Skip to main content Accessibility help
×
Hostname: page-component-5c6d5d7d68-sv6ng Total loading time: 0 Render date: 2024-08-07T02:17:27.020Z Has data issue: false hasContentIssue false

62 - Molecular oncology of basal cell carcinomas

from Part 3.3 - Molecular pathology: cancers of the skin

Published online by Cambridge University Press:  05 February 2015

Ervin H. Epstein
Affiliation:
Children’s Hospital Oakland Research Institute, Oakland, CA, USA
Edward P. Gelmann
Affiliation:
Columbia University, New York
Charles L. Sawyers
Affiliation:
Memorial Sloan-Kettering Cancer Center, New York
Frank J. Rauscher, III
Affiliation:
The Wistar Institute Cancer Centre, Philadelphia
Get access

Summary

Clinical and histologic aspects

Basal cell carcinomas (BCCs) are the most common human cancer, occurring in a predicted one-third of US Caucasians (1,2). Fortunately although their common occurrence makes them expensive in aggregate to treat, they metastasize only very rarely and cause morbidity generally via local invasion and damage to normal tissues. In the great majority of cases, they occur sporadically, in particular on the sun-exposed skin of older persons of Caucasian descent. Their incidence is rising, and this seems to correlate in part, but only in part, with increased exposure to UV mutagenesis. Histologically, BCC tumor cells resemble the keratinocytes of the basal layers of the inter-follicular epidermis and the outer root sheath of the hair follicles with large nuclei and relatively scant cytoplasm. Various clinical and histologic subtypes are recognized, the most common of which is the nodular variety in which the tumor is palpable, has a translucent appearance, often with a central “dell,” and typically is surmounted by visible telangectasia. Less common are more superficial BCCs with a “radial” growth pattern hugging the underside of the inter-follicular epidermis and occurring predominantly on the trunk rather than the face, and the morphea form, with smaller numbers of tumor cells embedded in a hyperplastic stroma. In general the histologic appearance of the BCC stroma differs from that of the surrounding normal stroma, and the expression pattern of the BCC stroma may differ from that of other cancers (3).

Type
Chapter
Information
Molecular Oncology
Causes of Cancer and Targets for Treatment
, pp. 693 - 697
Publisher: Cambridge University Press
Print publication year: 2013

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Epstein, EH.Basal cell carcinomas: attack of the hedgehog. Nature Reviews Cancer 2008;8:743–54.CrossRefGoogle ScholarPubMed
Rubin, AI, Chen, EH, Ratner, D.Basal-cell carcinoma. New England Journal of Medicine 2005;353:2262–9.CrossRefGoogle ScholarPubMed
Micke, P, Kappert, K, Ohshima, M, et al. In situ identification of genes regulated specifically in fibroblasts of human basal cell carcinoma. Journal of Investigative Dermatology 2007;127:1516–23.CrossRefGoogle ScholarPubMed
Gorlin, RJ.Nevoid basal-cell carcinoma syndrome. Medicine (Baltimore) 1987;66:98–113.CrossRefGoogle ScholarPubMed
Kimonis, VE, Goldstein, AM, Pastakia, B, et al. Clinical manifestations in 105 persons with nevoid basal cell carcinoma syndrome. American Journal of Medical Genetics 1997;69:299–308.3.0.CO;2-M>CrossRefGoogle ScholarPubMed
Kraemer, KH, Lee, M-M, Andrews, AD, Lambert, WC.The role of sunlight and dna repair in melanoma and nonmelanoma skin cancer. Archives of Dermatology 1994;130:1018–21.CrossRefGoogle ScholarPubMed
Parren, LJ, Frank, J.Hereditary tumour syndromes featuring basal cell carcinomas. British Journal of Dermatology 2011;165:30–4.CrossRefGoogle ScholarPubMed
Kricker, A, Armstrong, BK, English, DR, Heenan, PJ.Does intermittent sun exposure cause basal cell carcinoma? A case-control study in western Australia. International Journal of Cancer 1995;60:489–94.CrossRefGoogle ScholarPubMed
Rosso, S, Zanetti, R, Martinez, C, et al. The multicentre south European study “Helios”. II: Different sun exposure patterns in the aetiology of basal cell and squamous cell carcinomas of the skin. British Journal of Cancer 1996;73:1447–54.CrossRefGoogle Scholar
Chuang, TY, Reizner, GT, Elpern, DJ, Stone, JL, Farmer, ER.Nonmelanoma skin cancer in Japanese ethnic Hawaiians in Kauai, Hawaii: an incidence report. Journal of the American Academy of Dermatology 1995;33:422–6.CrossRefGoogle Scholar
Smith, R, Healy, E, Siddiqui, S, et al. Melanocortin 1 receptor variants in an Irish population. Journal of Investigative Dermatology 1998;111:119–22.CrossRefGoogle Scholar
Box, NF, Duffy, DL, Irving, RE, et al. Melanocortin-1 receptor genotype is a risk factor for basal and squamous cell carcinoma. Journal of Investigative Dermatology 2001;116:224–9.CrossRefGoogle ScholarPubMed
Gudbjartsson, DF, Sulem, P, Stacey, SN, et al. ASIP and TYR pigmentation variants associate with cutaneous melanoma and basal cell carcinoma. Nature Genetics 2008;40:886–91.CrossRefGoogle ScholarPubMed
Kong, A, Steinthorsdottir, V, Masson, G, et al. Parental origin of sequence variants associated with complex diseases. Nature 2009;462:868–74.CrossRefGoogle ScholarPubMed
Stacey, SN, Gudbjartsson, DF, Sulem, P, et al. Common variants on 1p36 and 1q42 are associated with cutaneous basal cell carcinoma but not with melanoma or pigmentation traits. Nature Genetics 2008;40:1313–18.CrossRefGoogle ScholarPubMed
Rafnar, T, Sulem, P, Stacey, SN, et al. Sequence variants at the TERT-CLPTM1L locus associate with many cancer types. Nature Genetics 2009;41:221–7.CrossRefGoogle ScholarPubMed
Stacey, SN, Sulem, P, Masson, G, et al. New common variants affecting susceptibility to basal cell carcinoma. Nature Genetics 2009;41:909–14.CrossRefGoogle ScholarPubMed
Liang, G, Qureshi, AA, Guo, Q, De Vivo, I, Han, J.No association between telomere length in peripheral blood leukocytes and the risk of nonmelanoma skin cancer. Cancer Epidemiology, Biomarkers and Prevention 2011;20:1043–5.CrossRefGoogle ScholarPubMed
Gailani, MR, Bale, SJ, Leffell, DJ, et al. Developmental defects in Gorlin syndrome related to a putative tumor suppressor gene on chromosome 9. Cell 1992;69:111–17.CrossRefGoogle ScholarPubMed
Hahn, H, Wicking, C, Zaphiropoulos, PG, et al. Mutations of the human homologue of Drosophila patched in the nevoid basal cell carcinoma syndrome. Cell 1996;85:841–51.CrossRefGoogle Scholar
Johnson, RL, Rothman, AL, Xie, J, et al. Human homolog of patched, a candidate gene for the basal cell nevus syndrome. Science 1996;272:1668–71.CrossRefGoogle ScholarPubMed
Fan, Z, Li, J, Du, J, et al. A missense mutation in PTCH2 underlies dominantly inherited NBCCS in a Chinese family. Journal of Medical Genetics 2008;45:303–8.CrossRefGoogle Scholar
Pastorino, L, Ghiorzo, P, Nasti, S, et al. Identification of a SUFU germline mutation in a family with Gorlin syndrome. American Journal of Medical Genetics A 2009;149A:1539–43.CrossRefGoogle Scholar
Yang, SH, Andl, T, Grachtchouk, V, et al. Pathological responses to oncogenic Hedgehog signaling in skin are dependent on canonical Wnt/beta3-catenin signaling. Nature Genetics 2008;40:1130–5.CrossRefGoogle ScholarPubMed
Aszterbaum, M, Epstein, J, Oro, A, et al. Ultraviolet and ionizing radiation enhance the growth of BCCs and trichoblastomas in patched heterozygous knockout mice. Nature Medicine 1999;5:1285–91.CrossRefGoogle ScholarPubMed
Mancuso, M, Pazzaglia, S, Tanori, M, et al. Basal cell carcinoma and its development: insights from radiation-induced tumors in Ptch1-deficient mice. Cancer Research 2004;64:934–41.CrossRefGoogle ScholarPubMed
Nilsson, M, Unden, AB, Krause, D, et al. Induction of basal cell carcinomas and trichoepitheliomas in mice overexpressing GLI-1. Proceedings of the National Academy of Sciences USA 2000;97:3438–43.CrossRefGoogle ScholarPubMed
Grachtchouk, M, Mo, R, Yu, S, et al. Basal cell carcinomas in mice overexpressing Gli2 in skin. Nature Genetics 2000;24:216–17.CrossRefGoogle ScholarPubMed
Youssef, KK, Van Keymeulen, A, Lapouge, G, et al. Identification of the cell lineage at the origin of basal cell carcinoma. Nature Cell Biology 12:299–305.
Grachtchouk, M, Pero, J, Yang, SH, et al. Basal cell carcinomas in mice arise from hair follicle stem cells and multiple epithelial progenitor populations. Journal of Clinical Investigation 121:1768–81.CrossRef
Kasper, M, Jaks, V, Are, A, et al. Wounding enhances epidermal tumorigenesis by recruiting hair follicle keratinocytes. Proceedings of the National Academy of Sciences USA 2011;108:4099–104.CrossRefGoogle ScholarPubMed
Wang, GY, Wang, J, Mancianti, ML, Epstein, EH, Jr. Basal cell carcinomas arise from hair follicle stem cells in ptch1(+/–) mice. Cancer Cell 2011;19:114–24.CrossRefGoogle ScholarPubMed
Wong, SY, Reiter, JF.Wounding mobilizes hair follicle stem cells to form tumors. Proceedings of the National Academy of Sciences USA 2011;108:4093–8.CrossRefGoogle ScholarPubMed
Williams, JA, Guicherit, OM, Zaharian, BI, et al. Identification of a small molecule inhibitor of the hedgehog signaling pathway: effects on basal cell carcinoma-like lesions. Proceedings of the National Academy of Sciences USA 2003;100:4616–21.CrossRefGoogle ScholarPubMed
Watkins, DN, Berman, DM, Burkholder, SG, et al. Hedgehog signalling within airway epithelial progenitors and in small-cell lung cancer. Nature 2003;422:313–17.CrossRefGoogle ScholarPubMed
Thayer, SP, di Magliano, MP, Heiser, PW, et al. Hedgehog is an early and late mediator of pancreatic cancer tumorigenesis. Nature 2003;425:851–6.CrossRefGoogle ScholarPubMed
Berman, DM, Karhadkar, SS, Maitra, A, et al. Widespread requirement for Hedgehog ligand stimulation in growth of digestive tract tumours. Nature 2003;425:846–51.CrossRefGoogle ScholarPubMed
Karhadkar, SS, Bova, GS, Abdallah, N, et al. Hedgehog signalling in prostate regeneration, neoplasia and metastasis. Nature 2004;431:707–12.CrossRefGoogle ScholarPubMed
Binns, W, James, LF, Shupe, JL, Everett, G.A congenital cyclopian-type malformation in lambs induced by maternal ingestion of a range plant, Veratrum californicum. American Journal of Veterinary Research 1963;24:1164–75.Google ScholarPubMed
Taipale, J, Chen, JK, Cooper, MK, et al. Effects of oncogenic mutations in Smoothened and Patched can be reversed by cyclopamine. Nature 2000;406:1005–9.CrossRefGoogle ScholarPubMed
Kim, J, Lee, JJ, Gardner, D, Beachy, PA.Arsenic antagonizes the Hedgehog pathway by preventing ciliary accumulation and reducing stability of the Gli2 transcriptional effector. Proceedings of the National Academy of Sciences USA 2010;107:13 432–7.CrossRefGoogle ScholarPubMed
Beauchamp, EM, Ringer, L, Bulut, G, et al. Arsenic trioxide inhibits human cancer cell growth and tumor development in mice by blocking Hedgehog/GLI pathway. Journal of Clinical Investigation 2011;121:148–60.CrossRefGoogle ScholarPubMed
Von Hoff, DD, LoRusso, PM, Rudin, CM, et al. Inhibition of the hedgehog pathway in advanced basal-cell carcinoma. New England Journal of Medicine 2009;361:1164–72.CrossRefGoogle Scholar
Sekulic, A, Migden, MR, Oro, AE, et al. Efficacy and safety of vismodegib in advanced basal-cell carcinoma. New England Journal of Medicine 2012;366:2171–9.CrossRefGoogle ScholarPubMed
Tang, JY, Mackay-Wiggan, JM, Aszterbaum, M, et al. Inhibiting the hedgehog pathway in patients with the basal-cell nevus syndrome. New England Journal of Medicine 2012;366:2180–8.CrossRefGoogle ScholarPubMed
Teperino, R, Amann, S, Bayer, M, et al. Hedgehog partial agonism drives Warburg-like metabolism in muscle and brown fat. Cell 2012;151:414–26.CrossRefGoogle ScholarPubMed
Yauch, RL, Dijkgraaf, GJ, Alicke, B, et al. Smoothened mutation confers resistance to a Hedgehog pathway inhibitor in medulloblastoma. Science 2009;326:572–4.CrossRefGoogle ScholarPubMed
Buonamici, S, Williams, J, Morrissey, M, et al. Interfering with resistance to smoothened antagonists by inhibition of the PI3K pathway in medulloblastoma. Science Translational Medicine 2010;2:51ra70.CrossRefGoogle ScholarPubMed
Dijkgraaf, GJ, Alicke, B, Weinmann, L, et al. Small molecule inhibition of GDC-0449 refractory smoothened mutants and downstream mechanisms of drug resistance. Cancer Research 2011;71:435–44.CrossRefGoogle ScholarPubMed
Nolan-Stevaux, O, Lau, J, Truitt, ML, et al. GLI1 is regulated through Smoothened-independent mechanisms in neoplastic pancreatic ducts and mediates PDAC cell survival and transformation. Genes and Development 2009;23:24–36.CrossRefGoogle Scholar
Jagani, Z, Mora-Blanco, EL, Sansam, CG, et al. Loss of the tumor suppressor Snf5 leads to aberrant activation of the Hedgehog-Gli pathway. Nature Medicine 2010;16:1429–33.CrossRefGoogle ScholarPubMed

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×