REFERENCES

1. Ley RD. Photoreactivation in humans. Proc Natl Acad Sci U S A 1993;90:4337.

2. Feehan RP, Shantz LM. Molecular signaling cascades involved in nonmelanoma skin carcinogenesis. Biochem J 2016;473:2973-94.

3. Sample A, He YY. Mechanisms and prevention of UV-induced melanoma. Photodermatol Photoimmunol Photomed 2018;34:13-24.

4. de Gruijl FR. Photocarcinogenesis: UVA vs. UVB radiation. Skin Pharmacol Appl Skin Physiol 2002;15:316-20.

5. Craig S, Earnshaw CH, Virós A. Ultraviolet light and melanoma. J Pathol 2018;244:578-85.

6. Patrick MH. Studies on thymine-derived UV photoproducts in DNA--I. Formation and biological role of pyrimidine adducts in DNA. Photochem Photobiol 1977;25:357-72.

7. Lee JH, Budanov AV, Karin M. Sestrins orchestrate cellular metabolism to attenuate aging. Cell Metabolism 2013;18:792-801.

8. Budanov AV, Karin M. p53 target genes Sestrin1 and Sestrin2 connect genotoxic stress and mTOR signaling. Cell 2008;134:451-60.

9. Holick MF. Biological effects of sunlight, ultraviolet radiation, visible light, infrared radiation and vitamin D for health. Anticancer Res 2016;36:1345-56.

10. Leavitt E, Lask G, Martin S. Sonic hedgehog pathway inhibition in the treatment of advanced basal cell carcinoma. Curr Treat Options Oncol 2019;20:84.

11. Bolognia JJ, Jorizzo JL, Rapini RP. Dermatology. Philadelphia: Elsevier Saunders; 2012.

12. Xie J, Murone M, Luoh SM, Ryan A, Gu Q, et al. Activating smoothened mutations in sporadic basal-cell carcinoma. Nature 1998;391:90-2.

13. Marzuka AG, Book SE. Basal cell carcinoma: pathogenesis, epidemiology, clinical features, diagnosis, histopathology, and management. Yale J Biol Med 2015;88:167-79.

14. Gutzmer R, Solomon JA. Hedgehog pathway inhibition for the treatment of basal cell carcinoma. Target Oncol 2019;14:253-67.

15. Pala R, Alomari N, Nauli SM. Primary cilium-dependent signaling mechanisms. Int J Mol Sci 2017;18:2272.

16. Hutchin ME, Kariapper MS, Grachtchouk M, Wang A, Wei L, et al. Sustained hedgehog signaling is required for basal cell carcinoma proliferation and survival: conditional skin tumorigenesis recapitulates the hair growth cycle. Genes Dev 2005;19:214-23.

17. Pontén F, Berg C, Ahmadian A, Ren ZP, Nistér M, et al. Molecular pathology in basal cell cancer with p53 as a genetic marker. Oncogene 1997;15:1059-67.

18. Sekulic A, Migden MR, Oro AE, Dirix L, Lewis KD, et al. Efficacy and safety of vismodegib in advanced basal-cell carcinoma. N Engl J Med 2012;366:2171-9.

19. Kong JH, Siebold C, Rohatgi R. Biochemical mechanisms of vertebrate hedgehog signaling. Development (Cambridge, England) 2019;146:dev166892.

20. Bakkenist CJ, Kastan MB. DNA damage activates ATM through intermolecular autophosphorylation and dimer dissociation. Nature 2003;421:499-506.

21. Jazayeri A, Falck J, Lukas C, Bartek J, Smith GC, et al. ATM- and cell cycle-dependent regulation of ATR in response to DNA double-strand breaks. Nat Cell Biol 2006;8:37-45.

22. Shen T, Huang S. The role of Cdc25A in the regulation of cell proliferation and apoptosis. Anticancer Agents Med Chem 2012;12:631-9.

23. Decraene D, Agostinis P, Pupe A, de Haes P, Garmyn M. Acute response of human skin to solar radiation: regulation and function of the p53 protein. J Photochem Photobiol B 2001;63:78-83.

24. Liu P, Gan W, Chin YR, Ogura K, Guo J, et al. PtdIns(3,4,5)P3-dependent activation of the mTORC2 kinase complex. Cancer Discov 2015;5:1194-209.

25. Zhang Y, Hunter T. Roles of Chk1 in cell biology and cancer therapy. Int J Cancer 2014;134:1013-23.

26. Xiong Y, Hannon GJ, Zhang H, Casso D, Kobayashi R, et al. p21 is a universal inhibitor of cyclin kinases. Nature 1993;366:701-4.

27. Rezvani HR, Mazurier F, Cario-André M, Pain C, Ged C, et al. Protective effects of catalase overexpression on UVB-induced apoptosis in normal human keratinocytes. J Biol Chem 2006;281:17999-8007.

28. Sander CS, Hamm F, Elsner P, Thiele JJ. Oxidative stress in malignant melanoma and non-melanoma skin cancer. Br J Dermatol 2003;148:913-22.

29. Zhang J, Bowden GT. Activation of p38 MAP kinase and JNK pathways by UVA irradiation. Photochem Photobiol Sci 2012;11:54-61.

30. Nadeau PJ, Charette SJ, Toledano MB, Landry J. Disulfide bond-mediated multimerization of Ask1 and its reduction by thioredoxin-1 regulate H(2)O(2)-induced c-Jun NH(2)-terminal kinase activation and apoptosis. Mol Biol Cell 2007;18:3903-13.

31. Gross S, Knebel A, Tenev T, Neininger A, Gaestel M, et al. Inactivation of protein-tyrosine phosphatases as mechanism of UV-induced signal transduction. J Biol Chem 1999;274:26378-86.

32. Reinhardt HC, Aslanian AS, Lees JA, Yaffe MB. p53-deficient cells rely on ATM- and ATR-mediated checkpoint signaling through the p38MAPK/MK2 pathway for survival after DNA damage. Cancer Cell 2007;11:175-89.

33. Hess J, Angel P, Schorpp-Kistner M. AP-1 subunits: quarrel and harmony among siblings. J Cell Sci 2004;117:5965-73.

34. Silvers AL, Finch JS, Bowden GT. Inhibition of UVA-induced c-Jun N-terminal kinase activity results in caspase-dependent apoptosis in human keratinocytes. Photochem Photobiol 2006;82:423-31.

35. Cooper SJ, MacGowan J, Ranger-Moore J, Young MR, Colburn NH, et al. Expression of dominant negative c-jun inhibits ultraviolet B-induced squamous cell carcinoma number and size in an SKH-1 hairless mouse model. Mol Cancer Res 2003;1:848-54.

36. Strozyk E, Kulms D. The role of AKT/mTOR pathway in stress response to UV-irradiation: implication in skin carcinogenesis by regulation of apoptosis, autophagy and senescence. Int J Mol Sci 2013;14:15260-85.

37. Coffer PJ, Burgering BM, Peppelenbosch MP, Bos JL, Kruijer W. UV activation of receptor tyrosine kinase activity. Oncogene 1995;11:561-9.

38. Arora A, Scholar EM. Role of tyrosine kinase inhibitors in cancer therapy. J Pharmacol Exp Ther 2005;315:971-9.

39. Spallone G, Botti E, Costanzo A. Targeted therapy in nonmelanoma skin cancers. Cancers (Basel) 2011;3:2255-73.

40. Laplante M, Sabatini DM. mTOR signaling in growth control and disease. Cell 2012;149:274-93.

41. Hafner C, Landthaler M, Vogt T. Activation of the PI3K/AKT signalling pathway in non-melanoma skin cancer is not mediated by oncogenic PIK3CA and AKT1 hotspot mutations. Exp Dermatol 2010;19:e222-7.

42. Ming M, He YY. PTEN: new insights into its regulation and function in skin cancer. J Invest Dermatol 2009;129:2109-12.

43. Hemmings BA, Restuccia DF. PI3K-PKB/Akt pathway. Cold Spring Harb Perspect Biol 2012;4:a011189.

44. Shimobayashi M, Hall MN. Making new contacts: the mTOR network in metabolism and signalling crosstalk. Nat Rev Mol Cell Biol 2014;15:155-62.

45. Gan X, Wang J, Su B, Wu D. Evidence for direct activation of mTORC2 kinase activity by phosphatidylinositol 3,4,5-trisphosphate. J Biol Chem 2011;286:10998-1002.

46. Tu Y, Ji C, Yang B, Yang Z, Gu H, et al. DNA-dependent protein kinase catalytic subunit (DNA-PKcs)-SIN1 association mediates ultraviolet B (UVB)-induced Akt Ser-473 phosphorylation and skin cell survival. Mol Cancer 2013;12:172.

47. Greer EL, Brunet A. FOXO transcription factors at the interface between longevity and tumor suppression. Oncogene 2005;24:7410-25.

48. Daitoku H, Fukamizu A. FOXO transcription factors in the regulatory networks of longevity. J Biochem 2007;141:769-74.

49. Bivik C, Ollinger K. JNK mediates UVB-induced apoptosis upstream lysosomal membrane permeabilization and Bcl-2 family proteins. Apoptosis 2008;13:1111-20.

50. Qin JZ, Bacon P, Panella J, Sitailo LA, Denning MF, et al. Low-dose UV-radiation sensitizes keratinocytes to TRAIL-induced apoptosis. J Cell Physiol 2004;200:155-66.

51. Kim DJ, Kataoka K, Sano S, Connolly K, Kiguchi K, et al. Targeted disruption of Bcl-xL in mouse keratinocytes inhibits both UVB- and chemically induced skin carcinogenesis. Mol Carcinog 2009;48:873-85.

52. Obsil T, Ghirlando R, Anderson DE, Hickman AB, Dyda F. Two 14-3-3 binding motifs are required for stable association of Forkhead transcription factor FOXO4 with 14-3-3 proteins and inhibition of DNA binding. Biochemistry 2003;42:15264-72.

53. Dhanasekaran DN, Reddy EP. JNK signaling in apoptosis. Oncogene 2008;27:6245-51.

54. Wellbrock C, Karasarides M, Marais R. The RAF proteins take centre stage. Nat Rev Mol Cell Biol 2004;5:875-85.

55. McCubrey JA, Steelman LS, Chappell WH, Abrams SL, Wong EW, et al. Roles of the Raf/MEK/ERK pathway in cell growth, malignant transformation and drug resistance. Biochim Biophys Acta 2007;1773:1263-84.

56. Omran OM, Ata HS. Expression of tumor necrosis factor-related apoptosis-inducing ligand death receptors DR4 and DR5 in human nonmelanoma skin cancer. Am J Dermatopathol 2014;36:710-7.

57. Shimizu T, Tolcher AW, Papadopoulos KP, Beeram M, Rasco DW, et al. The clinical effect of the dual-targeting strategy involving PI3K/AKT/mTOR and RAS/MEK/ERK pathways in patients with advanced cancer. Clin Cancer Res 2012;18:2316-25.

58. Paul G, Marchelletta RR, McCole DF, Barrett KE. Interferon-γ alters downstream signaling originating from epidermal growth factor receptor in intestinal epithelial cells: functional consequences for ion transport. J Biol Chem 2012;287:2144-55.

59. Chaisuparat R, Limpiwatana S, Kongpanitkul S, Yodsanga S, Jham BC. The Akt/mTOR pathway is activated in verrucous carcinoma of the oral cavity. J Oral Pathol Med 2016;45:581-5.

60. Kim C, Pasparakis M. Epidermal p65/NF-κB signalling is essential for skin carcinogenesis. EMBO Mol Med 2014;6:970-83.

61. Hoesel B, Schmid JA. The complexity of NF-κB signaling in inflammation and cancer. Mol Cancer 2013;12:86.

62. Carpenter RL, Lo HW. STAT3 target genes relevant to human cancers. Cancers (Basel) 2014;6:897-925.

63. Wilson NS, Dixit V, Ashkenazi A. Death receptor signal transducers: nodes of coordination in immune signaling networks. Nat Immunol 2009;10:348-55.

64. Gordon R. Skin cancer: an overview of epidemiology and risk factors. Semin Oncol Nurs 2013;29:160-9.

65. Burton KA, Ashack KA, Khachemoune A. Cutaneous squamous cell carcinoma: a review of high-risk and metastatic disease. Am J Clin Dermatol 2016;17:491-508.

66. Immunotherapy drug cemiplimab approved for advanced squamous cell skin cancer. Available from: https://www.cancer.gov/news-events/cancer-currents-blog/2018/cemiplimab-fda-squamous-cell-carcinoma. [Last accessed on 7 Sep 2020].

67. Benjamin CL, Ananthaswamy HN. p53 and the pathogenesis of skin cancer. Toxicol Appl Pharmacol 2007;224:241-8.

68. Kang D, Choi TH, Han K, Son D, Kim JH, et al. Regulation of K(+) channels may enhance wound healing in the skin. Med Hypotheses 2008;71:927-9.

69. Liu Q, Yu S, Zhao W, Qin S, Chu Q, et al. EGFR-TKIs resistance via EGFR-independent signaling pathways. Mol Cancer 2018;17:53.

70. Wenczak BA, Lynch JB, Nanney LB. Epidermal growth factor receptor distribution in burn wounds. Implications for growth factor-mediated repair. J Clin Invest 1992;90:2392-401.

71. Heo JS, Lee MY, Han HJ. Sonic hedgehog stimulates mouse embryonic stem cell proliferation by cooperation of Ca2+/protein kinase C and epidermal growth factor receptor as well as Gli1 activation. Stem Cells 2007;25:3069-80.

72. Carballo GB, Honorato JR, de Lopes GPF, Spohr TCLdSE. A highlight on Sonic hedgehog pathway. Cell Communication and Signaling 2018;16:11.

73. Panelos J, Tarantini F, Paglierani M, Di Serio C, Maio V, et al. Photoexposition discriminates Notch 1 expression in human cutaneous squamous cell carcinoma. Mod Pathol 2008;21:316-25.

74. Yang L, XL, Brooks YS. Dysregulated estrogen signaling through CYP1B1 contributes to notch deficiency in squamous cell carcinoma. Society of investigative dermatology: youtube presentation; 2020. Available from: https://www.youtube.com/watch?v=nxpbcf1xdca. [Last accessed on 7 Sep 2020].

75. Al Labban D, Jo SH, Ostano P, Saglietti C, Bongiovanni M, et al. Notch-effector CSL promotes squamous cell carcinoma by repressing histone demethylase KDM6B. J Clin Invest 2018;128:2581-99.

76. Konicke K, López-Luna A, Muñoz-Carrillo JL, Servín-González LS, Flores-de la Torre A, et al. The microRNA landscape of cutaneous squamous cell carcinoma. Drug Discov Today 2018;23:864-70.

77. Kollias N, Ruvolo E, Sayre RM. The value of the ratio of UVA to UVB in sunlight. Photochem Photobiol 2011;87:1474-5.

78. Mahler V. Skin diseases associated with environmental factors. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2017;60:605-17.

79. Agar N, Young AR. Melanogenesis: a photoprotective response to DNA damage? Mutat Res 2005;571:121-32.

80. Hung KF, Sidorova JM, Nghiem P, Kawasumi M. The 6-4 photoproduct is the trigger of UV-induced replication blockage and ATR activation. Proc Natl Acad Sci U S A 2020;117:12806-16.

81. Center V-IC. Cell cycle control: overview. Cell Cycle Control. Vanderbilt University; 2016.

82. Ra SH, Su A, Li X, Zhou J, Cochran AJ, et al. Keratoacanthoma and squamous cell carcinoma are distinct from a molecular perspective. Mod Pathol 2015;28:799-806.

83. Atasoy M, Anadolu-Braise R, Pirim I, Dogan H, Ikbal M. HLA antigen profile differences in patients with SCC (Squamous Cell Carcinoma) in-situ/actinic keratosis and invasive SCC: is there a genetic succeptibility for invasive SCC development? Eurasian J Med 2009;41:162-4.

84. Berhane T, Halliday GM, Cooke B, Barnetson RS. Inflammation is associated with progression of actinic keratoses to squamous cell carcinomas in humans. Br J Dermatol 2002;146:810-5.

85. Maru GB, Gandhi K, Ramchandani A, Kumar G. The role of inflammation in skin cancer. Adv Exp Med Biol 2014;816:437-69.

86. Yu H, Pardoll D, Jove R. STATs in cancer inflammation and immunity: a leading role for STAT3. Nat Rev Cancer 2009;9:798-809.

87. Scola N, Skrygan M, Wieland U, Kreuter A, Gambichler T. Altered gene expression in squamous cell carcinoma arising from congenital unilateral linear porokeratosis. Clin Exp Dermatol 2012;37:781-5.

88. Pi J, Diwan BA, Sun Y, Liu J, Qu W, et al. Arsenic-induced malignant transformation of human keratinocytes: involvement of Nrf2. Free Radic Biol Med 2008;45:651-8.

89. Colman JA. Arsenic and uranium in water from private wells completed in bedrock of east-central Massachusetts-Concentrations, correlations with bedrock units, and estimated probability maps. Sci Invest Rep 2011. doi: 10.3133/sir20115013

90. Li C, Srivastava RK, Elmets CA, Afaq F, Athar M. Arsenic-induced cutaneous hyperplastic lesions are associated with the dysregulation of Yap, a Hippo signaling-related protein. Biochem Biophys Res Commun 2013;438:607-12.

91. Poulalhon N, Dalle S, Balme B, Thomas L. Fast-growing cutaneous squamous cell carcinoma in a patient treated with vismodegib. Dermatology 2015;230:101-4.

92. Nissinen L, Farshchian M, Riihilä P, Kähäri VM. New perspectives on role of tumor microenvironment in progression of cutaneous squamous cell carcinoma. Cell Tissue Res 2016;365:691-702.

93. Zgraggen S, Huggenberger R, Kerl K, Detmar M. An important role of the SDF-1/CXCR4 axis in chronic skin inflammation. PLoS One 2014;9:e93665.

94. Sinha S, Su S, Workentine M, Agabalyan N, Cheng M, et al. Transcriptional analysis reveals evidence of chronically impeded ECM turnover and epithelium-to-mesenchyme transition in scar tissue giving rise to marjolin’s ulcer. J Burn Care Res 2017;38:e14-22.

95. Peters FS, Peeters AMA, Mandaviya PR, van Meurs JBJ, Hofland LJ, et al. Differentially methylated regions in T cells identify kidney transplant patients at risk for de novo skin cancer. Clin Epigenetics 2018;10:81.

96. Wysong A, Newman JG, Covington KR, Kurley SJ, Ibrahim SF, et al. Validation of a 40-gene expression profile test to predict metastatic risk in localized high-risk cutaneous squamous cell carcinoma. J Am Acad Dermatol 2020. doi: 10.1016/j.jaad.2020.04.088

97. Costache M, Desa LT, Mitrache LE, Pătraşcu OM, Dumitru A, et al. Cutaneous verrucous carcinoma - report of three cases with review of literature. Rom J Morphol Embryol 2014;55:383-8.

98. Schell BJ, Rosen T, Rády P, Arany I, Tschen JA, et al. Verrucous carcinoma of the foot associated with human papillomavirus type 16. J Am Acad Dermatol 2001;45:49-55.

99. Murao K, Kubo Y, Fukumoto D, Matsumoto K, Arase S. Verrucous carcinoma of the scalp associated with human papillomavirus type 33. Dermatol Surg 2005;31:1363-5.

100. Fujita S, Senba M, Kumatori A, Hayashi T, Ikeda T, et al. Human papillomavirus infection in oral verrucous carcinoma: genotyping analysis and inverse correlation with p53 expression. Pathobiology 2008;75:257-64.

101. Ren B. E2F integrates cell cycle progression with DNA repair, replication, and G2/M checkpoints. Genes Dev 2002;16:245-56.

102. Yim EK, Park JS. The role of HPV E6 and E7 oncoproteins in HPV-associated cervical carcinogenesis. Cancer Res Treat 2005;37:319.

103. Pătraşcu V, Geoloaica L, Ciurea R. Case report acral verrucous carcinoma. Curr Health Sci J 2019;45:235-40.

104. Schumann H, Roth W, Has C, Volz A, Erfurt-Berge C, et al. Verrucous carcinoma in epidermolysis bullosa simplex is possibly associated with a novel mutation in the keratin 5 gene. Br J Dermatol 2012;167:929-36.

105. Jung H, Seong HA, Ha H. Critical role of cysteine residue 81 of macrophage migration inhibitory factor (MIF) in MIF-induced inhibition of p53 activity. J Biol Chem 2008;283:20383-96.

106. Coulombe PA, Lee CH. Defining keratin protein function in skin epithelia: epidermolysis bullosa simplex and its aftermath. J Invest Dermatol 2012;132:763-75.

107. Bolling MC, Lemmink HH, Jansen GHL, Jonkman MF. Mutations in KRT5 and KRT14 cause epidermolysis bullosa simplex in 75% of the patients. Br J Dermatol 2011;164:637-44.

108. Porta C, Paglino C, Mosca A. Targeting PI3K/Akt/mTOR signaling in cancer. Front Oncol 2014;4.

109. Deng Z, Wang Y, Fang X, Yan F, Pan H, et al. Research on miRNA-195 and target gene CDK6 in oral verrucous carcinoma. Cancer Gene Ther 2017;24:282-8.

110. Deng Z, Wang Y, Fang X, Yan F, Pan H, et al. Research on miRNA-195 and target gene CDK6 in oral verrucous carcinoma. Cancer Gene Ther 2017;24.

111. Adegboyega PA, Boromound N, Freeman DH. Diagnostic utility of cell cycle and apoptosis regulatory proteins in verrucous squamous carcinoma. Appl Immunohistochem Mol Morphol 2005;13:171-7.

112. Kusume T, Tsuda H, Kawabata M, Inoue T, Umesaki N, et al. The p16-Cyclin D1/CDK4-pRb pathway and clinical outcome in epithelial ovarian cancer. Clin Cancer Res 1999;5.

113. Al-Mohanna MA, Manogaran PS, Al-Mukhalafi Z, A Al-Hussein K, Aboussekhra A. The tumor suppressor p16INK4a gene is a regulator of apoptosis induced by ultraviolet light and cisplatin. Oncogene 2004;23:201-12.

114. Bruno S, Darzynkiewicz Z. Cell cycle dependent expression and stability of the nuclear protein detected by Ki-67 antibody in HL-60 cells. Cell Proliferation 1992;25:31-40.

115. Mallick S, Breta M, Gupta SD, Dinda AK, Mohanty BK, et al. Angiogenesis, proliferative activity and DNA ploidy in oral verrucous carcinoma: a comparative study including verrucous hyperplasia and squamous cell carcinoma. Pathol Oncol Res 2015;21:1249-57.

116. Yang G, Rosen DG, Liu G, Yang F, Guo X, et al. CXCR2 promotes ovarian cancer growth through dysregulated cell cycle, diminished apoptosis, and enhanced angiogenesis. Clin Cancer Res 2010;16:3875-86.

117. Coussens LM, Werb Z. Inflammation and cancer. Nature 2002;420:860-7.

118. Langowski JL, Zhang X, Wu L, Mattson JD, Chen T, et al. IL-23 promotes tumour incidence and growth. Nature 2006;442:461-5.

119. Tchernev G, Guarneri C, Bevelacqua V, Wollina U. Carcinoma cuniculatum in course of etanercept: blocking autoimmunity but propagation of carcinogenesis? Int J Immunopathol Pharmacol 2014;27:261-6.

120. Chen Y, Sun J, Yang Y, Huang Y, Liu G. Malignancy risk of anti-tumor necrosis factor alpha blockers: an overview of systematic reviews and meta-analyses. Clin Rheumatol 2016;35:1-18.

121. Garcia C, Poletti E, Crowson AN. Basosquamous carcinoma. J Am Acad Dermatol 2009;60:137-43.

122. Basosquamous Cell Cancer. In: Hoffman H, editor. Iowa Head and Neck Protocols: University of Iowa Health Care; 2017.

123. Akay BN, Saral S, Heper AO, Erdem C, Rosendahl C. Basosquamous carcinoma: Dermoscopic clues to diagnosis. J Dermatol 2017;44:127-34.

124. Anand RL, Collins D, Chapman A. Basosquamous carcinoma: appearance and reality. Oxf Med Case Rep 2017;2017.

125. Tarapore E, Atwood SX. Defining the genetics of basosquamous carcinoma. J Invest Dermatol 2019;139:2258-60.

126. Murthaiah P, Truskinovsky A, Shah S, Dudek A. Collision tumor versus multiphenotypic differentiation: a case of carcinoma with features of colonic and lung primary tumors. Anticancer Res 2009;29:1495-7.

127. Lara F, Santamaría JR, Garbers LM. Recurrence rate of basal cell carcinoma with positive histopathological margins and related risk factors. An Bras Dermatol 2017;92:58-62.

128. Oldbury J, Wain R, Abas S, Dobson C, Iyer S. Basosquamous carcinoma: a single centre clinicopathological evaluation and proposal of an evidence-based protocol. J Skin Cancer 2018;2018.

129. Piva de Freitas P, Senna CG, Tabai M, Chone CT, Altemani A. Metastatic basal cell carcinoma: a rare manifestation of a common disease. Case Rep Med 2017;2017.

130. Major A, Anderson M. Not just skin deep: distant metastases from cutaneous squamous cell carcinoma. Am J Med 2017;130.

131. Alam M. Basal cell carcinoma recurrence after mohs surgery. U.S. National Laboratory of Medicine; 2012.

132. Metastatic Squamous Cell Carcinoma. Transplant Skin Cancer Network: University of California San Francisco. Available from: https://skincancer.ucsf.edu/metastatic-squamous-cell-carcinoma. [Last accessed on 7 Sep 2020].

133. van Lee CB, Roorda BM, Wakkee M, Voorham Q, Mooyaart AL, et al. Recurrence rates of cutaneous squamous cell carcinoma of the head and neck after Mohs micrographic surgery vs. standard excision: a retrospective cohort study. Br J Dermatol 2019;181:338-43.

134. Martin RC 2nd, Edwards MJ, Cawte TG, Sewell CL, McMasters KM. Basosquamous carcinoma: analysis of prognostic factors influencing recurrence. Cancer 2000;88:1365-9.

135. Chiang A, Tan CZ, Kuonen F, Hodgkinson LM, Chiang F, et al. Genetic mutations underlying phenotypic plasticity in basosquamous carcinoma. J Invest Dermatol 2019;139:2263-71.e5.

136. Zhao X, Ponomaryov T, Ornell KJ, Zhou P, Dabral SK, et al. RAS/MAPK activation drives resistance to smo inhibition, metastasis, and tumor evolution in shh pathway-dependent tumors. Cancer Res 2015;75:3623-35.

137. Osawa H, Ohnishi H, Takano K, Noguti T, Mashima H, et al. Sonic hedgehog stimulates the proliferation of rat gastric mucosal cells through ERK activation by elevating intracellular calcium concentration. BiochemBiophys Res Commun 2006;344:680-7.

138. Solomon JA, Iarrobino A, Shutty B. Calcium dependence of the SHH pathway and theoretical implications in oral treatment of locally advanced or metastatic basal cell carcinomas. Cutaneous Oncol Today 2011:9-13.

139. Ji J, Wernli M, Mielgo A, Buechner SA, Erb P. Fas-ligand gene silencing in basal cell carcinoma tissue with small interfering RNA. Gene Therapy 2005;12:678-84.

140. Hardwick JM, Soane L. Multiple functions of BCL-2 family proteins. Cold Spring Harb Perspect Biol 2013;5.

141. N Sivrikoz O, Kandiloğlu G. The effects of cyclin D1 and Bcl-2 expressıon on aggressive behavior in basal cell and basosquamous carcinoma. Iran J Pathol 2015;10:185-91.

142. Rishikaysh P, Dev K, Diaz D, Qureshi WMS, Filip S, et al. Signaling involved in hair follicle morphogenesis and development. Int J Mol Sci 2014;15:1648-70.

143. Ellis T, Smyth I, Riley E, Bowles J, Adolphe C, et al. Overexpression of sonic hedgehog suppresses embryonic hair follicle morphogenesis. Dev Biol 2003;263:203-15.

144. Kasper M, Jaks V, Are A, Bergström Å, Schwäger A, et al. Wounding enhances epidermal tumorigenesis by recruiting hair follicle keratinocytes. Proc Nat Acad Sci 2011;108:4099-104.

145. Peterson SC, Eberl M, Vagnozzi AN, Belkadi A, Veniaminova NA, et al. Basal cell carcinoma preferentially arises from stem cells within hair follicle and mechanosensory niches. Cell Stem Cell 2015;16:400-12.

146. Gutzmer R, Solomon J. Hedgehog pathway inhibition for the treatment of basal cell carcinoma. Target Oncol 2019;14:253-67.

147. Duman N, Şen Korkmaz N, Erol Z. Host immune responses and peritumoral stromal reactions in different basal cell carcinoma subtypes: histopathological comparison of basosquamous carcinoma and high-risk and low-risk basal cell carcinoma subtypes. Turk J Med Sci 2016;46:28-34.

148. Zainab H, Sultana A,  Shaimaa. Stromal desmoplasia as a possible prognostic indicator in different grades of oral squamous cell carcinoma. J Oral Maxillofac Pathol 2019;23:338-43.

149. Kaporis H, Guttman-Yassky E, Lowes M, Haider A, Fuentes-Dunclan J, et al. Human basal cell carcinoma is associated with Foxp3+ T cells in a Th2 dominant microenvironment. J Invest Dermatol 2007;127:2391-8.

150. Siegel RL, Miller KD, Jemal A. Cancer Statistics. CA Cancer J Clin 2017;67:7-30.

151. Skin Cancer Facts & Statistics. 2020. Available from: https://www.skincancer.org/skin-cancer-information/skin-cancer-facts/. [Last accessed on 7 Sep 2020].

152. Wells GL. Melanoma - Dermatologic Disorders. Merck Manuals Professional Edition 2019. Available from: https://www.msdmanuals.com/professional/dermatologic-disorders/cancers-of-the-skin/melanoma. [Last accessed on 7 Sep 2020].

153. Soufir N. Prevalence of p16 and CDK4 germline mutations in 48 melanoma-prone families in France. The French Familial Melanoma Study Group. Hum Mol Genet 1998;7:209-16.

154. Freedman DM, Sigurdson A, Rao RS, Hauptmann M, Alexander B, et al. Risk of melanoma among radiologic technologists in the United States. Int J Cancer 2003;103:556-62.

155. Miura K, Olsen CM, Rea S, Marsden J, Green AC. Do airline pilots and cabin crew have raised risks of melanoma and other skin cancers? Systematic review and meta-analysis. Br J Dermatol 2019;181:55-64.

156. Kubica AW, Brewer JD. Melanoma in immunosuppressed patients. Mayo Clinic Proceedings 2012;87:991-1003.

157. Budden T, Davey RJ, Vilain RE, Ashton KA, Braye SG, et al. Repair of UVB-induced DNA damage is reduced in melanoma due to low XPC and global genome repair. Oncotarget 2016;7:60940-53.

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

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

160. Palmieri G, Ombra M, Colombino M, Casula M, Sini M, et al. Multiple molecular pathways in melanomagenesis: characterization of therapeutic targets. Front Oncol 2015;5:183.

161. Liu J, Fukunaga-Kalabis M, Li L, Herlyn M. Developmental pathways activated in melanocytes and melanoma. Arch Biochem Biophys 2014;563:13-21.

162. Cancer Genome Atlas Network. Genomic classification of cutaneous melanoma. Cell 2015;161:1681-96.

163. Curtin JA, Fridlyand J, Kageshita T, Patel HN, Busam KJ, et al. Distinct sets of genetic alterations in melanoma. N Engl J Med 2005;353:2135-47.

164. Lopez-Bergami P, Fitchman B, Ronai ZE. Understanding signaling cascades in melanoma. Photochem Photobiol 2008;84:289-306.

165. Kunz M, Vera J. Modelling of protein kinase signaling pathways in melanoma and other cancers. Cancers 2019;11:465.

166. Paluncic J, Kovacevic Z, Jansson PJ, Kalinowski D, Merlot AM, et al. Roads to melanoma: key pathways and emerging players in melanoma progression and oncogenic signaling. Biochim Biophys Acta 2016;1863:770-84.

167. Orouji E, Orouji A, Gaiser T, Larribère L, Gebhardt C, et al. MAP kinase pathway gene copy alterations inNRAS/BRAFwild-type advanced melanoma. Int J Cancer 2016;138:2257-62.

168. Hodis E, Watson IR, Kryukov GV, Arold ST, Imielinski M, et al. A landscape of driver mutations in melanoma. Cell 2012;150:251-63.

169. Chen X, Wu Q, Depeille P, Chen P, Thornton S, et al. RasGRP3 mediates MAPK pathway activation in GNAQ mutant uveal melanoma. Cancer Cell 2017;31:685-96.e6.

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

171. Broussard L, Howland A, Ryu S, Song K, Norris D, et al. Melanoma cell death mechanisms. Chonnam Med J 2018;54:135.

172. Abbaspour Babaei M, Kamalidehghan B, Saleem M, Zaman Huri H, Ahmadipour F. Receptor tyrosine kinase (c-Kit) inhibitors: a potential therapeutic target in cancer cells. Drug Des Devel Ther 2016;10:2443-59.

173. Meng D, Carvajal RD. KIT as an oncogenic driver in melanoma: an update on clinical development. Am J Clin Dermatol 2019;20:315-23.

174. Sakaizawa K, Ashida A, Uchiyama A, Ito T, Fujisawa Y, et al. Clinical characteristics associated with BRAF, NRAS and KIT mutations in Japanese melanoma patients. J Dermatol Sci 2015;80:33-7.

175. Kong Y, Si L, Zhu Y, Xu X, Corless CL, et al. Large-scale analysis of KIT aberrations in Chinese patients with melanoma. Clin Cancer Res 2011;17:1684-91.

176. Avagliano A, Fiume G, Pelagalli A, Sanità G, Ruocco MR, et al. Metabolic plasticity of melanoma cells and their crosstalk with tumor microenvironment. Front Oncol 2020;10:722.

177. Hartman ML, Czyz M. MITF in melanoma: mechanisms behind its expression and activity. Cell Mol Life Sci 2015;72:1249-60.

178. Wellbrock C, Arozarena I. The complexity of the ERK/MAP-kinase pathway and the treatment of melanoma skin cancer. Front cell dev biol 2016;4:33.

179. Buscà R, Berra E, Gaggioli C, Khaled M, Bille K, et al. Hypoxia-inducible factor 1{alpha} is a new target of microphthalmia-associated transcription factor (MITF) in melanoma cells. J Cell Biol 2005;170:49-59.

180. McGill GG, Horstmann M, Widlund HR, Du J, Motyckova G, et al. Bcl2 regulation by the melanocyte master regulator mitf modulates lineage survival and melanoma cell viability. Cell 2002;109:707-18.

181. Šestáková B, Ondrušová L, Vachtenheim J. Cell cycle inhibitor p21/WAF1/CIP1 as a cofactor of MITF expression in melanoma cells. Pigment Cell Melanoma Res 2010;23:238-51.

182. McKibbin T. Melanoma: understanding relevant molecular pathways as well as available and emerging therapies. Am J Manag Care 2015;21:S224-33.

183. Wei SC, Levine JH, Cogdill AP, Zhao Y, Anang NAAS, et al. Distinct cellular mechanisms underlie anti-CTLA-4 and anti-PD-1 checkpoint blockade. Cell 2017;170:1120-33.e17.

184. Sznol M, Chen L. Antagonist antibodies to PD-1 and B7-H1 (PD-L1) in the treatment of advanced human cancer. Clin Cancer Res 2013;19:1021-34.

185. Zhang N, Wang L, Zhu G, Sun DJ, He H, et al. The association between trauma and melanoma in the Chinese population: a retrospective study. J Eur Acad Dermatol Venereol 2013;28.

186. Möhrle M, Häfner HM. Is subungual melanoma related to trauma? Dermatology 2002;204:259-61.

187. Elmore JG, Elder DE, Barnhill RL, Knezevich SR, Longton GM, et al. Concordance and reproducibility of melanoma staging according to the 7th vs 8th edition of the AJCC cancer staging manual. JAMA Network Open 2018;1:e180083.

188. Gastman BR, Gerami P, Kurley SJ, Cook RW, Leachman S, et al. Identification of patients at risk of metastasis using a prognostic 31-gene expression profile in subpopulations of melanoma patients with favorable outcomes by standard criteria. J Am Acad Dermatol 2019;80:149-57.e4.

189. Gerami P, Cook RW, Wilkinson J, Russell MC, Dhillon N, et al. Development of a prognostic genetic signature to predict the metastatic risk associated with cutaneous melanoma. Clin Cancer Res 2015;21:175-83.

190. Gastman BR, Zager JS, Messina JL, Cook RW, Covington KR, et al. Performance of a 31-gene expression profile test in cutaneous melanomas of the head and neck. Head Neck 2019;41:871-9.

191. Greenhaw BN, Covington KR, Kurley SJ, Yeniay Y, Cao NA, et al. Molecular risk prediction in cutaneous melanoma: a meta-analysis of the 31-gene expression profile prognostic test in 1,479 patients. J Am Acad Dermatol 2020. doi: 10.1016/j.jaad.2020.03.053

192. Kovarik CL, Chu EY, Adamson AS. Gene expression profile testing for thin melanoma: evidence to support clinical use remains thin. JAMA Dermatol 2020. doi: 10.1001/jamadermatol.2020.0894

193. Dalal M, Mitchell S, McCloskey C, Zagadailov E, Gautam A. The clinical and humanistic burden of cutaneous T-cell lymphomas and response to conventional and novel therapies: results of a systematic review. Expert Rev Hematol 2020;13:405-19.

194. Bagherani N, Smoller BR. An overview of cutaneous T cell lymphomas. F1000Res 2016;5:1882.

195. Jahan-Tigh RR, Huen AO, Lee GL, Pozadzides JV, Liu P, et al. Hydrochlorothiazide and cutaneous T cell lymphoma. Cancer 2013;119:825-31.

196. Emge DA, Bassett RL, Duvic M, Huen AO. Methicillin-resistant Staphylococcus aureus (MRSA) is an important pathogen in erythrodermic cutaneous T-cell lymphoma (CTCL) patients. Arch Dermatol Res 2020;312:283-8.

197. Pulitzer M. Cutaneous T-cell lymphoma. Clin Lab Med 2017;37:527-46.

198. Rasheed H, Hegazy RA, Gawdat HI, Mehaney DA, Kamel MM, et al. Serum vitamin D and vitamin D receptor gene polymorphism in mycosis fungoides patients: a case control study. PLoS One 2016;11:e0158014.

199. Choi J, Goh G, Walradt T, Hong BS, Bunick CG, et al. Genomic landscape of cutaneous T cell lymphoma. Nat Genet 2015;47:1011-9.

200. Ayhan A, Mao TL, Suryo Rahmanto Y, Zeppernick F, Ogawa H, et al. Increased proliferation in atypical hyperplasia/endometrioid intraepithelial neoplasia of the endometrium with concurrent inactivation of ARID1A and PTEN tumour suppressors. J Pathol Clin Res 2015;1:186-93.

201. Yang L, Rau R, Goodell MA. DNMT3A in haematological malignancies. Nature Reviews Cancer 2015;15:152-65.

202. Gallardo F, Sandoval J, Díaz-Lagares A, Garcia R, D’Altri T, et al. Notch1 pathway activation results from the epigenetic abrogation of notch-related micrornas in mycosis fungoides. J Invest Dermatol 2015;135:3144-52.

203. Dotto GP. Crosstalk of Notch with p53 and p63 in cancer growth control. Nat Rev Cancer 2009;9:587-95.

204. Espinosa L, Cathelin S, D’Altri T, Trimarchi T, Statnikov A, et al. The Notch/Hes1 pathway sustains NF-κB Activation through CYLD repression in T cell leukemia. Cancer Cell 2010;18:268-81.

205. Manfè V, Biskup E, Rosbjerg A, Kamstrup M, Skov AG, et al. miR-122 regulates p53/Akt signalling and the chemotherapy-induced apoptosis in cutaneous T-cell lymphoma. PLoS One 2012;7:e29541.

206. Walia R, Yeung CCS. An update on molecular biology of cutaneous T cell lymphoma. Front Oncol 2020;9:1558.

207. Wahnschaffe L, Braun T, Timonen S, Giri AK, Schrader A, et al. JAK/STAT-activating genomic alterations are a hallmark of T-PLL. Cancers 2019;11:1833.

208. Rawlings JS, Rosler KM, Harrison DA. The JAK/STAT signaling pathway. J Cell Sci 2004;117:1281-3.

209. Ungewickell A, Bhaduri A, Rios E, Reuter J, Lee CS, et al. Genomic analysis of mycosis fungoides and Sézary syndrome identifies recurrent alterations in TNFR2. Nat Genet 2015;47:1056-60.

210. Espinosa L, Cathelin S, D’Altri T, Trimarchi T, Statnikov A, et al. The Notch/Hes1 pathway sustains NF-κB activation through CYLD repression in T cell leukemia. Cancer Cell 2010;18:268-81.

211. Vaqué JP, Gómez-López G, Monsálvez V, Varela I, Martínez N, et al. PLCG1 mutations in cutaneous T-cell lymphomas. Blood 2014;123:2034-43.

212. Pomerantz JL, Denny EM, Baltimore D. CARD11 mediates factor-specific activation of NF-κB by the T cell receptor complex. EMBO J 2002;21:5184-94.

213. Braun FCM, Grabarczyk P, Möbs M, Braun FK, Eberle J, et al. Tumor suppressor TNFAIP3 (A20) is frequently deleted in Sézary syndrome. Leukemia 2011;25:1494-501.

214. McGirt LY, Degesys CA, Johnson VE, Zic JA, Zwerner JP, et al. TOX expression and role in CTCL. J Eur Acad Dermatol Venereol 2016;30:1497-502.

215. Kremer KN, Dinkel BA, Sterner RM, Osborne DG, Jevremovic D, et al. TCR-CXCR4 signaling stabilizes cytokine mRNA transcripts via a PREX1-Rac1 pathway: implications for CTCL. Blood 2017;130:982-94.

216. Tanase C, Popescu I, Enciu AM, Gheorghisan-Galateanu A, Codrici E, et al. Angiogenesis in cutaneous T-cell lymphoma - proteomic approaches (Review). Oncol Lett 2019;17:4060-7.

217. Liszewski W, Naym DG, Biskup E, Gniadecki R. Psoralen with ultraviolet A-induced apoptosis of cutaneous lymphoma cell lines is augmented by type I interferons via the JAK1-STAT1 pathway. Photodermatol Photoimmunol Photomed 2017;33:164-71.

218. Shea L, Mehta-Shah N. Brentuximab vedotin in the treatment of peripheral T cell lymphoma and cutaneous T cell lymphoma. Curr Hematol Malig Rep 2020;15:9-19.

219. Jawed SI, Myskowski PL, Horwitz S, Moskowitz A, Querfeld C. Primary cutaneous T-cell lymphoma (mycosis fungoides and Sézary syndrome): Part II. Prognosis, management, and future directions. J Am Acad Dermatol 2014;70:223.e1-17.

220. Rappaport SM, Smith MT. Environment and disease risks. Science 2010;330:460-1.

221. English DR, Armstrong BK. Identifying people at high risk of cutaneous malignant melanoma: results from a case-control study in Western Australia. Br Med J (Clin Res Ed) 1988;296:1285-8.

222. Rizzi A, Raddadi N, Sorlini C, Nordgrd L, Nielsen KM, et al. The stability and degradation of dietary DNA in the gastrointestinal tract of mammals: implications for horizontal gene transfer and the biosafety of GMOs. Crit Rev Food Sci Nutr 2012;52:142-61.

223. Potter VR. Initiation and promotion in cancer formation: the importance of studies on intercellular communication. Yale J Biol Med 1980;53:367-84.

224. Colon-Echevarria CB, Lamboy-Caraballo R, Aquino-Acevedo AN, Armaiz-Pena GN. Neuroendocrine regulation of tumor-associated immune cells. Front Oncol 2019;9:1077.

Journal of Cancer Metastasis and Treatment
ISSN 2454-2857 (Online) 2394-4722 (Print)

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/