 2005; 23(25): 5950-9. Madhusudan S, Foster M, Muthuramalingam SR, et al. A phase II study of etanercept (Enbrel), a tumor necrosis factor alpha inhibitor in patients with metastatic breast cancer. Clin Cancer Res 2004; 10(19): 6528-34. Nishimura N, Tohyama C, Satoh M, Nishimura H, Reeve VE. Defective immune response and severe skin damage following UVB irradiation in interleukin-6-deficient mice. Immunology 1999; 97(1): 77-83. Suganuma M, Okabe S, Kurusu M, et al. Discrete roles of cytokines, TNF-alpha, IL-1, IL-6 in tumor promotion and cell transformation. Int J Oncol 2002; 20(1): 131-6. Sebba A. Tocilizumab: the first interleukin-6-receptor inhibitor. Am J Health Syst Pharm 2008; 65(15): 1413-8.
Current Cancer Therapy Reviews, 2009, Vol. 5, No. 1 [62]
[63]
[64] [65] [66] [67]
[68]
[69]
[70]
[71] [72]
[73] [74]
[75] [76] [77]
[78] [79]
[80]
[81]
[82]
[83]
[84]
43
Nibbs RJ, Gilchrist DS, King V, et al. The atypical chemokine receptor D6 suppresses the development of chemically induced skin tumors. J Clin Invest 2007; 117(7): 1884-92. Stingl LA, Sauder DN, Iijima M, Wolff K, Pehamberger H, Stingl G. Mechanism of UV-B-induced impairment of the antigenpresenting capacity of murine epidermal cells. J Immunol 1983; 130(4): 1586-91. Schwarz T. Mechanisms of UV-induced immunosuppression. Keio J Med 2005; 54(4): 165-71. Bickers DR, Athar M. Oxidative stress in the pathogenesis of skin disease. J Invest Dermatol 2006; 126(12): 2565-75. Rundhaug JE, Fischer SM. Cyclo-oxygenase-2 plays a critical role in UV-induced skin carcinogenesis. Photochem Photobiol 2008; 84(2): 322-9. Shreedhar V, Giese T, Sung VW, Ullrich SE. A cytokine cascade including prostaglandin E2, IL-4, and IL-10 is responsible for UVinduced systemic immune suppression. J Immunol 1998; 160(8): 3783-9. Chen W, Tang Q, Gonzales MS, Bowden GT. Role of p38 MAP kinases and ERK in mediating ultraviolet-B induced cyclooxygenase-2 gene expression in human keratinocytes. Oncogene 2001; 20(29): 3921-6. Wilgus TA, Parrett ML, Ross MS, Tober KL, Robertson FM, Oberyszyn TM. Inhibition of ultraviolet light B-induced cutaneous inflammation by a specific cyclooxygenase-2 inhibitor. Adv Exp Med Biol 2002; 507: 85-92. Fischer SM, Pavone A, Mikulec C, Langenbach R, Rundhaug JE. Cyclooxygenase-2 expression is critical for chronic UV-induced murine skin carcinogenesis. Mol Carcinog 2007; 46(5): 363-71. Bol DK, Rowley RB, Ho CP, et al. Cyclooxygenase-2 overexpression in the skin of transgenic mice results in suppression of tumor development. Cancer Res 2002; 62(9): 2516-21. Thompson EJ, Gupta A, Vielhauer GA, Regan JW, Bowden GT. The growth of malignant keratinocytes depends on signaling through the PGE(2) receptor EP1. Neoplasia 2001; 3(5): 402-10. Pentland AP, Schoggins JW, Scott GA, Khan KN, Han R. Reduction of UV-induced skin tumors in hairless mice by selective COX2 inhibition. Carcinogenesis 1999; 20(10):1939-44. Fischer SM, Lo HH, Gordon GB, et al. Chemopreventive activity of celecoxib, a specific cyclooxygenase-2 inhibitor, and indomethacin against ultraviolet light-induced skin carcinogenesis. Mol Carcinog 1999; 25(4): 231-40. Yona D, Arber N. Coxibs and cancer prevention. J Cardiovasc Pharmacol 2006; 47 Suppl 1: S76-81. Afaq F, Mukhtar H. Photochemoprevention by botanical antioxidants. Skin Pharmacol Appl Skin Physiol 2002; 15(5): 297-306. Heck DE, Gerecke DR, Vetrano AM, Laskin JD. Solar ultraviolet radiation as a trigger of cell signal transduction. Toxicol Appl Pharmacol 2004; 195(3): 288-97. Finch JS, Tome ME, Kwei KA, Bowden GT. Catalase reverses tumorigenicity in a malignant cell line by an epidermal growth factor receptor pathway. Free Radic Biol Med 2006; 40(5): 863-75. Gupta A, Rosenberger SF, Bowden GT. Increased ROS levels contribute to elevated transcription factor and MAP kinase activities in malignantly progressed mouse keratinocyte cell lines. Carcinogenesis 1999; 20(11): 2063-73. Gupta A, Butts B, Kwei KA, et al. Attenuation of catalase activity in the malignant phenotype plays a functional role in an in vitro model for tumor progression. Cancer Lett 2001; 173(2): 115-25. Milczarek GJ, Chen W, Gupta A, Martinez JD, Bowden GT. Okadaic acid mediates p53 hyperphosphorylation and growth arrest in cells with wild-type p53 but increases aberrant mitoses in cells with non-functional p53. Carcinogenesis 1999; 20(6): 1043-8. Rosenberger SF, Finch JS, Gupta A, Bowden GT. Extracellular signal-regulated kinase 1/2-mediated phosphorylation of JunD and FosB is required for okadaic acid-induced activator protein 1 activation. J Biol Chem 1999; 274(2): 1124-30. Rezvani HR, Ged C, Bouadjar B, de Verneuil H, Taieb A. Catalase overexpression reduces UVB-induced apoptosis in a human xeroderma pigmentosum reconstructed epidermis. Cancer Gene Ther 2008; 15(4): 241-51. Rezvani HR, Mazurier F, Cario-Andre M, et al. Protective effects of catalase overexpression on UVB-induced apoptosis in normal human keratinocytes. J Biol Chem 2006; 281(26): 17999-18007.
44 Current Cancer Therapy Reviews, 2009, Vol. 5, No. 1 [85]
[86] [87]
[88] [89]
[90] [91] [92] [93]
[94]
[95] [96] [97]
[98]
[99]
Bachelor and Owens
Athar M, Back JH, Tang X, et al. Resveratrol: a review of preclinical studies for human cancer prevention. Toxicol Appl Pharmacol 2007; 224(3): 274-83. Jang M, Cai L, Udeani GO, et al. Cancer chemopreventive activity of resveratrol, a natural product derived from grapes. Science 1997; 275(5297): 218-20. Aziz MH, Reagan-Shaw S, Wu J, Longley BJ, Ahmad N. Chemoprevention of skin cancer by grape constituent resveratrol: relevance to human disease? FASEB J 2005; 19(9): 1193-5. Afaq F, Adhami VM, Ahmad N. Prevention of short-term ultraviolet B radiation-mediated damages by resveratrol in SKH-1 hairless mice. Toxicol Appl Pharmacol 2003; 186(1): 28-37. Murcia MA, Martinez-Tome M. Antioxidant activity of resveratrol compared with common food additives. J Food Prot 2001; 64(3): 379-84. Stivala LA, Savio M, Carafoli F, et al. Specific structural determinants are responsible for the antioxidant activity and the cell cycle effects of resveratrol. J Biol Chem 2001; 276(25): 22586-94. Kang D, Hamasaki N. Mitochondrial oxidative stress and mitochondrial DNA. Clin Chem Lab Med 2003; 41(10): 1281-8. Croteau DL, Stierum RH, Bohr VA. Mitochondrial DNA repair pathways. Mutat Res 1999; 434(3): 137-48. Birch-Machin MA, Tindall M, Turner R, Haldane F, Rees JL. Mitochondrial DNA deletions in human skin reflect photo- rather than chronologic aging. J Invest Dermatol 1998; 110(2): 149-52. Berneburg M, Grether-Beck S, Kurten V, et al. Singlet oxygen mediates the UVA-induced generation of the photoaging-associated mitochondrial common deletion. J Biol Chem 1999; 274(22): 15345-9. Ishikawa K, Takenaga K, Akimoto M, et al. ROS-generating mitochondrial DNA mutations can regulate tumor cell metastasis. Science 2008; 320(5876): 661-4. Wallace DC. Mitochondrial diseases in man and mouse. Science 1999; 283(5407): 1482-8. De Flora S, D'Agostini F, Masiello L, Giunciuglio D, Albini A. Synergism between N-acetylcysteine and doxorubicin in the prevention of tumorigenicity and metastasis in murine models. Int J Cancer 1996; 67(6): 842-8. Clayton E, Doupe DP, Klein AM, Winton DJ, Simons BD, Jones PH. A single type of progenitor cell maintains normal epidermis. Nature 2007; 446(7132): 185-9. Ghazizadeh S, Taichman LB. Multiple classes of stem cells in cutaneous epithelium: a lineage analysis of adult mouse skin. EMBO J 2001; 20(6): 1215-22.
Received: September 25, 2008
[100]
[101] [102]
[103] [104]
[105]
[106] [107] [108]
[109] [110]
[111] [112] [113]
Horsley V, O'Carroll D, Tooze R, et al. Blimp1 defines a progenitor population that governs cellular input to the sebaceous gland. Cell 2006; 126(3): 597-609. Tani H, Morris RJ, Kaur P. Enrichment for murine keratinocyte stem cells based on cell surface phenotype. Proc Natl Acad Sci U S A 2000; 97(20): 10960-5. Trempus CS, Morris RJ, Bortner CD, et al. Enrichment for living murine keratinocytes from the hair follicle bulge with the cell surface marker CD34. J Invest Dermatol 2003; 120(4): 501-11. Yan X, Owens DM. The skin: a home to multiple classes of epithelial progenitor cells. Stem Cell Rev 2008; 4(2): 113-8. Nijhof JG, Braun KM, Giangreco A, et al. The cell-surface marker MTS24 identifies a novel population of follicular keratinocytes with characteristics of progenitor cells. Development 2006; 133(15): 3027-37. Jensen UB, Lowell S, Watt FM. The spatial relationship between stem cells and their progeny in the basal layer of human epidermis: a new view based on whole-mount labelling and lineage analysis. Development 1999; 126(11): 2409-18. Arnold I, Watt FM. c-Myc activation in transgenic mouse epidermis results in mobilization of stem cells and differentiation of their progeny. Curr Biol 2001; 11(8): 558-8. Niemann C, Watt FM. Designer skin: lineage commitment in postnatal epidermis. Trends Cell Biol 2002; 12(4): 185-92. Benitah SA, Frye M, Glogauer M, Watt FM. Stem cell depletion through epidermal deletion of Rac1. Science 2005; 309(5736): 9335. Vidal VP, Chaboissier MC, Lutzkendorf S, et al. Sox9 is essential for outer root sheath differentiation and the formation of the hair stem cell compartment. Curr Biol 2005; 15(15): 1340-51. Vidal VP, Ortonne N, Schedl A. SOX9 expression is a general marker of basal cell carcinoma and adnexal-related neoplasms. J Cutan Pathol 2008; 35(4): 373-9. Leung C, Lingbeek M, Shakhova O, et al. Bmi1 is essential for cerebellar development and is overexpressed in human medulloblastomas. Nature 2004; 428(6980): 337-41. Taipale J, Beachy PA. The Hedgehog and Wnt signalling pathways in cancer. Nature 2001; 411(6835): 349-54. Malanchi I, Peinado H, Kassen D, et al. Cutaneous cancer stem cell maintenance is dependent on beta-catenin signalling. Nature 2008; 452(7187): 650-3.
Revised: December 11, 2008
Accepted: December 13, 2008