Genetic Testing for Melanoma


Key Points

  • Approximately 10% of melanomas occur in family clusters.

  • In North American populations, mutations in the CDKN2A gene are associated with up to a 76% lifetime risk for melanoma. CDKN2A may also increase the risk for pancreatic cancer to as much as 25%.

  • Clinical genetic testing is available to identify families with mutations in the CDKN2A gene.

  • In moderate to high melanoma incidence areas, referral for genetic counseling and testing should be considered for: (1) individuals with three or more primary invasive melanomas; (2) families with at least one invasive melanoma and two or more other diagnoses of invasive melanoma and/or pancreatic cancer.

  • Carriers of a deleterious CDKN2A mutation should receive aggressive melanoma surveillance and be educated about the risk for pancreatic cancer, including the possibility of participating in a pancreatic cancer surveillance program.

Introduction

Approximately 10% of melanomas present in familial clusters ( Fig. 30.1 ). Aggregation of melanoma in a family may occur due to a combination of shared environmental exposure and genetic factors. Melanoma risk factors are summarized in Table 30.1 . A recent study has found that approximately 40% of risk in families is due to shared variants in genes associated with pigmentation. While shared phenotypic features and environmental exposures accounts for a significant amount of familial melanoma risk, rare families harbor mutations in highly penetrant cancer predisposition genes. Patients who have inherited predisposition for melanoma are at 30–70-fold higher risk of developing melanoma than the general population.

Figure 30.1, Prevalence of hereditary melanoma and of p16 mutation in hereditary melanoma.

Table 30.1
Risk Factors for Cutaneous Melanoma
Risk Factor Estimated Relative Risk *
Skin type I (fair skin) 1.4
Freckling 2–3
Blue eyes 1.6
Red hair 2.4–4
History of blistering sunburns 2–3
≥6 atypical nevi 6.3
≥10 dysplastic nevi 12
100 or more nevi * 3.1–16.5
Family history of melanoma 2–3
Previous primary cutaneous melanoma 8.5
Member of melanoma-prone family Up to 35–70

* Relative risk in the United Kingdom population.

One or more affected first-degree relatives.

Multiple affected relatives on the same side of the family.

To date, two high penetrance genes definitively associated with hereditary melanoma, cyclin-dependent kinase inhibitor 2A ( CDKN2A) on chromosome 19p21 and cyclin-dependent kinase 4 ( CDK4 ) on chromosome 12q14, have been identified. Mutations in CDKN2A account for approximately 20–40% of hereditary melanoma, and 0.2–1% of all melanomas ( Fig. 30.1 ). Mutations in CDK4 are rare and have been reported in fewer than fifteen families worldwide, leaving CDKN2A as the most significant melanoma predisposition gene identified thus far.

An efficient method for identifying this subset of particularly high-risk patients is needed so that additional clinical resources can be devoted to their care, even in fast-paced outpatient practices. Genetic testing is one mechanism by which high-risk patients and their families can be quickly identified. While CDKN2A was identified in 1994, incorporation of hereditary melanoma genetic testing into clinical practice has lagged behind that of other cancer predisposition genes such as BRCA1/BRCA2 (hereditary breast/ovarian cancer) and MSH2/MLH1 (Lynch syndrome – hereditary colon cancer). Recent studies suggest a strong interest in genetic testing among families with a history of melanoma, and that receiving genetic test results with appropriate education and counseling may help promote positive screening and behavioral changes among high-risk individuals. In order to address this growing demand for genetic services for hereditary melanoma, an international consortium recently published guidelines for appropriately identifying patients who are candidates for genetic counseling and testing for hereditary melanoma. In this chapter we will summarize current information about CDKN2A , the process of identifying high-risk families and incorporating genetic testing into clinical practice.

Gene identification

CDKN2A is a tumor suppressor gene that regulates cellular proliferation and growth by inhibiting RB (retinoblastoma protein) phosphorylation and entry into the cell cycle. CDKN2A produces two protein products, p16 INK4a , which is transcribed from exons 1α, 2, and 3, and p14 ARF , which is transcribed from an alternate reading frame and includes exons 1β, 2, and 3. Depending on the location, mutations in CDKN2A may disrupt one or both of the proteins produced by this gene. Most mutations that have been found to cause a risk for melanoma affect p16 INK4a , and at least one study has suggested that mutations which affect both proteins may be more penetrant. However, rare mutations which only affect p14 ARF have also been reported to segregate with the melanoma diagnoses in families, establishing it as an independent bona fide melanoma predisposition gene. Inheriting a deleterious mutation in one copy of CDKN2A is sufficient to cause an individual to have a melanoma predisposition. Parents who have a mutation in the CDKN2A gene will have a 50% chance of passing on the mutated allele and a 50% chance of passing on the functional copy of the gene with each offspring.

CDKN2A mutation penetrance estimates vary between populations and geographic regions due to gene–gene and gene–environment interactions. Studies of families ascertained on the basis of multiple cases have demonstrated penetrance estimates of 91%, 76%, and 58% in Australia, the United States, and Europe, respectively. Penetrance estimates are greatest in areas with high background rates of melanoma. This is likely because there are additional factors such as high ultraviolet radiation (UV) intensity and other shared genetic factors which work synergistically with the CDKN2A gene to increase melanoma risk. One of the best-studied genetic interactions is between CDKN2A and MC1R . Box et al. found the penetrance of CDKN2A mutations increased from 50% to 84% when inherited with a MCIR variant. Other genetic factors that contribute to incomplete penetrance have not yet been well defined. Yang et al. conducted an association study looking for potential CDKN2A interactions with 152 genes associated with pigmentation, DNA repair, apoptosis, and immune response. The only significant association identified was with IL9 (a gene involved with Th1:Th2 immune pathway), but several other candidate genes showed possible interactions that may become more evident with a larger sample size. Further studies are needed to characterize the gene–environment interactions that contribute to the differing melanoma risk estimates. Population-based studies have resulted in lower, but still significant, risk estimates. Begg et al. estimated a 28% risk for melanoma to age 80 in a population-based sample. However, for patients presenting in a clinical setting, penetrance estimates based on multiple case families are likely to be the most appropriate estimates to use for counseling when they are referred based on family history.

In addition to predisposing to melanoma, CDKN2A mutations are also associated with an increased risk for pancreatic cancer in some families. GenoMEL, an international melanoma genetics consortium, found that 28% of CDKN2A families had diagnoses of pancreatic cancer, compared to 6% of families without mutations. However, when data were evaluated by country, no excess risk of pancreatic cancer was seen in Australian families. Therefore, the nature of the association between CDKN2A and pancreatic cancer is still incompletely understood. Studies of families with CDKN2A mutations exhibiting pancreatic cancer have found that the risk to age 80 may be as high as 25%.

Genetic testing in clinical practice

There are several required steps for incorporating genetic testing into clinical practice, including identification of high-risk patients, providing genetic counseling, appropriately ordering genetic testing, interpreting results and incorporating findings into management recommendations. All of these steps can be time-intensive and referral to a local genetic counseling resource may be the most efficient approach for providing patients with these services. Table 30.2 provides a list of online resources for identifying cancer genetic services.

Table 30.2
Directories of Genetic Counseling, Genetic Testing, and Research Resources
Resource Website Services
National Society of Genetic Counselors www.nsgc.org International directory of genetic counselors that is searchable by location and specialty
National Cancer Institute Cancer Genetics Service Directory www.cancer.gov/search/geneticsservices/ Directory of clinical cancer genetic services that is searchable by location and specialty
GeneTests www.genetests.org International directory of clinical and research laboratories providing genetic testing Directory of genetics clinics
GenoMEL www.genomel.org International melanoma research consortium

Patient selection criteria

Because CDKN2A mutations are a rare cause of melanoma, routine testing of all melanoma patients is not an efficient or appropriate approach for identifying high-risk families. Recently, an international consortium established guidelines for identifying candidates for referral for genetic counseling and potential testing. Just as CDKN2A penetrance varies by region, the likelihood of detecting a mutation also varies. In very high melanoma incidence areas there is a greater chance that melanoma may cluster in a family due to other factors, and therefore more diagnoses in a family are required to suggest the presence of a CDKN2A mutation. In areas where melanoma is rare, fewer cases are needed to raise the possibility of a CDKN2A mutation. Therefore, criteria for genetic counseling and testing referral need to be tailored to the patterns of melanoma development in specific areas ( Table 30.3 ). In moderate to high melanoma incidence areas, referral for genetic services is recommended for individuals with three or more invasive primary melanomas (synchronous cases) and/or families with a diagnosis of an invasive melanoma plus two or more additional cases of invasive melanoma and/or pancreatic cancer in first- or second-degree relatives (metachronous cases). In low melanoma incidence areas, individuals with two primary melanomas or families with an invasive melanoma and one or more additional diagnoses of melanoma or pancreatic cancer are sufficient to warrant referral. Current studies of CDKN2A have focused on cases with invasive melanoma. The relationship of CDKN2A mutations to melanoma in situ or lentigo maligna has been less well described and it is not currently possible to make express recommendations for in-situ disease.

Table 30.3
Criteria for Referral for Genetic Services Based on Geographic or Population Background Melanoma Incidence
Moderate to High Melanoma Incidence Areas * Low Melanoma Incidence Areas *
Individuals with three or more (synchronous or metachronous) primary melanomas Individuals with two or more (synchronous or metachronous) primary melanomas
Families with a diagnosis of melanoma and two or more first- or second-degree relatives with melanoma and/or pancreatic cancer Families with a diagnosis of melanoma and one or more first- or second-degree relatives with melanoma and/or pancreatic cancer

* Moderate to high melanoma incidence areas include Australia, North America, and Northern Europe, while most other areas of the world have low melanoma incidence. However, clinicians must use clinical judgment and take into consideration a patient's ethnic background, sun exposure, and other melanoma risk factors when evaluating a family history.

This table refers to pathologically confirmed invasive melanoma.

Relatives should be on the same side of the family.

Taking a thorough family history is a crucial step in identifying high-risk families. A family history should include information on parents, siblings, children, aunts, uncles, grandparents and cousins. All cancer diagnoses in the family should be elicited since the presence of pancreatic cancer is a strong predictor of CDKN2A mutation identification. Aitken et al. found that approximately 40% of proxy reports of melanoma diagnoses by first-degree relatives are inaccurate. The most common error is misreporting basal or squamous cell carcinomas as melanoma. Reported family history tends to be even less accurate for more distantly related family members. Confirming verbal reports with medical records, particularly pathology reports, provides the most accurate basis for risk assessment.

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here