sábado, 26 de octubre de 2019

Genetics of Skin Cancer (PDQ®) 2/3 –Health Professional Version - National Cancer Institute

Genetics of Skin Cancer (PDQ®)–Health Professional Version - National Cancer Institute

National Cancer Institute

Genetics of Skin Cancer (PDQ®)–Health Professional Version





Interventions

Prevention and treatment of skin cancers

A phase III, double-blind, placebo-controlled clinical trial evaluated the effects of oral nicotinamide (vitamin B3) in 386 individuals with a history of at least two NMSCs within 5 years before study enrollment.[231] After 12 months of treatment, those taking nicotinamide 500 mg twice daily had a 30% reduction in the incidence of new SCCs (95% CI, 0%–51%; P = .05). A statistically significant reduction was also seen in actinic keratoses, the precursor skin lesions to SCCs. The rate of new NMSCs was 23% lower in the nicotinamide group (95% CI, 4–38, P = .02) than in the placebo group. No clinically significant differences in adverse events were observed between the two groups, and there was no evidence of benefit after discontinuation of nicotinamide. Of note, this study was not conducted in a population with an identified genetic predisposition to SCC.
Because many of the syndromes described above are rare, few clinical trials have been conducted in these specific populations. However, valuable information has been developed from the clinical management experience related to skin cancer risk and treatment in the XP population. Strict sun avoidance beginning in infancy, use of protective clothing, and close clinical monitoring of the skin are key components to management of XP. Full-body photography of the skin, conjunctivae, and eyelids is recommended to aid in follow-up.[232] Although few studies on treatment of SCC in the XP population have been done, in most cases treatment is similar to what would be recommended for the general population. Actinic keratoses are treated with topical therapies such as 5-fluorouracil (5-FU), cryotherapy with liquid nitrogen, or dermabrasion, whereas cutaneous cancers are generally managed surgically.[233]
Oral isotretinoin has been used as chemoprevention in XP patients with promising results. A small study of daily use of isotretinoin (13-cis retinoic acid; given as 2 mg/kg/day) reduced NMSC incidence by 63% in a small number of people with XP. Toxicities associated with this treatment included mucocutaneous symptoms, abnormalities in liver function tests and triglyceride levels, and musculoskeletal symptoms such as arthralgias, calcifications of tendons and ligaments, and osteoporosis.[234,235] Dose reduction to 0.5 mg/kg/day reduced toxicity and decreased skin cancer frequency in three of seven subjects (43%); increasing the dose to 1 mg/kg/day resulted in decreased skin cancer frequency in three of the four subjects who did not respond at the lower dose.[236] Oral isotretinoin use may be useful as a chemopreventive agent in other hereditary skin cancer syndromes, including basal cell nevus syndrome (BCNS), Rombo syndrome, EB, and epidermodysplasia verruciformis.[237,238]
Topical T4N5 liposome lotion, containing the bacterial enzyme T4 endonuclease V, was also investigated as a chemopreventive agent in a randomized, placebo-controlled trial of 30 XP patients.[239] Although no effect was seen on incidence of SCC, 17.7 fewer actinic keratoses per year were seen in the treatment group. Additionally, 1.6 fewer BCCs per year were observed in patients being treated with this therapy. Both of these results were statistically significant. The risk of BCC was reduced by 47%, which was of borderline statistical significance. No significant adverse effects of this agent were reported. To date, this agent has not been approved for use by the U.S. Food and Drug Administration.
For patients with XP and unresectable SCC, therapy with 5-FU has been investigated. Several treatment methods were used in this prospective study, including topical therapy to the lesions, short systemic infusion with folic acid, and continuous systemic infusion in combination with cisplatin. Topical 5-FU demonstrated some efficacy, but in some cases viable tumor remained in the deeper dermis. The systemic chemotherapy resulted in one complete response and three partial responses in a total of five patients, suggesting that this therapy may be an option for treatment of extensive lesions.[240] A dose reduction of 30% to 50% has been recommended for systemic chemotherapeutic agents in this population because of the increased sensitivity of XP cells.[241]
For patients with EB, wide local excision of SCC with 2 cm margins remains the treatment of choice. Amputation may be considered as an option to reduce disease recurrence, although it is not clear that this has an impact on survival. The role of sentinel lymph node biopsy remains unclear in this population.[238]
Current guidelines recommend that individuals with EB and unresectable SCC be treated with radiation therapy, but the dose may need to be given in smaller fractions in order to decrease the risk of skin desquamation. Systemic therapy with epidermal growth factor receptor antagonists or tyrosine kinase inhibitors may also be considered for individuals with advanced SCC.[238]
For people who have genetic disorders other than XP, data are lacking, but general sun-safety measures remain important. Careful protection of the skin and eyes is the mainstay of prevention in all patients with increased susceptibility to skin cancer. Key points include avoidance of sun exposure at peak hours, protective clothing and lenses, and vigilant use of sunscreen. Avoidance of x-ray therapy has also been advocated for some groups with hereditary skin cancer syndromes, such as those with epidermodysplasia verruciformis.[144] However, XP patients with unresectable skin cancers or internal cancers, such as spinal cord astrocytoma or glioblastomas of the brain, have been treated successfully with standard therapeutic doses of x-ray radiation.[48] Some experts recommend dermatologic evaluation every 6 months and ophthalmologic evaluation at least annually in these high-risk populations. Guidelines for the management of patients with EB recommend skin examinations every 3 to 6 months starting at age 10 years for individuals with the RDEB-sev gen subtype of the disease.[238] For individuals with other subtypes of EB, skin examination every 6 to 12 months starting at age 20 years is recommended in the absence of an established SCC diagnosis. Dental examination every 6 months is also recommended in this population.[238]
For individuals with DEB, wound care is paramount. Use of silver sulfadiazine cream, medical grade honey, and soft silicone dressings can be helpful in these settings. Attention to nutritional status, which may be compromised because of esophageal strictures, iron-deficiency anemia, infection, and inflammation, is another critical consideration for wound healing for these patients. Multivitamin supplementation, often at higher doses than those routinely recommended for the general population, may be warranted.[242]
Bone marrow transplantation has been explored in patients with DEB; however, there is no evidence that this intervention results in a reduction of skin cancer.[243] A double-blind, randomized, placebo-controlled trial of infusion of nonhematopoietic bone marrow stem cells with or without cyclosporine was conducted in 14 patients with recessive DEB. The rationale for this study was that mesenchymal stem cells (MSCs) have the potential to differentiate into dermal fibroblasts, the main expressor of type VII collagen. Seven subjects were randomly assigned to receive MSCs with 5 mg/kg/day of cyclosporine and an additional seven subjects received only MSCs. The number of new blisters and the rate of blister healing were significantly improved in both groups (P = .003 for the number of new blisters in the combination therapy group and P = .004 in the group receiving MSCs only; P < .001 for the rate of blister healing in both groups). However, no difference was seen between the groups.[244]

Future therapies for epidermolysis bullosa

Researchers are taking advantage of recent technological advances to study new strategies for the treatment of dominant and recessive EB.[245-248] Clustered regularly interspaced short palindromic repeats (CRISPR)-associated protein 9 (Cas9) is a technology that can be used to edit DNA. One research group used CRISPR/Cas9 to correct an inherited pathogenic variant in COL7A1 in keratinocytes isolated from a patient with RDEB.[245] Keratinocytes that contained the corrected version of COL7A1 were successfully transplanted onto mice and staining of skin grafts after transplant showed normal skin. Another study used a different approach, retrovirus infection, to introduce normal COL7A1 into keratinocytes from four RDEB patients.[247] The corrected keratinocytes were then assembled into epidermal graft sheets and transplanted onto six wound areas of each of the four patients. The grafts were well tolerated and showed greater healing capabilities than did noncorrected skin after further study. All of these therapies are still in early research stages and have not yet been evaluated in clinical trials.
Level of evidence: None assigned


References
  1. Rogers HW, Weinstock MA, Harris AR, et al.: Incidence estimate of nonmelanoma skin cancer in the United States, 2006. Arch Dermatol 146 (3): 283-7, 2010. [PUBMED Abstract]
  2. American Cancer Society: Cancer Facts and Figures 2019. Atlanta, Ga: American Cancer Society, 2019. Available onlineExit Disclaimer. Last accessed June 7, 2019.
  3. Rogers HW, Weinstock MA, Feldman SR, et al.: Incidence Estimate of Nonmelanoma Skin Cancer (Keratinocyte Carcinomas) in the U.S. Population, 2012. JAMA Dermatol 151 (10): 1081-6, 2015. [PUBMED Abstract]
  4. Flohil SC, van der Leest RJ, Arends LR, et al.: Risk of subsequent cutaneous malignancy in patients with prior keratinocyte carcinoma: a systematic review and meta-analysis. Eur J Cancer 49 (10): 2365-75, 2013. [PUBMED Abstract]
  5. Armstrong BK, Kricker A: The epidemiology of UV induced skin cancer. J Photochem Photobiol B 63 (1-3): 8-18, 2001. [PUBMED Abstract]
  6. 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. Br J Cancer 73 (11): 1447-54, 1996. [PUBMED Abstract]
  7. Gallagher RP, Hill GB, Bajdik CD, et al.: Sunlight exposure, pigmentation factors, and risk of nonmelanocytic skin cancer. II. Squamous cell carcinoma. Arch Dermatol 131 (2): 164-9, 1995. [PUBMED Abstract]
  8. Lindelöf B, Sigurgeirsson B, Tegner E, et al.: PUVA and cancer risk: the Swedish follow-up study. Br J Dermatol 141 (1): 108-12, 1999. [PUBMED Abstract]
  9. Lim JL, Stern RS: High levels of ultraviolet B exposure increase the risk of non-melanoma skin cancer in psoralen and ultraviolet A-treated patients. J Invest Dermatol 124 (3): 505-13, 2005. [PUBMED Abstract]
  10. Karagas MR, Stannard VA, Mott LA, et al.: Use of tanning devices and risk of basal cell and squamous cell skin cancers. J Natl Cancer Inst 94 (3): 224-6, 2002. [PUBMED Abstract]
  11. Guo X, Fujino Y, Ye X, et al.: Association between multi-level inorganic arsenic exposure from drinking water and skin lesions in China. Int J Environ Res Public Health 3 (3): 262-7, 2006. [PUBMED Abstract]
  12. Chen Y, Hall M, Graziano JH, et al.: A prospective study of blood selenium levels and the risk of arsenic-related premalignant skin lesions. Cancer Epidemiol Biomarkers Prev 16 (2): 207-13, 2007. [PUBMED Abstract]
  13. Karagas MR, Stukel TA, Morris JS, et al.: Skin cancer risk in relation to toenail arsenic concentrations in a US population-based case-control study. Am J Epidemiol 153 (6): 559-65, 2001. [PUBMED Abstract]
  14. Schwartz RA: Arsenic and the skin. Int J Dermatol 36 (4): 241-50, 1997. [PUBMED Abstract]
  15. Koh D, Wang H, Lee J, et al.: Basal cell carcinoma, squamous cell carcinoma and melanoma of the skin: analysis of the Singapore Cancer Registry data 1968-97. Br J Dermatol 148 (6): 1161-6, 2003. [PUBMED Abstract]
  16. Asgari MM, Warton EM, Whittemore AS: Family history of skin cancer is associated with increased risk of cutaneous squamous cell carcinoma. Dermatol Surg 41 (4): 481-6, 2015. [PUBMED Abstract]
  17. Halder RM, Bang KM: Skin cancer in blacks in the United States. Dermatol Clin 6 (3): 397-405, 1988. [PUBMED Abstract]
  18. Asuquo ME, Ebughe G: Major dermatological malignancies encountered in the University of Calabar Teaching Hospital, Calabar, southern Nigeria. Int J Dermatol 51 (Suppl 1): 32-6, 36-40, 2012. [PUBMED Abstract]
  19. English DR, Armstrong BK, Kricker A, et al.: Demographic characteristics, pigmentary and cutaneous risk factors for squamous cell carcinoma of the skin: a case-control study. Int J Cancer 76 (5): 628-34, 1998. [PUBMED Abstract]
  20. Kricker A, Armstrong BK, English DR, et al.: Pigmentary and cutaneous risk factors for non-melanocytic skin cancer--a case-control study. Int J Cancer 48 (5): 650-62, 1991. [PUBMED Abstract]
  21. Akgüner M, Barutçu A, Yilmaz M, et al.: Marjolin's ulcer and chronic burn scarring. J Wound Care 7 (3): 121-2, 1998. [PUBMED Abstract]
  22. Friedman R, Hanson S, Goldberg LH: Squamous cell carcinoma arising in a Leishmania scar. Dermatol Surg 29 (11): 1148-9, 2003. [PUBMED Abstract]
  23. Jensen P, Hansen S, Møller B, et al.: Skin cancer in kidney and heart transplant recipients and different long-term immunosuppressive therapy regimens. J Am Acad Dermatol 40 (2 Pt 1): 177-86, 1999. [PUBMED Abstract]
  24. Hartevelt MM, Bavinck JN, Kootte AM, et al.: Incidence of skin cancer after renal transplantation in The Netherlands. Transplantation 49 (3): 506-9, 1990. [PUBMED Abstract]
  25. Lindelöf B, Sigurgeirsson B, Gäbel H, et al.: Incidence of skin cancer in 5356 patients following organ transplantation. Br J Dermatol 143 (3): 513-9, 2000. [PUBMED Abstract]
  26. Krynitz B, Edgren G, Lindelöf B, et al.: Risk of skin cancer and other malignancies in kidney, liver, heart and lung transplant recipients 1970 to 2008--a Swedish population-based study. Int J Cancer 132 (6): 1429-38, 2013. [PUBMED Abstract]
  27. Glover MT, Niranjan N, Kwan JT, et al.: Non-melanoma skin cancer in renal transplant recipients: the extent of the problem and a strategy for management. Br J Plast Surg 47 (2): 86-9, 1994. [PUBMED Abstract]
  28. Kaplan AL, Cook JL: Cutaneous squamous cell carcinoma in patients with chronic lymphocytic leukemia. Skinmed 4 (5): 300-4, 2005 Sep-Oct. [PUBMED Abstract]
  29. Euvrard S, Kanitakis J, Decullier E, et al.: Subsequent skin cancers in kidney and heart transplant recipients after the first squamous cell carcinoma. Transplantation 81 (8): 1093-100, 2006. [PUBMED Abstract]
  30. Herrero JI, España A, D'Avola D, et al.: Subsequent nonmelanoma skin cancer after liver transplantation. Transplant Proc 44 (6): 1568-70, 2012 Jul-Aug. [PUBMED Abstract]
  31. Cantwell MM, Murray LJ, Catney D, et al.: Second primary cancers in patients with skin cancer: a population-based study in Northern Ireland. Br J Cancer 100 (1): 174-7, 2009. [PUBMED Abstract]
  32. Epstein E: Value of follow-up after treatment of basal cell carcinoma. Arch Dermatol 108 (6): 798-800, 1973. [PUBMED Abstract]
  33. Møller R, Nielsen A, Reymann F: Multiple basal cell carcinoma and internal malignant tumors. Arch Dermatol 111 (5): 584-5, 1975. [PUBMED Abstract]
  34. Bergstresser PR, Halprin KM: Multiple sequential skin cancers. The risk of skin cancer in patients with previous skin cancer. Arch Dermatol 111 (8): 995-6, 1975. [PUBMED Abstract]
  35. Robinson JK: Risk of developing another basal cell carcinoma. A 5-year prospective study. Cancer 60 (1): 118-20, 1987. [PUBMED Abstract]
  36. 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 323 (12): 789-95, 1990. [PUBMED Abstract]
  37. Karagas MR, Stukel TA, Greenberg ER, et al.: 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 267 (24): 3305-10, 1992. [PUBMED Abstract]
  38. Helgadottir H, Höiom V, Tuominen R, et al.: CDKN2a mutation-negative melanoma families have increased risk exclusively for skin cancers but not for other malignancies. Int J Cancer 137 (9): 2220-6, 2015. [PUBMED Abstract]
  39. Hussain SK, Sundquist J, Hemminki K: The effect of having an affected parent or sibling on invasive and in situ skin cancer risk in Sweden. J Invest Dermatol 129 (9): 2142-7, 2009. [PUBMED Abstract]
  40. Hemminki K, Zhang H, Czene K: Familial invasive and in situ squamous cell carcinoma of the skin. Br J Cancer 88 (9): 1375-80, 2003. [PUBMED Abstract]
  41. Lindström LS, Yip B, Lichtenstein P, et al.: Etiology of familial aggregation in melanoma and squamous cell carcinoma of the skin. Cancer Epidemiol Biomarkers Prev 16 (8): 1639-43, 2007. [PUBMED Abstract]
  42. Mucci LA, Hjelmborg JB, Harris JR, et al.: Familial Risk and Heritability of Cancer Among Twins in Nordic Countries. JAMA 315 (1): 68-76, 2016. [PUBMED Abstract]
  43. DiGiovanna JJ, Kraemer KH: Shining a light on xeroderma pigmentosum. J Invest Dermatol 132 (3 Pt 2): 785-96, 2012. [PUBMED Abstract]
  44. Kraemer KH, Lee MM, Andrews AD, et al.: The role of sunlight and DNA repair in melanoma and nonmelanoma skin cancer. The xeroderma pigmentosum paradigm. Arch Dermatol 130 (8): 1018-21, 1994. [PUBMED Abstract]
  45. Bradford PT, Goldstein AM, Tamura D, et al.: Cancer and neurologic degeneration in xeroderma pigmentosum: long term follow-up characterises the role of DNA repair. J Med Genet 48 (3): 168-76, 2011. [PUBMED Abstract]
  46. Moussaid L, Benchikhi H, Boukind EH, et al.: [Cutaneous tumors during xeroderma pigmentosum in Morocco: study of 120 patients] Ann Dermatol Venereol 131 (1 Pt 1): 29-33, 2004. [PUBMED Abstract]
  47. Brooks BP, Thompson AH, Bishop RJ, et al.: Ocular manifestations of xeroderma pigmentosum: long-term follow-up highlights the role of DNA repair in protection from sun damage. Ophthalmology 120 (7): 1324-36, 2013. [PUBMED Abstract]
  48. DiGiovanna JJ, Patronas N, Katz D, et al.: Xeroderma pigmentosum: spinal cord astrocytoma with 9-year survival after radiation and isotretinoin therapy. J Cutan Med Surg 2 (3): 153-8, 1998. [PUBMED Abstract]
  49. Robbins JH: Xeroderma pigmentosum complementation group H is withdrawn and reassigned to group D. Hum Genet 88 (2): 242, 1991. [PUBMED Abstract]
  50. Khan SG, Oh KS, Shahlavi T, et al.: Reduced XPC DNA repair gene mRNA levels in clinically normal parents of xeroderma pigmentosum patients. Carcinogenesis 27 (1): 84-94, 2006. [PUBMED Abstract]
  51. Hirai Y, Noda A, Kodama Y, et al.: Increased risk of skin cancer in Japanese heterozygotes of xeroderma pigmentosum group A. J Hum Genet 63 (11): 1181-1184, 2018. [PUBMED Abstract]
  52. Messaoud O, Ben Rekaya M, Cherif W, et al.: Genetic homogeneity of mutational spectrum of group-A xeroderma pigmentosum in Tunisian patients. Int J Dermatol 49 (5): 544-8, 2010. [PUBMED Abstract]
  53. Ben Rekaya M, Messaoud O, Talmoudi F, et al.: High frequency of the V548A fs X572 XPC mutation in Tunisia: implication for molecular diagnosis. J Hum Genet 54 (7): 426-9, 2009. [PUBMED Abstract]
  54. Cartault F, Nava C, Malbrunot AC, et al.: A new XPC gene splicing mutation has lead to the highest worldwide prevalence of xeroderma pigmentosum in black Mahori patients. DNA Repair (Amst) 10 (6): 577-85, 2011. [PUBMED Abstract]
  55. Doubaj Y, Laarabi FZ, Elalaoui SC, et al.: Carrier frequency of the recurrent mutation c.1643_1644delTG in the XPC gene and birth prevalence of the xeroderma pigmentosum in Morocco. J Dermatol 39 (4): 382-4, 2012. [PUBMED Abstract]
  56. Hirai Y, Kodama Y, Moriwaki S, et al.: Heterozygous individuals bearing a founder mutation in the XPA DNA repair gene comprise nearly 1% of the Japanese population. Mutat Res 601 (1-2): 171-8, 2006. [PUBMED Abstract]
  57. Vogelstein B, Knizler K: Nucleotide excision repair syndromes: xeroderma pigmentosum, Cockayne syndrome, and trichothiodystrophy. In: Vogelstein B, Kinzler KW, eds.: The Genetic Basis of Human Cancer. 2nd ed. New York, NY: McGraw-Hill, 2002, pp 211-37.
  58. Kraemer KH, Slor H: Xeroderma pigmentosum. Clin Dermatol 3 (1): 33-69, 1985 Jan-Mar. [PUBMED Abstract]
  59. Moriwaki S, Kraemer KH: Xeroderma pigmentosum--bridging a gap between clinic and laboratory. Photodermatol Photoimmunol Photomed 17 (2): 47-54, 2001. [PUBMED Abstract]
  60. Ben Rekaya M, Laroussi N, Messaoud O, et al.: A founder large deletion mutation in Xeroderma pigmentosum-Variant form in Tunisia: implication for molecular diagnosis and therapy. Biomed Res Int 2014: 256245, 2014. [PUBMED Abstract]
  61. Fassihi H, Sethi M, Fawcett H, et al.: Deep phenotyping of 89 xeroderma pigmentosum patients reveals unexpected heterogeneity dependent on the precise molecular defect. Proc Natl Acad Sci U S A 113 (9): E1236-45, 2016. [PUBMED Abstract]
  62. Amr K, Messaoud O, El Darouti M, et al.: Mutational spectrum of Xeroderma pigmentosum group A in Egyptian patients. Gene 533 (1): 52-6, 2014. [PUBMED Abstract]
  63. Schriver C, Cleaver J, et al., eds.: Xeroderma pigmentosum and cockayne syndrome. In: Cleaver J, Kraemer K, eds.: The Metabolic and Molecular Bases of Inherited Disease. 7th ed. New York, NY: McGraw-Hill Book Co, 1995, pp 4397.
  64. Lambert WC, Gagna CE, Lambert MW: Xeroderma pigmentosum: its overlap with trichothiodystrophy, Cockayne syndrome and other progeroid syndromes. Adv Exp Med Biol 637: 128-37, 2008. [PUBMED Abstract]
  65. Robbins JH, Kraemer KH, Lutzner MA, et al.: Xeroderma pigmentosum. An inherited diseases with sun sensitivity, multiple cutaneous neoplasms, and abnormal DNA repair. Ann Intern Med 80 (2): 221-48, 1974. [PUBMED Abstract]
  66. Weeda G, Eveno E, Donker I, et al.: A mutation in the XPB/ERCC3 DNA repair transcription gene, associated with trichothiodystrophy. Am J Hum Genet 60 (2): 320-9, 1997. [PUBMED Abstract]
  67. Lehmann AR: The xeroderma pigmentosum group D (XPD) gene: one gene, two functions, three diseases. Genes Dev 15 (1): 15-23, 2001. [PUBMED Abstract]
  68. Broughton BC, Berneburg M, Fawcett H, et al.: Two individuals with features of both xeroderma pigmentosum and trichothiodystrophy highlight the complexity of the clinical outcomes of mutations in the XPD gene. Hum Mol Genet 10 (22): 2539-47, 2001. [PUBMED Abstract]
  69. Goudie DR, Yuille MA, Leversha MA, et al.: Multiple self-healing squamous epitheliomata (ESS1) mapped to chromosome 9q22-q31 in families with common ancestry. Nat Genet 3 (2): 165-9, 1993. [PUBMED Abstract]
  70. Goudie DR, D'Alessandro M, Merriman B, et al.: Multiple self-healing squamous epithelioma is caused by a disease-specific spectrum of mutations in TGFBR1. Nat Genet 43 (4): 365-9, 2011. [PUBMED Abstract]
  71. Bose S, Morgan LJ, Booth DR, et al.: The elusive multiple self-healing squamous epithelioma (MSSE) gene: further mapping, analysis of candidates, and loss of heterozygosity. Oncogene 25 (5): 806-12, 2006. [PUBMED Abstract]
  72. Mabula JB, Chalya PL, Mchembe MD, et al.: Skin cancers among Albinos at a University teaching hospital in Northwestern Tanzania: a retrospective review of 64 cases. BMC Dermatol 12: 5, 2012. [PUBMED Abstract]
  73. Luande J, Henschke CI, Mohammed N: The Tanzanian human albino skin. Natural history. Cancer 55 (8): 1823-8, 1985. [PUBMED Abstract]
  74. Knöpfel N, Martín-Santiago A, Del Pozo LJ, et al.: Amelanotic naevoid melanoma in a 16-month-old albino infant. Clin Exp Dermatol 42 (1): 84-88, 2017. [PUBMED Abstract]
  75. Iversen U, Iversen OH: Tumours of the skin. In: Templeton AC, ed.: Tumours in a Tropical Country: A Survey of Uganda, 1964-1968. Berlin, Germany: Springer, 1973, pp 180-99.
  76. Hutton SM, Spritz RA: A comprehensive genetic study of autosomal recessive ocular albinism in Caucasian patients. Invest Ophthalmol Vis Sci 49 (3): 868-72, 2008. [PUBMED Abstract]
  77. Brilliant MH: The mouse p (pink-eyed dilution) and human P genes, oculocutaneous albinism type 2 (OCA2), and melanosomal pH. Pigment Cell Res 14 (2): 86-93, 2001. [PUBMED Abstract]
  78. Sviderskaya EV, Bennett DC, Ho L, et al.: Complementation of hypopigmentation in p-mutant (pink-eyed dilution) mouse melanocytes by normal human P cDNA, and defective complementation by OCA2 mutant sequences. J Invest Dermatol 108 (1): 30-4, 1997. [PUBMED Abstract]
  79. Mauri L, Manfredini E, Del Longo A, et al.: Clinical evaluation and molecular screening of a large consecutive series of albino patients. J Hum Genet 62 (2): 277-290, 2017. [PUBMED Abstract]
  80. Inagaki K, Suzuki T, Shimizu H, et al.: Oculocutaneous albinism type 4 is one of the most common types of albinism in Japan. Am J Hum Genet 74 (3): 466-71, 2004. [PUBMED Abstract]
  81. Mauri L, Barone L, Al Oum M, et al.: SLC45A2 mutation frequency in Oculocutaneous Albinism Italian patients doesn't differ from other European studies. Gene 533 (1): 398-402, 2014. [PUBMED Abstract]
  82. Simeonov DR, Wang X, Wang C, et al.: DNA variations in oculocutaneous albinism: an updated mutation list and current outstanding issues in molecular diagnostics. Hum Mutat 34 (6): 827-35, 2013. [PUBMED Abstract]
  83. Ibarrola-Villava M, Hu HH, Guedj M, et al.: MC1R, SLC45A2 and TYR genetic variants involved in melanoma susceptibility in southern European populations: results from a meta-analysis. Eur J Cancer 48 (14): 2183-91, 2012. [PUBMED Abstract]
  84. Kausar T, Bhatti MA, Ali M, et al.: OCA5, a novel locus for non-syndromic oculocutaneous albinism, maps to chromosome 4q24. Clin Genet 84 (1): 91-3, 2013. [PUBMED Abstract]
  85. Wei AH, Zang DJ, Zhang Z, et al.: Exome sequencing identifies SLC24A5 as a candidate gene for nonsyndromic oculocutaneous albinism. J Invest Dermatol 133 (7): 1834-40, 2013. [PUBMED Abstract]
  86. Morice-Picard F, Lasseaux E, François S, et al.: SLC24A5 mutations are associated with non-syndromic oculocutaneous albinism. J Invest Dermatol 134 (2): 568-571, 2014. [PUBMED Abstract]
  87. Grønskov K, Dooley CM, Østergaard E, et al.: Mutations in c10orf11, a melanocyte-differentiation gene, cause autosomal-recessive albinism. Am J Hum Genet 92 (3): 415-21, 2013. [PUBMED Abstract]
  88. Tomita Y, Miyamura Y: Oculocutaneous albinism and analysis of tyrosinase gene in Japanese patients. Nagoya J Med Sci 61 (3-4): 97-102, 1998. [PUBMED Abstract]
  89. Liu N, Kong XD, Shi HR, et al.: Tyrosinase gene mutations in the Chinese Han population with OCA1. Genet Res (Camb) 96: e14, 2014. [PUBMED Abstract]
  90. FROGGATT P: Albinism in Northern Ireland. Ann Hum Genet 24: 213-38, 1960. [PUBMED Abstract]
  91. McLeod R, Lowry RB: Incidence of albinism in British Columbia (B.C.). Separation by hairbulb test. Clin Genet 9 (1): 77-80, 1976. [PUBMED Abstract]
  92. Martínez-Frías ML, Bermejo E: Prevalence of congenital anomaly syndromes in a Spanish gypsy population. J Med Genet 29 (7): 483-6, 1992. [PUBMED Abstract]
  93. Grønskov K, Ek J, Sand A, et al.: Birth prevalence and mutation spectrum in danish patients with autosomal recessive albinism. Invest Ophthalmol Vis Sci 50 (3): 1058-64, 2009. [PUBMED Abstract]
  94. Chiang PW, Drautz JM, Tsai AC, et al.: A new hypothesis of OCA1B. Am J Med Genet A 146A (22): 2968-70, 2008. [PUBMED Abstract]
  95. Okoro AN: Albinism in Nigeria. A clinical and social study. Br J Dermatol 92 (5): 485-92, 1975. [PUBMED Abstract]
  96. Kagore F, Lund PM: Oculocutaneous albinism among schoolchildren in Harare, Zimbabwe. J Med Genet 32 (11): 859-61, 1995. [PUBMED Abstract]
  97. Lee ST, Nicholls RD, Bundey S, et al.: Mutations of the P gene in oculocutaneous albinism, ocular albinism, and Prader-Willi syndrome plus albinism. N Engl J Med 330 (8): 529-34, 1994. [PUBMED Abstract]
  98. WOOLF CM: ALBINISM AMONG INDIANS IN ARIZONA AND NEW MEXICO. Am J Hum Genet 17: 23-35, 1965. [PUBMED Abstract]
  99. Manga P, Kromberg JG, Box NF, et al.: Rufous oculocutaneous albinism in southern African Blacks is caused by mutations in the TYRP1 gene. Am J Hum Genet 61 (5): 1095-101, 1997. [PUBMED Abstract]
  100. Rundshagen U, Zühlke C, Opitz S, et al.: Mutations in the MATP gene in five German patients affected by oculocutaneous albinism type 4. Hum Mutat 23 (2): 106-10, 2004. [PUBMED Abstract]
  101. Bertolotti A, Lasseaux E, Plaisant C, et al.: Identification of a homozygous mutation of SLC24A5 (OCA6) in two patients with oculocutaneous albinism from French Guiana. Pigment Cell Melanoma Res 29 (1): 104-6, 2016. [PUBMED Abstract]
  102. Mondal M, Sengupta M, Samanta S, et al.: Molecular basis of albinism in India: evaluation of seven potential candidate genes and some new findings. Gene 511 (2): 470-4, 2012. [PUBMED Abstract]
  103. Perry PK, Silverberg NB: Cutaneous malignancy in albinism. Cutis 67 (5): 427-30, 2001. [PUBMED Abstract]
  104. Fukai K, Oh J, Frenk E, et al.: Linkage disequilibrium mapping of the gene for Hermansky-Pudlak syndrome to chromosome 10q23.1-q23.3. Hum Mol Genet 4 (9): 1665-9, 1995. [PUBMED Abstract]
  105. Wildenberg SC, Oetting WS, Almodóvar C, et al.: A gene causing Hermansky-Pudlak syndrome in a Puerto Rican population maps to chromosome 10q2. Am J Hum Genet 57 (4): 755-65, 1995. [PUBMED Abstract]
  106. Anikster Y, Huizing M, White J, et al.: Mutation of a new gene causes a unique form of Hermansky-Pudlak syndrome in a genetic isolate of central Puerto Rico. Nat Genet 28 (4): 376-80, 2001. [PUBMED Abstract]
  107. Suzuki T, Li W, Zhang Q, et al.: Hermansky-Pudlak syndrome is caused by mutations in HPS4, the human homolog of the mouse light-ear gene. Nat Genet 30 (3): 321-4, 2002. [PUBMED Abstract]
  108. Zhang Q, Zhao B, Li W, et al.: Ru2 and Ru encode mouse orthologs of the genes mutated in human Hermansky-Pudlak syndrome types 5 and 6. Nat Genet 33 (2): 145-53, 2003. [PUBMED Abstract]
  109. Li W, Zhang Q, Oiso N, et al.: Hermansky-Pudlak syndrome type 7 (HPS-7) results from mutant dysbindin, a member of the biogenesis of lysosome-related organelles complex 1 (BLOC-1). Nat Genet 35 (1): 84-9, 2003. [PUBMED Abstract]
  110. Morgan NV, Pasha S, Johnson CA, et al.: A germline mutation in BLOC1S3/reduced pigmentation causes a novel variant of Hermansky-Pudlak syndrome (HPS8). Am J Hum Genet 78 (1): 160-6, 2006. [PUBMED Abstract]
  111. Cullinane AR, Curry JA, Carmona-Rivera C, et al.: A BLOC-1 mutation screen reveals that PLDN is mutated in Hermansky-Pudlak Syndrome type 9. Am J Hum Genet 88 (6): 778-87, 2011. [PUBMED Abstract]
  112. Toro J, Turner M, Gahl WA: Dermatologic manifestations of Hermansky-Pudlak syndrome in patients with and without a 16-base pair duplication in the HPS1 gene. Arch Dermatol 135 (7): 774-80, 1999. [PUBMED Abstract]
  113. Dell'Angelica EC, Shotelersuk V, Aguilar RC, et al.: Altered trafficking of lysosomal proteins in Hermansky-Pudlak syndrome due to mutations in the beta 3A subunit of the AP-3 adaptor. Mol Cell 3 (1): 11-21, 1999. [PUBMED Abstract]
  114. Nagle DL, Karim MA, Woolf EA, et al.: Identification and mutation analysis of the complete gene for Chediak-Higashi syndrome. Nat Genet 14 (3): 307-11, 1996. [PUBMED Abstract]
  115. Perou CM, Moore KJ, Nagle DL, et al.: Identification of the murine beige gene by YAC complementation and positional cloning. Nat Genet 13 (3): 303-8, 1996. [PUBMED Abstract]
  116. Barbosa MD, Nguyen QA, Tchernev VT, et al.: Identification of the homologous beige and Chediak-Higashi syndrome genes. Nature 382 (6588): 262-5, 1996. [PUBMED Abstract]
  117. Engle LJ, Kennett RH: Cloning, analysis, and chromosomal localization of myoxin (MYH12), the human homologue to the mouse dilute gene. Genomics 19 (3): 407-16, 1994. [PUBMED Abstract]
  118. Ménasché G, Pastural E, Feldmann J, et al.: Mutations in RAB27A cause Griscelli syndrome associated with haemophagocytic syndrome. Nat Genet 25 (2): 173-6, 2000. [PUBMED Abstract]
  119. Ménasché G, Ho CH, Sanal O, et al.: Griscelli syndrome restricted to hypopigmentation results from a melanophilin defect (GS3) or a MYO5A F-exon deletion (GS1). J Clin Invest 112 (3): 450-6, 2003. [PUBMED Abstract]
  120. Montaudié H, Chiaverini C, Sbidian E, et al.: Inherited epidermolysis bullosa and squamous cell carcinoma: a systematic review of 117 cases. Orphanet J Rare Dis 11 (1): 117, 2016. [PUBMED Abstract]
  121. Vahidnezhad H, Youssefian L, Saeidian AH, et al.: Multigene Next-Generation Sequencing Panel Identifies Pathogenic Variants in Patients with Unknown Subtype of Epidermolysis Bullosa: Subclassification with Prognostic Implications. J Invest Dermatol 137 (12): 2649-2652, 2017. [PUBMED Abstract]
  122. Fine JD, Bruckner-Tuderman L, Eady RA, et al.: Inherited epidermolysis bullosa: updated recommendations on diagnosis and classification. J Am Acad Dermatol 70 (6): 1103-26, 2014. [PUBMED Abstract]
  123. Bruckner-Tuderman L: Hereditary skin diseases of anchoring fibrils. J Dermatol Sci 20 (2): 122-33, 1999. [PUBMED Abstract]
  124. van den Akker PC, Jonkman MF, Rengaw T, et al.: The international dystrophic epidermolysis bullosa patient registry: an online database of dystrophic epidermolysis bullosa patients and their COL7A1 mutations. Hum Mutat 32 (10): 1100-7, 2011. [PUBMED Abstract]
  125. Fine JD: Epidemiology of Inherited Epidermolysis Bullosa Based on Incidence and Prevalence Estimates From the National Epidermolysis Bullosa Registry. JAMA Dermatol 152 (11): 1231-1238, 2016. [PUBMED Abstract]
  126. Fine J, Johnson L, Suchindran C, et al.: Cancer and inherited epidermolysis bullosa. In: Fine J, Bauer E, McGuire J, et al., eds.: Epidermolysis Bullosa; Clinical, Epidemiologic, and Laboratory Advances and the Findings of the National Epidermolysis Bullosa Registry. Baltimore, Md: The Johns Hopkins University Press, 1999, pp 175-92.
  127. Fine JD, Johnson LB, Weiner M, et al.: Chemoprevention of squamous cell carcinoma in recessive dystrophic epidermolysis bullosa: results of a phase 1 trial of systemic isotretinoin. J Am Acad Dermatol 50 (4): 563-71, 2004. [PUBMED Abstract]
  128. Fine JD, Johnson LB, Weiner M, et al.: Cause-specific risks of childhood death in inherited epidermolysis bullosa. J Pediatr 152 (2): 276-80, 2008. [PUBMED Abstract]
  129. van den Akker PC, van Essen AJ, Kraak MM, et al.: Long-term follow-up of patients with recessive dystrophic epidermolysis bullosa in the Netherlands: expansion of the mutation database and unusual phenotype-genotype correlations. J Dermatol Sci 56 (1): 9-18, 2009. [PUBMED Abstract]
  130. Farhi D: Surgical management of epidermolysis bullosa: the importance of a multidisciplinary management. Int J Dermatol 46 (8): 815-6, 2007. [PUBMED Abstract]
  131. Fine JD, Eady RA, Bauer EA, et al.: The classification of inherited epidermolysis bullosa (EB): Report of the Third International Consensus Meeting on Diagnosis and Classification of EB. J Am Acad Dermatol 58 (6): 931-50, 2008. [PUBMED Abstract]
  132. Sawamura D, Nakano H, Matsuzaki Y: Overview of epidermolysis bullosa. J Dermatol 37 (3): 214-9, 2010. [PUBMED Abstract]
  133. Wessagowit V, Ashton GH, Mohammedi R, et al.: Three cases of de novo dominant dystrophic epidermolysis bullosa associated with the mutation G2043R in COL7A1. Clin Exp Dermatol 26 (1): 97-9, 2001. [PUBMED Abstract]
  134. Cserhalmi-Friedman PB, Garzon MC, Guzman E, et al.: Maternal germline mosaicism in dominant dystrophic epidermolysis bullosa. J Invest Dermatol 117 (5): 1327-8, 2001. [PUBMED Abstract]
  135. Cuadrado-Corrales N, Sánchez-Jimeno C, García M, et al.: A prevalent mutation with founder effect in Spanish Recessive Dystrophic Epidermolysis Bullosa families. BMC Med Genet 11: 139, 2010. [PUBMED Abstract]
  136. Ben Brick AS, Laroussi N, Mesrati H, et al.: Mutational founder effect in recessive dystrophic epidermolysis bullosa families from Southern Tunisia. Arch Dermatol Res 306 (4): 405-11, 2014. [PUBMED Abstract]
  137. Ortiz-Urda S, Garcia J, Green CL, et al.: Type VII collagen is required for Ras-driven human epidermal tumorigenesis. Science 307 (5716): 1773-6, 2005. [PUBMED Abstract]
  138. Fine JD: Inherited epidermolysis bullosa. Orphanet J Rare Dis 5: 12, 2010. [PUBMED Abstract]
  139. Aumailley M, Bruckner-Tuderman L, Carter WG, et al.: A simplified laminin nomenclature. Matrix Biol 24 (5): 326-32, 2005. [PUBMED Abstract]
  140. Nakano A, Chao SC, Pulkkinen L, et al.: Laminin 5 mutations in junctional epidermolysis bullosa: molecular basis of Herlitz vs. non-Herlitz phenotypes. Hum Genet 110 (1): 41-51, 2002. [PUBMED Abstract]
  141. Schumann H, Hammami-Hauasli N, Pulkkinen L, et al.: Three novel homozygous point mutations and a new polymorphism in the COL17A1 gene: relation to biological and clinical phenotypes of junctional epidermolysis bullosa. Am J Hum Genet 60 (6): 1344-53, 1997. [PUBMED Abstract]
  142. Fine JD, Mellerio JE: Extracutaneous manifestations and complications of inherited epidermolysis bullosa: part I. Epithelial associated tissues. J Am Acad Dermatol 61 (3): 367-84; quiz 385-6, 2009. [PUBMED Abstract]
  143. Kiritsi D, Kern JS, Schumann H, et al.: Molecular mechanisms of phenotypic variability in junctional epidermolysis bullosa. J Med Genet 48 (7): 450-7, 2011. [PUBMED Abstract]
  144. Majewski S, Jabłońska S: Epidermodysplasia verruciformis as a model of human papillomavirus-induced genetic cancer of the skin. Arch Dermatol 131 (11): 1312-8, 1995. [PUBMED Abstract]
  145. Sterling JC: Human papillomaviruses and skin cancer. J Clin Virol 32 (Suppl 1): S67-71, 2005. [PUBMED Abstract]
  146. Karagas MR, Nelson HH, Sehr P, et al.: Human papillomavirus infection and incidence of squamous cell and basal cell carcinomas of the skin. J Natl Cancer Inst 98 (6): 389-95, 2006. [PUBMED Abstract]
  147. Ramoz N, Rueda LA, Bouadjar B, et al.: Mutations in two adjacent novel genes are associated with epidermodysplasia verruciformis. Nat Genet 32 (4): 579-81, 2002. [PUBMED Abstract]
  148. Mulvihill J, Miller R, Fraumeni J, eds.: Nosology among the neoplastic genedermatoses. In: Mulvihill J, Miller R, Fraumeni J, eds.: Genetics of Human Cancer. New York, NY: Raven Press, 1977, pp 145-67.
  149. Jabłońska S, Orth G, Jarzabek-Chorzelska M, et al.: Twenty-one years of follow-up studies of familial epidermodysplasia verruciformis. Dermatologica 158 (5): 309-27, 1979. [PUBMED Abstract]
  150. McDermott DF, Gammon B, Snijders PJ, et al.: Autosomal dominant epidermodysplasia verruciformis lacking a known EVER1 or EVER2 mutation. Pediatr Dermatol 26 (3): 306-10, 2009 May-Jun. [PUBMED Abstract]
  151. Lazarczyk M, Pons C, Mendoza JA, et al.: Regulation of cellular zinc balance as a potential mechanism of EVER-mediated protection against pathogenesis by cutaneous oncogenic human papillomaviruses. J Exp Med 205 (1): 35-42, 2008. [PUBMED Abstract]
  152. Patel AS, Karagas MR, Pawlita M, et al.: Cutaneous human papillomavirus infection, the EVER2 gene and incidence of squamous cell carcinoma: a case-control study. Int J Cancer 122 (10): 2377-9, 2008. [PUBMED Abstract]
  153. Crequer A, Picard C, Patin E, et al.: Inherited MST1 deficiency underlies susceptibility to EV-HPV infections. PLoS One 7 (8): e44010, 2012. [PUBMED Abstract]
  154. Ramoz N, Taïeb A, Rueda LA, et al.: Evidence for a nonallelic heterogeneity of epidermodysplasia verruciformis with two susceptibility loci mapped to chromosome regions 2p21-p24 and 17q25. J Invest Dermatol 114 (6): 1148-53, 2000. [PUBMED Abstract]
  155. Rosenberg PS, Tamary H, Alter BP: How high are carrier frequencies of rare recessive syndromes? Contemporary estimates for Fanconi Anemia in the United States and Israel. Am J Med Genet A 155A (8): 1877-83, 2011. [PUBMED Abstract]
  156. Rosenberg PS, Greene MH, Alter BP: Cancer incidence in persons with Fanconi anemia. Blood 101 (3): 822-6, 2003. [PUBMED Abstract]
  157. Alter BP: Cancer in Fanconi anemia, 1927-2001. Cancer 97 (2): 425-40, 2003. [PUBMED Abstract]
  158. Puligandla B, Stass SA, Schumacher HR, et al.: Terminal deoxynucleotidyl transferase in Fanconi's anaemia. Lancet 2 (8102): 1263, 1978. [PUBMED Abstract]
  159. Alter BP, Frissora CL, Halpérin DS, et al.: Fanconi's anaemia and pregnancy. Br J Haematol 77 (3): 410-8, 1991. [PUBMED Abstract]
  160. Berger R, Le Coniat M, Schaison G: Chromosome abnormalities in bone marrow of Fanconi anemia patients. Cancer Genet Cytogenet 65 (1): 47-50, 1993. [PUBMED Abstract]
  161. Lebbé C, Pinquier L, Rybojad M, et al.: Fanconi's anaemia associated with multicentric Bowen's disease and decreased NK cytotoxicity. Br J Dermatol 129 (5): 615-8, 1993. [PUBMED Abstract]
  162. Bagby GC, Alter BP: Fanconi anemia. Semin Hematol 43 (3): 147-56, 2006. [PUBMED Abstract]
  163. Chang L, Yuan W, Zeng H, et al.: Whole exome sequencing reveals concomitant mutations of multiple FA genes in individual Fanconi anemia patients. BMC Med Genomics 7: 24, 2014. [PUBMED Abstract]
  164. Wang W: Emergence of a DNA-damage response network consisting of Fanconi anaemia and BRCA proteins. Nat Rev Genet 8 (10): 735-48, 2007. [PUBMED Abstract]
  165. Howlett NG, Taniguchi T, Olson S, et al.: Biallelic inactivation of BRCA2 in Fanconi anemia. Science 297 (5581): 606-9, 2002. [PUBMED Abstract]
  166. Seal S, Thompson D, Renwick A, et al.: Truncating mutations in the Fanconi anemia J gene BRIP1 are low-penetrance breast cancer susceptibility alleles. Nat Genet 38 (11): 1239-41, 2006. [PUBMED Abstract]
  167. Berwick M, Satagopan JM, Ben-Porat L, et al.: Genetic heterogeneity among Fanconi anemia heterozygotes and risk of cancer. Cancer Res 67 (19): 9591-6, 2007. [PUBMED Abstract]
  168. Rafnar T, Sigurjonsdottir GR, Stacey SN, et al.: Association of BRCA2 K3326* With Small Cell Lung Cancer and Squamous Cell Cancer of the Skin. J Natl Cancer Inst 110 (9): 967-974, 2018. [PUBMED Abstract]
  169. Walne AJ, Vulliamy T, Beswick R, et al.: TINF2 mutations result in very short telomeres: analysis of a large cohort of patients with dyskeratosis congenita and related bone marrow failure syndromes. Blood 112 (9): 3594-600, 2008. [PUBMED Abstract]
  170. Alter BP, Giri N, Savage SA, et al.: Cancer in dyskeratosis congenita. Blood 113 (26): 6549-57, 2009. [PUBMED Abstract]
  171. Vulliamy T, Dokal I: Dyskeratosis congenita. Semin Hematol 43 (3): 157-66, 2006. [PUBMED Abstract]
  172. Knight SW, Heiss NS, Vulliamy TJ, et al.: X-linked dyskeratosis congenita is predominantly caused by missense mutations in the DKC1 gene. Am J Hum Genet 65 (1): 50-8, 1999. [PUBMED Abstract]
  173. Vulliamy T, Marrone A, Szydlo R, et al.: Disease anticipation is associated with progressive telomere shortening in families with dyskeratosis congenita due to mutations in TERC. Nat Genet 36 (5): 447-9, 2004. [PUBMED Abstract]
  174. Vulliamy TJ, Walne A, Baskaradas A, et al.: Mutations in the reverse transcriptase component of telomerase (TERT) in patients with bone marrow failure. Blood Cells Mol Dis 34 (3): 257-63, 2005 May-Jun. [PUBMED Abstract]
  175. Vulliamy T, Beswick R, Kirwan M, et al.: Mutations in the telomerase component NHP2 cause the premature ageing syndrome dyskeratosis congenita. Proc Natl Acad Sci U S A 105 (23): 8073-8, 2008. [PUBMED Abstract]
  176. Walne AJ, Vulliamy T, Marrone A, et al.: Genetic heterogeneity in autosomal recessive dyskeratosis congenita with one subtype due to mutations in the telomerase-associated protein NOP10. Hum Mol Genet 16 (13): 1619-29, 2007. [PUBMED Abstract]
  177. Savage SA, Giri N, Baerlocher GM, et al.: TINF2, a component of the shelterin telomere protection complex, is mutated in dyskeratosis congenita. Am J Hum Genet 82 (2): 501-9, 2008. [PUBMED Abstract]
  178. Marrone A, Walne A, Tamary H, et al.: Telomerase reverse-transcriptase homozygous mutations in autosomal recessive dyskeratosis congenita and Hoyeraal-Hreidarsson syndrome. Blood 110 (13): 4198-205, 2007. [PUBMED Abstract]
  179. Ballew BJ, Yeager M, Jacobs K, et al.: Germline mutations of regulator of telomere elongation helicase 1, RTEL1, in Dyskeratosis congenita. Hum Genet 132 (4): 473-80, 2013. [PUBMED Abstract]
  180. Walne AJ, Vulliamy T, Kirwan M, et al.: Constitutional mutations in RTEL1 cause severe dyskeratosis congenita. Am J Hum Genet 92 (3): 448-53, 2013. [PUBMED Abstract]
  181. Fedick AM, Shi L, Jalas C, et al.: Carrier screening of RTEL1 mutations in the Ashkenazi Jewish population. Clin Genet 88 (2): 177-81, 2015. [PUBMED Abstract]
  182. Batista LF, Pech MF, Zhong FL, et al.: Telomere shortening and loss of self-renewal in dyskeratosis congenita induced pluripotent stem cells. Nature 474 (7351): 399-402, 2011. [PUBMED Abstract]
  183. Neveling K, Bechtold A, Hoehn H: Genetic instability syndromes with progeroid features. Z Gerontol Geriatr 40 (5): 339-48, 2007. [PUBMED Abstract]
  184. Zhong F, Savage SA, Shkreli M, et al.: Disruption of telomerase trafficking by TCAB1 mutation causes dyskeratosis congenita. Genes Dev 25 (1): 11-6, 2011. [PUBMED Abstract]
  185. Walne AJ, Vulliamy T, Beswick R, et al.: Mutations in C16orf57 and normal-length telomeres unify a subset of patients with dyskeratosis congenita, poikiloderma with neutropenia and Rothmund-Thomson syndrome. Hum Mol Genet 19 (22): 4453-61, 2010. [PUBMED Abstract]
  186. Colombo EA, Bazan JF, Negri G, et al.: Novel C16orf57 mutations in patients with Poikiloderma with Neutropenia: bioinformatic analysis of the protein and predicted effects of all reported mutations. Orphanet J Rare Dis 7: 7, 2012. [PUBMED Abstract]
  187. Alter BP, Baerlocher GM, Savage SA, et al.: Very short telomere length by flow fluorescence in situ hybridization identifies patients with dyskeratosis congenita. Blood 110 (5): 1439-47, 2007. [PUBMED Abstract]
  188. Vulliamy TJ, Marrone A, Knight SW, et al.: Mutations in dyskeratosis congenita: their impact on telomere length and the diversity of clinical presentation. Blood 107 (7): 2680-5, 2006. [PUBMED Abstract]
  189. Vulliamy TJ, Dokal I: Dyskeratosis congenita: the diverse clinical presentation of mutations in the telomerase complex. Biochimie 90 (1): 122-30, 2008. [PUBMED Abstract]
  190. Borg MF, Olver IN, Hill MP: Rothmund-Thomson syndrome and tolerance of chemoradiotherapy. Australas Radiol 42 (3): 216-8, 1998. [PUBMED Abstract]
  191. Haneke E, Gutschmidt E: Premature multiple Bowen's disease in poikiloderma congenitale with warty hyperkeratoses. Dermatologica 158 (5): 384-8, 1979. [PUBMED Abstract]
  192. Wang LL, Levy ML, Lewis RA, et al.: Clinical manifestations in a cohort of 41 Rothmund-Thomson syndrome patients. Am J Med Genet 102 (1): 11-7, 2001. [PUBMED Abstract]
  193. Piquero-Casals J, Okubo AY, Nico MM: Rothmund-thomson syndrome in three siblings and development of cutaneous squamous cell carcinoma. Pediatr Dermatol 19 (4): 312-6, 2002 Jul-Aug. [PUBMED Abstract]
  194. Petkovic M, Dietschy T, Freire R, et al.: The human Rothmund-Thomson syndrome gene product, RECQL4, localizes to distinct nuclear foci that coincide with proteins involved in the maintenance of genome stability. J Cell Sci 118 (Pt 18): 4261-9, 2005. [PUBMED Abstract]
  195. Wang LL, Gannavarapu A, Kozinetz CA, et al.: Association between osteosarcoma and deleterious mutations in the RECQL4 gene in Rothmund-Thomson syndrome. J Natl Cancer Inst 95 (9): 669-74, 2003. [PUBMED Abstract]
  196. Kitao S, Lindor NM, Shiratori M, et al.: Rothmund-thomson syndrome responsible gene, RECQL4: genomic structure and products. Genomics 61 (3): 268-76, 1999. [PUBMED Abstract]
  197. Werner SR, Prahalad AK, Yang J, et al.: RECQL4-deficient cells are hypersensitive to oxidative stress/damage: Insights for osteosarcoma prevalence and heterogeneity in Rothmund-Thomson syndrome. Biochem Biophys Res Commun 345 (1): 403-9, 2006. [PUBMED Abstract]
  198. Nakayama H: RecQ family helicases: roles as tumor suppressor proteins. Oncogene 21 (58): 9008-21, 2002. [PUBMED Abstract]
  199. Cunniff C, Djavid AR, Carrubba S, et al.: Health supervision for people with Bloom syndrome. Am J Med Genet A 176 (9): 1872-1881, 2018. [PUBMED Abstract]
  200. Ellis NA, Groden J, Ye TZ, et al.: The Bloom's syndrome gene product is homologous to RecQ helicases. Cell 83 (4): 655-66, 1995. [PUBMED Abstract]
  201. Bugreev DV, Yu X, Egelman EH, et al.: Novel pro- and anti-recombination activities of the Bloom's syndrome helicase. Genes Dev 21 (23): 3085-94, 2007. [PUBMED Abstract]
  202. German J, Ellis N: Bloom syndrome. In: Vogelstein B, Kinzler KW, eds.: The Genetic Basis of Human Cancer. 2nd ed. New York, NY: McGraw-Hill, 2002, pp 267-88.
  203. German J, Sanz MM, Ciocci S, et al.: Syndrome-causing mutations of the BLM gene in persons in the Bloom's Syndrome Registry. Hum Mutat 28 (8): 743-53, 2007. [PUBMED Abstract]
  204. Ellis NA, Ciocci S, Proytcheva M, et al.: The Ashkenazic Jewish Bloom syndrome mutation blmAsh is present in non-Jewish Americans of Spanish ancestry. Am J Hum Genet 63 (6): 1685-93, 1998. [PUBMED Abstract]
  205. Lu X, Lane DP: Differential induction of transcriptionally active p53 following UV or ionizing radiation: defects in chromosome instability syndromes? Cell 75 (4): 765-78, 1993. [PUBMED Abstract]
  206. Meetei AR, Sechi S, Wallisch M, et al.: A multiprotein nuclear complex connects Fanconi anemia and Bloom syndrome. Mol Cell Biol 23 (10): 3417-26, 2003. [PUBMED Abstract]
  207. Yamamoto K, Imakiire A, Miyagawa N, et al.: A report of two cases of Werner's syndrome and review of the literature. J Orthop Surg (Hong Kong) 11 (2): 224-33, 2003. [PUBMED Abstract]
  208. Huang S, Lee L, Hanson NB, et al.: The spectrum of WRN mutations in Werner syndrome patients. Hum Mutat 27 (6): 558-67, 2006. [PUBMED Abstract]
  209. Goto M, Miller RW, Ishikawa Y, et al.: Excess of rare cancers in Werner syndrome (adult progeria). Cancer Epidemiol Biomarkers Prev 5 (4): 239-46, 1996. [PUBMED Abstract]
  210. Tsuchiya H, Tomita K, Ohno M, et al.: Werner's syndrome combined with quintuplicate malignant tumors: a case report and review of literature data. Jpn J Clin Oncol 21 (2): 135-42, 1991. [PUBMED Abstract]
  211. Lauper JM, Krause A, Vaughan TL, et al.: Spectrum and risk of neoplasia in Werner syndrome: a systematic review. PLoS One 8 (4): e59709, 2013. [PUBMED Abstract]
  212. Machino H, Miki Y, Teramoto T, et al.: Cytogenetic studies in a patient with porokeratosis of Mibelli, multiple cancers and a forme fruste of Werner's syndrome. Br J Dermatol 111 (5): 579-86, 1984. [PUBMED Abstract]
  213. Goto M, Imamura O, Kuromitsu J, et al.: Analysis of helicase gene mutations in Japanese Werner's syndrome patients. Hum Genet 99 (2): 191-3, 1997. [PUBMED Abstract]
  214. Oshima J, Yu CE, Piussan C, et al.: Homozygous and compound heterozygous mutations at the Werner syndrome locus. Hum Mol Genet 5 (12): 1909-13, 1996. [PUBMED Abstract]
  215. Uhrhammer NA, Lafarge L, Dos Santos L, et al.: Werner syndrome and mutations of the WRN and LMNA genes in France. Hum Mutat 27 (7): 718-9, 2006. [PUBMED Abstract]
  216. Yu CE, Oshima J, Wijsman EM, et al.: Mutations in the consensus helicase domains of the Werner syndrome gene. Werner's Syndrome Collaborative Group. Am J Hum Genet 60 (2): 330-41, 1997. [PUBMED Abstract]
  217. Shen JC, Loeb LA: The Werner syndrome gene: the molecular basis of RecQ helicase-deficiency diseases. Trends Genet 16 (5): 213-20, 2000. [PUBMED Abstract]
  218. Shen J, Loeb LA: Unwinding the molecular basis of the Werner syndrome. Mech Ageing Dev 122 (9): 921-44, 2001. [PUBMED Abstract]
  219. Brosh RM, Bohr VA: Roles of the Werner syndrome protein in pathways required for maintenance of genome stability. Exp Gerontol 37 (4): 491-506, 2002. [PUBMED Abstract]
  220. Furuichi Y: Premature aging and predisposition to cancers caused by mutations in RecQ family helicases. Ann N Y Acad Sci 928: 121-31, 2001. [PUBMED Abstract]
  221. Lebel M: Werner syndrome: genetic and molecular basis of a premature aging disorder. Cell Mol Life Sci 58 (7): 857-67, 2001. [PUBMED Abstract]
  222. Bohr VA, Brosh RM, von Kobbe C, et al.: Pathways defective in the human premature aging disease Werner syndrome. Biogerontology 3 (1-2): 89-94, 2002. [PUBMED Abstract]
  223. Chen L, Oshima J: Werner Syndrome. J Biomed Biotechnol 2 (2): 46-54, 2002. [PUBMED Abstract]
  224. Opresko PL, Cheng WH, von Kobbe C, et al.: Werner syndrome and the function of the Werner protein; what they can teach us about the molecular aging process. Carcinogenesis 24 (5): 791-802, 2003. [PUBMED Abstract]
  225. Pirzio LM, Pichierri P, Bignami M, et al.: Werner syndrome helicase activity is essential in maintaining fragile site stability. J Cell Biol 180 (2): 305-14, 2008. [PUBMED Abstract]
  226. Crabbe L, Jauch A, Naeger CM, et al.: Telomere dysfunction as a cause of genomic instability in Werner syndrome. Proc Natl Acad Sci U S A 104 (7): 2205-10, 2007. [PUBMED Abstract]
  227. Friedrich K, Lee L, Leistritz DF, et al.: WRN mutations in Werner syndrome patients: genomic rearrangements, unusual intronic mutations and ethnic-specific alterations. Hum Genet 128 (1): 103-11, 2010. [PUBMED Abstract]
  228. Moser MJ, Oshima J, Monnat RJ: WRN mutations in Werner syndrome. Hum Mutat 13 (4): 271-9, 1999. [PUBMED Abstract]
  229. Satoh M, Imai M, Sugimoto M, et al.: Prevalence of Werner's syndrome heterozygotes in Japan. Lancet 353 (9166): 1766, 1999. [PUBMED Abstract]
  230. Goto M, Yamabe Y, Shiratori M, et al.: Immunological diagnosis of Werner syndrome by down-regulated and truncated gene products. Hum Genet 105 (4): 301-7, 1999. [PUBMED Abstract]
  231. Chen AC, Martin AJ, Choy B, et al.: A Phase 3 Randomized Trial of Nicotinamide for Skin-Cancer Chemoprevention. N Engl J Med 373 (17): 1618-26, 2015. [PUBMED Abstract]
  232. Tamura D, DiGiovanna JJ, Khan SG, et al.: Living with xeroderma pigmentosum: comprehensive photoprotection for highly photosensitive patients. Photodermatol Photoimmunol Photomed 30 (2-3): 146-52, 2014 Apr-Jun. [PUBMED Abstract]
  233. Tamura D, DiGiovanna JJ, Kraemer KH: Xeroderma pigmentosum. In: Lebwohl MG, Birth-Jones J, Heymann WR, et al., eds.: Treatment of Skin Disease: Comprehensive Therapeutic Strategies. 3rd ed. London, England: Saunders Elsevier, 2010, pp 789-92.
  234. Kraemer KH, DiGiovanna JJ, Moshell AN, et al.: Prevention of skin cancer in xeroderma pigmentosum with the use of oral isotretinoin. N Engl J Med 318 (25): 1633-7, 1988. [PUBMED Abstract]
  235. DiGiovanna JJ: Retinoid chemoprevention in the high-risk patient. J Am Acad Dermatol 39 (2 Pt 3): S82-5, 1998. [PUBMED Abstract]
  236. DiGiovanna J: Oral isotretinoin chemoprevention of skin cancer in xeroderma pigmentosum. J Eur Acad Derm Venereol 5 (Suppl 1): 27, 1995.
  237. Otley CC, Stasko T, Tope WD, et al.: Chemoprevention of nonmelanoma skin cancer with systemic retinoids: practical dosing and management of adverse effects. Dermatol Surg 32 (4): 562-8, 2006. [PUBMED Abstract]
  238. Mellerio JE, Robertson SJ, Bernardis C, et al.: Management of cutaneous squamous cell carcinoma in patients with epidermolysis bullosa: best clinical practice guidelines. Br J Dermatol 174 (1): 56-67, 2016. [PUBMED Abstract]
  239. Yarosh D, Klein J, O'Connor A, et al.: Effect of topically applied T4 endonuclease V in liposomes on skin cancer in xeroderma pigmentosum: a randomised study. Xeroderma Pigmentosum Study Group. Lancet 357 (9260): 926-9, 2001. [PUBMED Abstract]
  240. Boussen H, Zwik J, Mili-Boussen I, et al.: [Therapeutic results of 5-fluorouracil in multiple and unresectable facial carcinoma secondary to xeroderma pigmentosum] Therapie 56 (6): 751-4, 2001 Nov-Dec. [PUBMED Abstract]
  241. Sarasin A: Progress and prospects of xeroderma pigmentosum therapy. Adv Exp Med Biol 637: 144-51, 2008. [PUBMED Abstract]
  242. Mellerio JE, Weiner M, Denyer JE, et al.: Medical management of epidermolysis bullosa: Proceedings of the IInd International Symposium on Epidermolysis Bullosa, Santiago, Chile, 2005. Int J Dermatol 46 (8): 795-800, 2007. [PUBMED Abstract]
  243. Wagner JE, Ishida-Yamamoto A, McGrath JA, et al.: Bone marrow transplantation for recessive dystrophic epidermolysis bullosa. N Engl J Med 363 (7): 629-39, 2010. [PUBMED Abstract]
  244. El-Darouti M, Fawzy M, Amin I, et al.: Treatment of dystrophic epidermolysis bullosa with bone marrow non-hematopoeitic stem cells: a randomized controlled trial. Dermatol Ther 29 (2): 96-100, 2016 Mar-Apr. [PUBMED Abstract]
  245. Hainzl S, Peking P, Kocher T, et al.: COL7A1 Editing via CRISPR/Cas9 in Recessive Dystrophic Epidermolysis Bullosa. Mol Ther 25 (11): 2573-2584, 2017. [PUBMED Abstract]
  246. Shinkuma S, Guo Z, Christiano AM: Site-specific genome editing for correction of induced pluripotent stem cells derived from dominant dystrophic epidermolysis bullosa. Proc Natl Acad Sci U S A 113 (20): 5676-81, 2016. [PUBMED Abstract]
  247. Siprashvili Z, Nguyen NT, Gorell ES, et al.: Safety and Wound Outcomes Following Genetically Corrected Autologous Epidermal Grafts in Patients With Recessive Dystrophic Epidermolysis Bullosa. JAMA 316 (17): 1808-1817, 2016. [PUBMED Abstract]
  248. Webber BR, Osborn MJ, McElroy AN, et al.: CRISPR/Cas9-based genetic correction for recessive dystrophic epidermolysis bullosa. NPJ Regen Med 1: , 2016. [PUBMED Abstract]

Melanoma



Introduction

Rare, high-penetrance and common, low-penetrance genetic factors for melanoma have been identified, and approximately 5% to 10% of all melanomas arise in multiple-case families. However, a significant fraction of these families do not have detectable pathogenic variants in specific susceptibility genes. The frequency with which multiple-case families are ascertained and specific genetic variants are identified differs substantially between populations and geographic regions. A major population-based study has concluded that the high-penetrance susceptibility gene CDKN2A does not make a large contribution to the incidence of melanoma.[1]

Risk Factors for Melanoma

This section focuses on risk factors in individuals at increased hereditary risk of developing melanoma. (Refer to the PDQ summary on Skin Cancer Prevention for information about risk factors for melanoma in the general population.)

Sun exposure

Sun exposure is well established as a major etiologic factor in all forms of skin cancer, although its effects differ by cancer type. The relationship between sun exposure, sunscreen use, and the development of skin cancer is complex. It is complicated by negative confounding (i.e., subjects who are extremely sun sensitive deliberately engage in fewer activities in direct sunlight, and they are more likely to wear sunscreen when they do). These subjects are genetically susceptible to the development of skin cancer by virtue of their cutaneous phenotype and thus may develop skin cancer regardless of the amount of sunlight exposure or the sun protection factor of the sunscreen.[2,3]

Pigmentary characteristics

Pigmentary characteristics are important determinants of melanoma susceptibility. There is an inverse correlation between melanoma risk and skin color that goes from lightest skin to darkest skin. Dark-skinned ethnic groups have a very low risk of melanoma on pigmented skin surfaces; however, individuals in these groups develop melanoma on less-pigmented acral surfaces (palms, soles, nail beds) at the same frequency as light-skinned individuals. Among relatively light-skinned individuals, skin color is modified by genetics and behavior. Melanocortin 1 receptor (MC1R) is one of the major genes controlling pigmentation (refer to the MC1R section of this summary); other pigmentation genes are under investigation.[4]
Clinically, several pigmentary characteristics are evaluated to assess the risk of melanoma and other types of skin cancer. These include the following:
  • Fitzpatrick skin type. The following six skin phenotypes were defined on the basis of response to sun exposure at the beginning of summer.[5]
    1. Type I: Extremely fair skin, always burns, never tans.
    2. Type II: Fair skin, always burns, sometimes tans.
    3. Type III: Medium skin, sometimes burns, always tans.
    4. Type IV: Olive skin, rarely burns, always tans.
    5. Type V: Moderately pigmented brown skin, never burns, always tans.
    6. Type VI: Markedly pigmented black skin, never burns, always tans.
  • Number of nevi or nevus density.
  • Abnormal or atypical nevi.
  • Freckling.

Nevi

Nevi (or moles) are sharply circumscribed benign pigmented lesions of the skin or mucosa composed of nest melanocytes. Patients with multiple nevi demonstrate increased risk of melanoma. While there is evidence that both the presence of multiple nevi and the presence of multiple clinically atypical nevi are associated with an increased risk of melanoma, most studies demonstrate a stronger risk of melanoma with the presence of atypical nevi.[6-9] In addition, patients with multiple atypical nevi, regardless of personal and/or family history of melanoma, are at significantly increased risk of developing melanoma than are patients without atypical nevi.[10] A population-based study in the United Kingdom that identified genetic risk factors for the development of nevi showed that some of the same variants are modestly associated with melanoma risk.[11]
The phenotype of multiple nevi has both familial and environmental affecters. The number of nevi can increase with childhood sun exposure.[12,13] The analysis of this association is complex because the use of sun protection strongly correlates with sun exposure. Inheritance of the specific phenotype of a high number of nevi, including clinically atypical nevi, was initially reported as an autosomal dominant trait under the names dysplastic nevus syndrome [14] and familial atypical multiple mole-melanoma syndrome.[15] A portion of this inherited phenotype is attributed to the major melanoma risk gene CDKN2A discussed below. Even within gene carriers in high-risk families, sun exposure seems to affect nevus number.[16]

Family history

Generally, a family history of melanoma appears to increase risk of melanoma by about twofold. A family cancer registry study assessed over 20,000 individuals with melanoma and found a standardized incidence ratio (SIR) of 2.62 for offspring of individuals with melanoma and 2.94 for siblings.[17] Slightly higher melanoma risks were found in a population-based study of 1,506,961 individuals in Western Australia; first-degree relatives (FDRs) of 5,660 individuals with melanoma showed an HR for melanoma of 3.58 (95% confidence interval [CI], 2.43–5.43).[18] Another population-based study of more than 238,000 FDRs of 23,000 melanoma patients found a lifetime cumulative risk of melanoma of 2.5% to 3%, which is about double the risk of the general population.[19] Risk based on family history is dependent not only on the number of individuals in the family who have a melanoma but also on the number of melanomas in each family member.[19] For example, the familial risk of melanoma was found to increase 2.2-fold (95% CI, 2.2–2.3) with a single FDR who has one melanoma and up to 16.3-fold (95% CI, 9.5–26.1) with a single FDR who has five or more melanomas.[19] When two or more family members were diagnosed with melanoma before age 30 years, the lifetime cumulative risk for the family members rose to 14%.[20]
A study on the heritability of cancer among 80,309 monozygotic and 123,382 dizygotic twins showed that melanoma has a heritability of 58% (95% CI, 43%–73%), suggesting that more than half of the risk of melanoma is caused by inherited factors.[21] A study looking at the contribution of family history to melanoma risk showed a population-attributable fraction ranging from less than 1% in northern Europe to 6.4% in Australia,[22] suggesting that only a small percentage of melanoma cases are caused by familial factors. Rarely, however, in some families many generations and multiple individuals develop melanoma and are at much higher risk. For individuals from these families, the incidence of melanoma is higher for sun-protected rather than sun-exposed skin.[23]
The major hereditary melanoma susceptibility gene, CDKN2A, is found to be altered in approximately 35% to 40% of families with three or more melanoma cases. To date, more than half of the families with multiple cases of melanoma have no identified pathogenic variant.[24,25] The definition of a familial cluster of melanoma varies by geographical region, worldwide, because of the role played by UV radiation in melanoma pathogenesis. In heavily insolated regions (regions with high ambient sun exposure), three or more affected family members are required; in regions with lower levels of ambient sunlight, two or more affected family members are considered sufficient to define a familial cluster. The American College of Medical Genetics and Genomics and the National Society of Genetic Counselors recommend that an individual with any of the following characteristics be referred for a cancer genetics consultation:[26]
  • A personal history of three or more primary melanomas.
  • A personal history of melanoma and pancreatic cancer.
  • A personal history of melanoma and astrocytoma.
  • Three or more cases of melanoma and/or pancreatic cancer in FDRs.
  • Melanoma and astrocytoma in two FDRs.

Personal history of melanoma

A previous melanoma places one at high risk of developing additional primary melanomas, particularly for people with the most common risk factors for melanoma, such as cutaneous phenotype, family history, a pathogenic variant in CDKN2A, a great deal of early-life sun exposure, and numerous or atypical nevi. In the sporadic setting, approximately 5% of melanoma patients develop more than one primary cancer, while in the familial setting the corresponding estimate is 30%. This greater-than-expected rate of multiple primary cancers of the same organ is a common feature of hereditary cancer susceptibility syndromes; it represents a clinical finding that should raise the level of suspicion that a given patient’s melanoma may be related to an underlying genetic predisposition. Risk of a second primary melanoma after diagnosis of a first primary melanoma is approximately 5% and is greater for males and older patients.[27-30] A study in Sweden of more than 65,000 individuals with melanoma found a SIR of 2.8 (95% CI, 2.3–3.4) for a second melanoma in individuals with a family history of melanoma and a SIR of 2.5 (95% CI, 2.3–2.7) in individuals with no family history.[31] The risk of a second melanoma increased when the first melanoma was diagnosed before age 40 years (SIR, 4.7; 95% CI, 3.9–5.6%). The SIRs increased with increasing numbers of melanomas.

Personal history of nonmelanoma skin cancer

Having a personal history of basal cell carcinoma (BCC) or squamous cell carcinoma (SCC) is also associated with an increase in risk of a subsequent melanoma.[32-34] Depending on the study, this risk ranges from a nonsignificant increase for melanoma with a previous SCC of 1.04 (95% CI, 0.13–8.18) to a highly significant risk of 7.94 (95% CI, 4.11–15.35).[35,36] It is likely that this relationship is the result of shared risk factors (of which sun exposure is presumably one), rather than a specific genetic factor that increases risk of both. Pigmentary characteristics are critically important for the development of melanoma, and cutaneous phenotype (described above), in combination with excessive sun exposure, is associated with an increased risk of all three types of skin cancers.

Major Genes for Melanoma

CDKN2A/p16 and p14/ARF

The major gene associated with melanoma is CDKN2A/p16, cyclin-dependent kinase inhibitor 2A, which is located on chromosome 9p21. This gene has multiple names (MTS1INK4, and MLM) and is commonly called by the name of its protein, p16. It is an upstream regulator of the retinoblastoma gene pathway, acting through the cyclin D1/cyclin-dependent kinase 4 complex. This tumor suppressor gene has been intensively studied in multiple-case families and in population-based series of melanoma cases. CDKN2A controls the passage of cells through the cell cycle and provides a mechanism for holding damaged cells at the G1/S checkpoint to permit repair of DNA damage before cellular replication. Loss of function of tumor suppressor genes—a good example of which is CDKN2A—is a critical step in carcinogenesis for many tumor systems.
CDKN2A encodes two proteins, p16INK4a and p14ARF, both inhibitors of cellular senescence. The protein produced when the alternate reading frame (ARF) for exon 1 is transcribed instead of the standard reading frame exerts its biological effects through the p53 pathway. It mediates cell cycle arrest at the G1 and G2/M checkpoints, complementing p16’s block of G1/S progression—thereby facilitating cellular repair of DNA damage.
Pathogenic variants in CDKN2A account for 35% to 40% of familial melanomas [24] and fewer than 1% of unselected melanoma cases.[37] A study of more than 1,000 individuals in Spain showed that 6.6% of individuals with melanoma have a family history of two or more FDRs with melanoma, and up to 15% have a family history suggestive of familial melanoma that includes melanoma or pancreatic cancer diagnoses in FDRs or second-degree relatives (SDRs).[38] A large case series from Britain found that CDKN2A pathogenic variants were present in 100% of families with seven to ten individuals affected with melanoma, 60% to 71% of families with four to six cases, and 14% of families with two cases.[25] A similar study of Greek individuals with melanoma found CDKN2A pathogenic variants in 3.3% of single melanoma cases, 22% of familial melanoma cases, and 57% of individuals with multiple primary melanomas (MPM).[39] A study of 92 sequential cases of Italian individuals with familial atypical multiple mole-melanoma syndrome (defined as three or more individuals with primary cutaneous melanoma or one individual with MPM) found CDKN2A pathogenic variants in 20% of individuals, including three unrelated individuals with a p.D84V variant.[40Cascade testing identified 14 of 40 unaffected family members undergoing testing who carried their family’s CDKN2A pathogenic variant. However, a second study of 106 familial melanoma cases (defined as at least two melanoma cases) only found CDKN2A pathogenic variants in 8.3% of cases.[41] The frequency of CDKN2A pathogenic variants is as high as 22% in families with two cases of melanoma who have other features of hereditary melanoma, such as an age at diagnosis younger than 50 years or one or more individuals diagnosed with MPM.[42] A study of 587 individuals with a single primary melanoma or MPM found CDKN2A pathogenic variants in 19% of individuals with MPM relative to 4.4% of individuals with a single primary melanoma.[43CDKN2A pathogenic variants were found in 29.6% of individuals with three or more primary melanomas. Individuals with more than three primary melanomas and a family history of melanoma (undefined) had a frequency of CDKN2A pathogenic variants of 58.8%. Many pathogenic variants reported among families consist of founder variants, which are unique to specific populations and the geographic areas from which they originate.[44-51]
Depending on the study design and target population, melanoma penetrance related to CDKN2A pathogenic variants differs widely. One study of 80 multiple-case families demonstrated that the penetrance varied by country, an observation that was attributed to major differences in sun exposure.[52] For example, in Australia, the penetrance was 30% by age 50 years and 91% by age 80 years; in the United States, the penetrance was 50% by age 50 years and 76% by age 80 years; in Europe, the penetrance was 13% by age 50 years and 58% by age 80 years. In contrast, a comparison of families with the CDKN2A pathogenic variant in the United Kingdom and Australia demonstrated the same cumulative risk of melanoma; for CDKN2A carriers, the risk of developing melanoma seemed independent of ambient UV radiation.[53] Another study of individuals with melanoma identified in eight population-based cancer registries and one hospital-based sample obtained a self-reported family history and sequenced CDKN2A in all individuals. The penetrance was estimated as 14% by age 50 years and 28% by age 80 years.[30] The explanation for these differences lies in the method of identifying the individuals tested, with penetrance estimates increasing with the number of affected family members. The method of family ascertainment in the latter study made it much less likely that “heavily loaded” melanoma families would be identified. Coinheritance of MC1R variants also increases CDKN2A penetrance; this genetic variant, described in further detail below, is therefore both a low-penetrance susceptibility gene and a modifier gene.[54] (Refer to the MC1R section of this summary for more information.) Other modifier loci have also been assessed in CDKN2A carriers; interleukin-9 (IL9) and GSTT1 were the only loci with effects that reached statistical significance, suggesting that other minor risk factors may interact with major risk loci.[55,56]
One study reported a melanoma incidence rate of 9.9 per 1,000 person years among 354 FDRs and 2.1 per 1,000 person years among 391 SDRs of probands with a p16-Leiden (c.225-243del19) CDKN2A pathogenic variant (95% CIs of 7.4–13.3 and 1.2–3.8, respectively). These data indicate a melanoma rate that is much higher than that of the general population (12.9-fold increased incidence) for SDRs in untested relatives of carriers of CDKN2A pathogenic variants.[57]
A comparison of clinical features from 182 patients with CDKN2A pathogenic variants and 7,513 individuals without variants found that individuals with CDKN2A pathogenic variants were statistically significantly younger at diagnosis (mean age at diagnosis, 39.0 y vs. 54.3 y; P < .001). There was also a 5-year cumulative incidence of a second melanoma of 23.4% in carriers of pathogenic variants and a rate of 2.3% in controls who were negative for a pathogenic variant.[58] A study of pediatric patients with melanoma (aged 9–19 y) in melanoma-prone families reported a significant increase in melanoma prevalence (6-fold to 28-fold) relative to the general population. In this series, 7 of 21 patients (33%) with CDKN2A pathogenic variants were diagnosed with MPMs before age 20 years.[59] An Italian study performed genotype-phenotype correlations in 100 families with familial melanoma to determine clinical features predictive of the identification of a CDKN2A pathogenic variant. Probands with MPM, at least one melanoma with Breslow thickness greater than 0.4 mm, and more than three affected family members had a greater than 90% likelihood of having a pathogenic variant; probands with none of these features had less than a 1% likelihood of having a CDKN2A pathogenic variant. The most predictive feature was MPM.[60]
Melanomas in carriers of CDKN2A pathogenic variants largely resemble those found sporadically. A large study that compared melanoma pathology between CDKN2A carriers and individuals with sporadic melanoma found few significant differences, with a minor trend of increased pigmentation among pathogenic variant carriers.[61] Another study of more than 670 carriers of CDKN2A pathogenic variants and 1,258 carriers of wild-type or benign CDKN2A variants found that participants with pathogenic variants were more likely to be diagnosed at an earlier age (median age, 38 vs. 46 y) and have MPM (average number of melanomas, 2.3 vs. 1.4).[62] Two pathogenic variants in CDKN2A (p.Arg112dup, p.Pro48Leu) may be prognostic factors in patients with melanoma. After adjusting for age, sex, and tumor classification, carriers of these CDKN2A pathogenic variants had poorer melanoma-specific survival than did non-CDKN2A carriers (hazard ratio [HR], 2.5; 95% CI, 1.49–2.21).[63] An early study suggested that somatic NRAS mutations occurred at a higher rate in melanomas diagnosed in Swedish families who carry CDKN2A pathogenic variants as compared with those with sporadic melanomas.[64] However, subsequent studies have found that the rates of the common somatic mutations (BRAFNRAS) in melanomas from CDKN2A carriers resemble or are lower than those described in the sporadic melanoma population.[65,66] Of note, melanomas from several patients with CDKN2A variants had coexisting BRAF and NRAS mutations, which is an uncommon occurrence in sporadic melanomas.[66]
CDKN2A exon 1ß pathogenic variants (p14ARF) have been identified in a small percentage of families negative for p16INK4a pathogenic variants. In a study of 94 Italian families with two or more cases of melanoma, 3.2% of families had variants in p14ARF.[67] A patient with a balanced translocation between chromosomes 9 and 22 that disrupted p14ARF had melanoma, DNA repair deficiency, and features of DiGeorge syndrome, including deafness and malformed inner ears.[68]
CDKN2A, cutaneous phenotypes, and cancers other than melanoma
In a Melanoma Genetics Consortium (GenoMEL) study of 1,641 family members of melanoma probands, family members with a CDKN2A pathogenic variant were more likely to have atypical nevi than were family members of CDKN2A noncarriers (odds ratio [OR], 1.65; 95% CI, 1.18–2.28).[69] Another study of individuals in Sweden with MPM and two or more cases of melanoma in their first-, second-, or third-degree relatives found CDKN2A pathogenic variants in 43 of 100 cases. Familial MPM cases with CDKN2A variants, familial MPM cases wild-type for CDKN2A, and nonfamilial MPM cases all showed increased risks of future cutaneous SCCs compared with controls (relative risk [RR], 4.8; 95% CI, 1.5–15.1).[70]
Results from the Genes, Environment, and Melanoma study showed that FDRs of carriers of CDKN2A pathogenic variants with melanoma had an approximately 50% increased risk of cancers other than melanoma, compared with FDRs of other melanoma patients.[71] Cancers with increased risk in this population included gastrointestinal cancers (RR, 2.4; 95% CI, 1.4–3.7), pancreatic cancers (RR, 7.4; 95% CI, 2.3–18.7), and Wilms tumor (RR, 40.4; 95% CI, 3.4–352.7). A Spanish study of the FDRs of 66 melanoma patients with known CDKN2A pathogenic variants also showed an increase in prevalence of other cancers, including pancreatic (prevalence ratio [PR], 2.97; 95% CI, 1.72–5.15), lung (PR, 3.04; 95% CI, 1.93–4.80), and breast cancers (PR, 2.19; 95% CI, 1.36–3.55).[72] A large registry study from Sweden that included 27 families carrying the Arg112dup pathogenic variant in CDKN2A observed excess nonmelanoma cancers in both carriers (n = 120) and FDRs (n = 275). For carriers of CDKN2A pathogenic variants, increased risks relative to a control population were seen for pancreatic (RR, 43.8; 95% CI, 13.8–139), upper digestive (RR, 17.1; 95% CI, 6.3–46.5), respiratory (RR, 15.6; 95% CI, 5.4–46.0), and breast cancers (RR 3.0; 95% CI, 0.9–9.9), among others (all cancers: RR, 5.0; 95% CI, 3.7–7.3). The RRs in FDRs were 20.6 (95% CI, 11.6–36.7) for pancreatic cancers, 6.0 (95% CI, 2.8–13.1) for respiratory cancers, 3.3 (95% CI, 1.5–7.6) for upper digestive cancers, and 1.9 (95% CI, 0.9–4.0) for breast cancers, with a RR of all cancers of 2.1 (95% CI, 1.6–2.7). A lesser-increased cancer risk was seen among SDRs. They also observed a significant association between smoking and risk of pancreatic, respiratory, and upper digestive cancers, with an OR of 9.3 (95% CI, 1.9–44.7) for ever-smoking carriers compared with never-smoking carriers.[73]
A few studies have identified individuals with sarcoma who have germline pathogenic variants in CDKN2A, but the number of cases is too small to determine the risk of sarcoma associated with this gene.[74,75] One patient with features of Li-Fraumeni syndrome did not carry a TP53 pathogenic variant, but a deletion of CDKN2A and CDKN2B.[75] A whole-exome sequencing study of a Li-Fraumeni–like family with three individuals with soft tissue sarcoma identified a shared pathogenic CDKN2A variant.[74] An evaluation of 474 melanoma families with cases of sarcoma and 190 TP53 variant–negative Li-Fraumeni–like families found eight additional individuals with sarcoma and pathogenic CDKN2A variants.
Pancreatic cancer
A subset of families carrying a CDKN2A pathogenic variant also displays an increased risk of pancreatic cancer.[76,77] The overall lifetime risk of pancreatic cancer in these families ranges from 11% to 17%.[78] The RR has been reported as high as 47.8.[79] Although at least 18 different variants in p16 have been identified in such families, specific pathogenic variants appear to have a particularly elevated risk of pancreatic cancer.[24,80] Pathogenic variants affecting splice sites or Ankyrin repeats were found more commonly in families with both pancreatic cancer and melanoma than in those with melanoma alone. The p16 Leiden variant is a 19-base pair deletion in CDKN2A exon 2 and is a founder pathogenic variant originating in the Netherlands. In one major Dutch study, 19 families with 86 members who had melanoma also had 19 members with pancreatic cancer in their families, a cumulative risk of 17% by age 75 years. In this study, the median age of pancreatic cancer onset was 58 years, similar to the median age at onset for sporadic pancreatic cancer.[81] However, other reports indicate that the average age at diagnosis is 5.8 years earlier for these carriers of pathogenic variants than for those with sporadic pancreatic cancer.[82] Geographic variation may play a role in determining pancreatic risk in these families carrying known pathogenic variants. In a multicontinent study of the features of germline CDKN2A pathogenic variants, Australian families carrying these variants did not have an increased risk of pancreatic cancer.[83] It was also reported that similar CDKN2A variants were involved in families with and without pancreatic cancer;[84] therefore, there are additional factors involved in the development of melanoma and pancreatic cancer. Some families with CDKN2A pathogenic variants may have a pattern of site-specific pancreatic cancer only.[85-87] Conversely, melanoma-prone families that do not have a CDKN2A pathogenic variant have not been shown to have an increased risk of pancreatic cancer.[81]
In a review of 110 families with multiple cases of pancreatic cancer, 18 showed an association between pancreatic cancer and melanoma.[88] Only 5 of the 18 families with cases of both pancreatic cancer and melanoma had individuals with multiple dysplastic nevi. These 18 families were assessed for pathogenic variants in CDKN2A; variants were identified in only 2 of the 18 families, neither of which had a dysplastic nevi phenotype.
Melanoma-astrocytoma syndrome
The melanoma-astrocytoma syndrome is another phenotype caused by pathogenic variants in CDKN2A. The possible existence of this disorder was first described in 1993.[89] A study of 904 individuals with melanoma and their families found 15 families with 17 members who had both melanoma and multiple types of tumors of the nervous system.[90] Another study found a family with multiple melanoma and neural cell tumors that appeared to be caused by loss of p14ARF function or to disruption of expression of p16.[91] Plexiform neurofibromas have also been reported in individuals with deleterious CDKN2A variants.[92-95]

CDK4 and CDK6

Cyclin-dependent kinases have important roles in progression of cells from G1 to S phase. CDK4 and CDK6 partner with the cyclin–D associated kinases to accelerate the function of the cell cycle. Phosphorylation of the retinoblastoma (Rb) protein in G1 by cyclin-dependent kinases releases transcription factors, inducing gene expression and metabolic changes that precede DNA replication, thus allowing the cell to progress through the cell cycle. These genes are of conceptual significance because they are in the same signaling pathway as CDKN2A.
Germline CDK4 pathogenic variants are very rare, being found in only a handful of melanoma kindreds.[96-98] All described families demonstrated a substitution of amino acid 24, suggesting this position as a variant hotspot within the CDK4 gene. Three Latvian families with melanoma have a R24H substitution arising on the same haplotype, which suggests that it could be a founder pathogenic variant in this population.[99] A CDK4 pathogenic variant affects binding of p16 with its subsequent inhibition of CDK4 functionality. With constitutive activation of germline CDK4CDK4 acts as a dominant oncogene. A small study showed that the melanoma cancer risk in 17 families with CDK4 pathogenic variants was similar to the risk seen in families with CDKN2A variants.[100] (Refer to the CDKN2A/p16 and p14/ARF section of this summary for more information.) In addition, the melanomas found in CDK4 families appear to have similar rates of somatic BRAF mutations to those found in sporadic populations, although because of the rare nature of CDK4 germline variants, the data are necessarily limited.[101]
Despite its functional similarity to CDK4germline variants in CDK6 have not been identified in any melanoma kindreds.[102]

Telomere maintenance genes

Telomerase reverse transcriptase (TERT)
Linkage of melanoma to a region of chromosome 5p was observed in a single, large kindred with multiple melanomas and other cancers.[103] Sequencing demonstrated a pathogenic variant in the promoter region of a subunit of TERT, which demonstrated increased promoter activity in construct assays. This variant cosegregated with melanoma and other cancers (ovarian, renal, bladder, and lung), with multiple cancers observed in single individuals. At least one affected family member was observed to have numerous nevi. Somatic mutations in the same region were observed in 125 of 168 sporadic melanomas in the same report.[103] A separate study reported pathogenic variants that also increased promoter activity in the same TERT promoter region in 50 of 70 sporadic melanomas.[104] Similar pathogenic variants were seen in 16% of a diverse set of established cancer cell lines, suggesting this might be a common activation variant in multiple cancer types. The frequency of this variant in melanoma families has not been fully investigated, but one study of 273 families with three or more cases of melanoma identified only one family (with 7 melanoma cases) that carried a c.-57 T>G promoter variant.[105] A study of 106 familial melanoma cases (defined as at least two melanoma cases or MPM in the proband) found that 47% of MPM cases and 58% of familial melanoma cases carried a risk-associated TERT promoter variant, rs2853669.[41] The prevalence of this variant in the general population is estimated to be between 25% and 29%.[106]
POT1
Exome and genome-sequencing in individuals from hereditary melanoma families led to the identification of missense pathogenic variants in POT1 that segregate with disease in numerous studies.[107,108] A POT1 Ser270Asn missense pathogenic variant was found in 5 of 56 unrelated melanoma families from Italy.[107] This variant was not observed in over 2,000 Italian controls. Ser270Asn is thought to be a founder pathogenic variant, as all families with the variant shared a haplotype. Additional POT1 missense pathogenic variants, including Tyr89Cys, Arg137His, and Gln623His, were identified in other melanoma families and were not seen in unaffected controls.[107,108] Together, POT1 pathogenic variants were found in approximately 4% of melanoma families who lacked CDKN2A or CDK4 variants, suggesting it may be another gene in hereditary melanoma. POT1 binds to single-stranded telomeric repeat regions and is thought to aid in maintenance of telomere length. Most of the variants segregating in families occur in the two oligonucleotide/oligosaccharide-binding domains of the protein, which are the portion of the protein critical for binding DNA. Individuals carrying POT1 pathogenic variants showed longer telomere lengths than melanoma cases without the POT1 variants, suggesting a link between disruption in normal telomere length and melanoma.[107,108] The clinical utility of testing this gene has not yet been established.
ACD and TERF2IP
In one study, 510 melanoma families were screened by next-generation sequencing for pathogenic variants in genes in the shelterin complex, which protects chromosomal ends. Six families were found to have variants in ACD, and four families had variants in TERF2IP.[109] The ACD variants clustered in the POT1 binding domain. Because some of these variants did not lead to a truncated protein, the functional significance is not confirmed.

DNA repair genes

Xeroderma pigmentosum (XP) patients with defective DNA repair have a more than 1,000-fold increase in melanoma risk. These patients are diagnosed with melanoma at a significantly younger age than individuals in the general population; on average, melanoma diagnosis occurs at age 22 years in XP patients.[110] The anatomic site distribution of melanomas in XP patients is similar to that of the general population.[111,112]
Genetic polymorphisms associated with DNA repair genes have been associated with mildly increased melanoma risk in the general population.[113] A meta-analysis of eight case-control studies comprising more than 5,000 cases and 7,000 controls found that individuals carrying the Asp1104His polymorphism in XPG had an increased risk of melanoma (OR, 2.42; 95% CI, 2.26–2.60).[114]
(Refer to the Xeroderma pigmentosum section in the Squamous Cell Carcinoma section of this summary for more information.)

BRCA1-associated protein 1 (BAP1)

BAP1 has recently emerged as a gene implicated both in sporadic and hereditary melanomas.[115] Originally described in a cohort of uveal melanoma patients, BAP1 is a tumor suppressor gene that was found to be inactivated in 84% of uveal melanoma patients with metastases.[116] Although the majority of these variants were somatic, one patient was found to have a germline frameshift variant. A phenotype associated with BAP1 pathogenic variants was subsequently described.[117] Two families with multiple, elevated melanocytic tumors that were clinically and histopathologically distinct from other melanocytic neoplasms were found to have inactivating germline pathogenic variants of BAP1. These tumors, which have been termed melanocytic BAP1-mutated atypical intradermal tumors, or MBAITs, are found throughout the body, generally measure approximately 5 mm, and begin to appear in the second decade of life. MBAITs are 2 mm to 10 mm in diameter, and affected individuals (about 67% of BAP1 pathogenic variant carriers) can have 5 to more than 50 skin lesions.[117,118] Cases of cutaneous melanoma were present in these families, but the rate of malignant progression is thought to be low due to the relative lack of melanomas in comparison with the number of more papular tumors. This syndrome has been called BAP1 tumor syndrome or the COMMON (cutaneous and ocular melanoma and atypical melanocytic proliferation with other internal neoplasms) syndrome, and it is inherited in an autosomal dominant pattern.[119] Further investigation has supported the association between familial cutaneous melanoma and uveal melanoma in BAP1 carriers.[120-124] However, potentially pathogenic BAP1 germline variants occur in a low percentage of melanoma cases. One targeted sequencing study of 1,109 unselected cutaneous melanoma cases found only seven germline missense pathogenic variants (<1%).[37] A second series of 1,977 melanoma cases and 754 controls identified 22 rare variants in BAP1 among cases and 5 rare variants among controls; three of the variants found only among cases were confirmed to disrupt BAP1 function and were associated with family histories of other BAP1-associated cancers.[125] In support of a link between melanoma risk and BAP1, in one series, about 18% of individuals with a BAP1 pathogenic variant developed melanoma.[121] In addition, although data are currently limited, patients with germline pathogenic variants in BAP1 may be at increased risk of lung adenocarcinoma, mesothelioma, BCC, and clear cell carcinoma of the kidney.[118,120,122,123,126,127]
Other studies have reported pathogenic variants in BAP1. A missense pathogenic variant (p.Leu570Val) in a family with multiple cases of melanoma was described to affect splicing and result in a frameshift. This family also had cases of uveal melanoma and paraganglioma.[126] Another family with a Y646X BAP1 pathogenic variant had multiple cancers, including multiple cutaneous melanomas and BCCs, uveal melanoma, and mesotheliomas.[128] The authors hypothesized that a gene-environment interaction between BAP1 pathogenic variants and UV radiation and asbestos exposure contributed to the high incidence of multiple cancers in this family.

PTEN hamartoma tumor syndromes (including Cowden syndrome)

Cowden syndrome and Bannayan-Riley-Ruvalcaba syndrome (BRRS) are part of a spectrum of conditions known collectively as PTEN hamartoma tumor syndromes. Approximately 85% of patients diagnosed with Cowden syndrome, and approximately 60% of patients with BRRS have an identifiable PTEN pathogenic variant.[129] In addition, PTEN pathogenic variants have been identified in patients with very diverse clinical phenotypes.[130] The term PTEN hamartoma tumor syndromes refers to any patient with a PTEN pathogenic variant, irrespective of clinical presentation.
PTEN functions as a dual-specificity phosphatase that removes phosphate groups from tyrosine, serine, and threonine. Pathogenic variants of PTEN are diverse, including nonsense, missense, frameshift, and splice-site variants. Approximately 40% of variants are found in exon 5, which encodes the phosphatase core motif, and several recurrent pathogenic variants have been observed.[131] Individuals with variants in the 5’ end or within the phosphatase core of PTEN tend to have more organ systems involved.[132]
Operational criteria for the diagnosis of Cowden syndrome have been published and subsequently updated.[133,134] These included major, minor, and pathognomonic criteria consisting of certain mucocutaneous manifestations and adult-onset dysplastic gangliocytoma of the cerebellum (Lhermitte-Duclos disease). An updated set of criteria based on a systematic literature review has been suggested [135] and is currently utilized in the National Comprehensive Cancer Network (NCCN) guidelines.[136] Contrary to previous criteria, the authors concluded that there was insufficient evidence for any features to be classified as pathognomonic. With increased utilization of genetic testing, especially the use of multigene panels, clinical criteria for Cowden syndrome will need to be reconciled with the phenotype of individuals with documented germline PTEN pathogenic variants who do not meet these criteria. Until then, whether Cowden syndrome and the other PTEN hamartoma tumor syndromes will be defined clinically or based on the results of genetic testing remains ambiguous. The American College of Medical Genetics and Genomics (ACMG) suggests that referral for genetics consultation be considered for individuals with a personal history of or a first-degree relative with 1) adult-onset Lhermitte-Duclos disease or 2) any three of the major or minor criteria that have been established for the diagnosis of Cowden syndrome.[26] Detailed recommendations, including diagnostic criteriaExit Disclaimer for Cowden syndrome, can be found in the NCCN and ACMG guidelines.[26,136] Additionally, a predictive modelExit Disclaimer that uses clinical criteria to estimate the probability of a PTEN pathogenic variant is available; a cost-effectiveness analysis suggests that germline PTEN testing is cost effective if the probability of a variant is greater than 10%.[137]
Over a 10-year period, the International Cowden Consortium (ICC) prospectively recruited a consecutive series of adult and pediatric patients meeting relaxed ICC criteria for PTEN testing in the United States, Europe, and Asia.[138] Most individuals did not meet the clinical criteria for a diagnosis of Cowden syndrome or BRRS. Of the 3,399 individuals recruited and tested, 295 probands (8.8%) and an additional 73 family members were found to harbor germline PTEN pathogenic variants. In addition to breast, thyroid, and endometrial cancers, the authors concluded that on the basis of cancer risk, melanoma, kidney cancer, and colorectal cancers should be considered part of the cancer spectra arising from germline PTEN pathogenic variants. A second study of approximately 100 patients with a germline PTEN pathogenic variant confirmed these findings and suggested a cumulative cancer risk of 85% by age 70 years.[139]
The risk of melanoma in PTEN carriers is controversial. In the study of 100 patients referenced above, four women and four men were diagnosed with melanoma and less than one case was expected, for a SIR of 28.3 for women (95% CI, 7.6–35.4) and 39.4 for men (95% CI, 10.6–100.9) (P < .001).[139] In the ICC study described above, an elevated SIR of 8.5 (95% CI, 4.1–15.6) was reported in 368 carriers of PTEN pathogenic variants.[138] In this cohort, the estimated lifetime risk of melanoma in carriers of PTEN pathogenic variants was 6% (range, 1.6%–9.4%). However, it is important to recognize that a subsequent prospective study did not observe an elevated melanoma risk.[140] In this study, only 1 of 180 carriers was diagnosed with melanoma. (Refer to the PDQ summaries on the Genetics of Colorectal Cancer and the Genetics of Breast and Gynecologic Cancers for more information about risks of other cancers in PTEN hamartoma tumor syndromes.)



No hay comentarios:

Publicar un comentario