Context.—

There have been major advances in the understanding of melanoma since the last revision of the World Health Organization (WHO) classification in 2006.

Objective.—

To discuss development of the 9 distinct types of melanoma and distinguishing them by their epidemiology, clinical and histologic morphology, and genomic characteristics. Each melanoma subtype is placed at the end of an evolutionary pathway that is rooted in its respective precursor, wherever appropriate and feasible, based on currently known data. Each precursor has a variable risk of progression culminating in its fully evolved, invasive melanoma.

Data Sources.—

This review is based on the “Melanocytic Tumours” section of the 4th edition of the WHO Classification of Skin Tumours, published in 2018.

Conclusions.—

Melanomas were divided into those etiologically related to sun exposure and those that are not, as determined by their mutational signatures, anatomic site, and epidemiology. Melanomas on the sun-exposed skin were further divided by the histopathologic degree of cumulative solar damage (CSD) of the surrounding skin, into low and high CSD, on the basis of degree of associated solar elastosis. Low-CSD melanomas include superficial spreading melanomas and high-CSD melanomas incorporate lentigo maligna and desmoplastic melanomas. The “nonsolar” category includes acral melanomas, some melanomas in congenital nevi, melanomas in blue nevi, Spitz melanomas, mucosal melanomas, and uveal melanomas. The general term melanocytoma is proposed to encompass “intermediate” tumors that have an increased (though still low) probability of disease progression to melanoma.

This monograph represents a summary and discussion of a classification of melanoma that was developed for the WHO Classification of Skin Tumours, 4th edition, published in 2018.1  As in other World Health Organization (WHO) “Blue Books,” the classification of melanocytic tumors is based on that of melanomas because the focus of the book is on skin cancer rather than on benign lesions. It is also important to recognize the existence of benign tumors that may be related to the melanomas as potential precursors or as simulants.2  Although many (approximately 30%–50%) melanomas arise in association with a preexisting benign putative precursor melanocytic nevus, the overwhelming majority of nevi are stable and are more likely to regress than progress to melanoma.3  The risk of an individual nevus progressing to melanoma has been estimated to be in the order of 1 in 33 000 or less per year.4,5  Therefore, the wholesale excision of these lesions is not recommended to potentially prevent melanoma. Indeed, individuals with large numbers of nevi are at higher risk of developing melanomas not only within nevi but also within their skin, unassociated with nevi, and hence excising nevi as a strategy to prevent melanoma may have limited effect and may give patients a false sense of security. Nevertheless, the recent identification of the presence of shared genomic abnormalities between melanomas and associated nevi has provided support for this precursor role of nevi.6  The range of variation of nevi from a morphologic perspective, both clinically and histologically, and also from consideration of their genomic attributes, makes them significant as potential simulants that need to be distinguished from melanomas by reliable diagnostic techniques. Accurate diagnosis of nevi is facilitated by recognizing their biologically distinct subtypes and classifying them into the appropriate evolutionary pathway that leads to a specific melanoma subtype. Such nosology not only allows for their specific recognition but also assists in their distinction from simulants, particularly from melanoma.7  We present here a classification based on clinical, histologic, epidemiologic, and genomic characteristics, in which 9 distinct subsets or “pathways” for the development of cutaneous, mucosal, and uveal melanomas are recognized and associated with their potential precursor and simulant lesions.

The gold standard for melanoma diagnosis continues to be histopathology, in conjunction with clinical characteristics, despite the sometimes important contributions of immunohistochemistry (IHC), and rapid recent advances in genomic analysis of tumors. The currently used clinicopathologic classification of melanoma can be attributed to contemporaneous work by Vincent McGovern8  in Australia and Wallace Clark9  in the United States. These contributions led to the recognition that the vast majority of cutaneous melanomas arise from melanocytes in the epidermis and most of them evolve through 2 major stages of progression. In the first of these, the early lesions may be recognized as a pigmented patch or plaque, which expands more or less along the radii of an imperfect circle in the horizontal axis within the skin and for this reason has been termed the radial growth phase (RGP). In the next stage of progression, a tumor is formed that may infiltrate into the dermis or elevate the epidermis to form a nodule whose net direction of growth includes the vertical axis (below and/or above the level of the skin), so therefore termed the vertical growth phase (VGP). Most VGP lesions are obvious tumors; however, in the limiting case the definition of early “tumorigenic” VGP is the presence of a cluster of cells in the dermis that is larger than the largest cluster in the epidermis, or of any mitotic activity in the dermis, consistent with a lesion whose focus of proliferation is shifting from entirely within the epidermis to within the dermis as well.10  There is evidence that “RGP only” melanomas have an excellent prognosis,11,12  while VGP lesions have potential competence for metastasis, the likelihood of which increases with attributes that include increasing thickness, ulceration, microsatellites that currently form the basis of melanoma staging,13  and others such as higher mitotic rate, lymphovascular invasion, and the absence of or minimal tumor-infiltrating lymphocytes.14 

From the presence or absence of RGP and its variants, 3 major categories of melanoma were initially recognized.15,16  One of these, termed nodular melanoma (NM) is a lesion that lacks a recognizable RGP but forms a tumor from its earliest recognition, and therefore has potential competence for metastasis from first diagnosis. Another variant that was recognized in the early studies was termed superficial spreading melanoma (SSM) by Clark et al15  and pagetoid melanoma by McGovern.16  These terms respectively recognized the major clinical property of these lesions, namely, a spreading lesion that changes over time, and a major histologic property, the presence of neoplastic cells scattered throughout the epidermis in a pattern reminiscent of Paget disease of the breast. The third major variant of melanoma was termed lentigo maligna melanoma (LMM),15  also known as melanoma arising in a Hutchinson melanotic freckle,16  and represents a form of melanoma that is associated with histologic evidence of severe solar damage and has a “lentiginous” rather than “pagetoid” pattern of growth within the epidermis. This lentiginous pattern of growth resembles that seen in actinic (or solar) lentigo, a lesion that occurs in skin with severe cumulative solar damage (CSD), and is characterized by melanocytic proliferation as single cells along the dermal-epidermal junction. These variants represent the major patterns of melanoma in skin that is susceptible to CSD and is exposed to sunlight, such as in populations of northern European ancestry, especially those living in sunny climates like Australia, New Zealand, and the United States, or traveling on sun-seeking vacations.17,18 

It is the growing and changing RGP stage of melanoma that gives rise to the characteristic signs of early melanoma, described in the well-known ABCDE mnemonic.19,20  “A” stands for “asymmetry,” where one half of the lesion, for example, differs from the other half in shape or color. “B” stands for “border irregularity,” whereby lesions begin to take on the morphology of an island with a highly indented coastline. “C” stands for “color variegation” whereby lesions evolve from mainly tan macules to papular/plaque lesions with a variety of colors including brown, black, and red-white-and-blue.” “D” stands for “diameter,” initially characterized as greater than 4 mm, although there is strictly no lower limit in the size that a melanoma can be formed. Lesions smaller than 4 mm can be diagnosed as melanoma but criteria should be stringent to avoid the phenomenon of “overdiagnosis,” whereby lesions are diagnosed as melanomas that would not have ability for causing harm to the host unless they progress. Most of the histologically convincing small melanomas will be examples of NM, which are pure tumorigenic VGP lesions that can have competence for metastasis even when small and form an important exception to the ABCDE criteria.21  “E” stands for elevation. However, not all melanomas—especially those early SSM lesions and lesions of the lentigo maligna, acral, and mucosal lentiginous types—are elevated when entirely in the in situ RGP. “E” also stands for “evolution,” whereby a history of growth and other changes is often the discerning feature of the lesion. In a person with multiple atypical pigmented lesions, an evolving melanoma often presents as an “ugly duckling” sign where the lesion in question is noticeably out of step with the patient's other pigmented lesions.22 

More recently, a theory of “divergent pathways” to melanoma formation/pathogenesis was proposed by Whiteman et al,23  who postulated that cutaneous melanomas may arise through 2 distinct pathways, one associated with melanocyte proliferation and broadly corresponding to the SSM subtype, and the other with chronic exposure to sunlight and corresponding to LMM. Independently, genetic analyses of BRAFV600E mutations by Maldonadoet al24  and Curtin et al25  indicated that they were particularly common in melanomas on sun-exposed skin with little solar elastosis but comparatively infrequent in those arising in skin with marked solar elastosis. These observations thus laid the groundwork for a classification of melanoma that encompasses not only histologic but also clinical, epidemiologic, and genetic characteristics. Subsequently, it has become realized that other genomic aspects of melanoma also correlate with different pathways,7  and together with epidemiologic, clinical and histopathologic features, allow for the distinction of 9 pathways of melanoma (including uveal melanoma, which is not further discussed here). This classification is presented in Table 1. Similar to other tumors, the patterns of genetic alterations in melanomas and their respective precursor lesions indicate that the neoplastic proliferation is initiated by gain-of-function mutations of growth-promoting genes. This can occur through point mutations, gene fusions, and gene amplification. These alterations are typically followed by loss of suppressor function through inactivating mutations, deletions, or epigenetic silencing and are followed by activation of additional growth and survival-related genes.6  Examples of common driver oncogenes, which are characteristically mutually exclusive in any given primary tumor, include mutations of BRAF or NRAS in cutaneous melanomas and others, to be discussed in additional pathways in sections following.6  Examples of driver fusion genes include fusions of the kinase domains of ALK, ROS, NTRK, MET, RET, PRKCA, MAP3K8, BRAF, and others, with a variety of 5′ partner genes, which occur also in a mutually exclusive pattern of each other and other oncogenic driver mutations.2630  In more advanced melanomas the mutually exclusive pattern of these mutations can change, as several driver mutations can exist in subclones, which can become selected out, especially under systemic therapy targeting signaling pathway components.

Table 1

Classification of Melanoma (Modified From 2018 WHO Classification)

Classification of Melanoma (Modified From 2018 WHO Classification)
Classification of Melanoma (Modified From 2018 WHO Classification)

Kinase fusions occur predominantly in clinically benign but morphologically atypical lesions termed Spitz tumors but also in some non-Spitz melanomas.31  Examples of genetic changes that promote disease progression and may be identified in melanomas include homozygous loss of CDKN2A, which encodes p16; inactivating mutations of CDKN2A (or other functional defects in the p16 protein or its expression, or in its pathway), of TP53, NF1, and other suppressor genes; and activation of various other pathways such as telomerase (often through TERT promoter mutations). These genetic events are characteristically associated with corresponding lesional changes that herald progression from precursor lesions (wholly benign and intermediate) to in situ and subsequently invasive melanomas, and also from invasive RGP to VGP, and continuing in metastases.6 

Benign tumors of melanocytes are very common, generally termed melanocytic nevi, or often simply nevi. Although the term nevus reflects an old idea that these lesions are hamartomas, it continues to be used despite recent convincing evidence that nevi are benign neoplasms, having mutations or fusions of the same single driver oncogenes that also occur in melanomas, but are generally lacking the additional progression-related genomic changes.6  There is also an “intermediate” category of lesions that have 1 or a few of these progression-related genomic changes (such as hemizygous loss of CDKN2A or a TERT promoter mutation) but insufficient to establish the malignant clinical behavior of a melanoma.6  Benign nevi, and particularly the intermediate lesions, may provide challenges of diagnostic differentiation from melanomas in clinical and pathology practice. These challenges often result in diagnostic uncertainty, and the classification recognizes that, in some instances, definitive classification may not always be possible. When this occurs, it may be appropriate to use descriptive terms for them, such as intraepidermal atypical proliferation of uncertain significance (used for in situ proliferations), superficial atypical proliferation of uncertain significance (used for invasive RGP-only proliferations), or melanocytic tumor of uncertain malignant potential (used for tumorigenic lesions), accompanied by a differential diagnosis to allow for selection of rational clinical management.32  These terms, when used, are always written out in full.

These benign nevi along with their corresponding intermediate lesions and melanomas were placed by Bastian7  into an “integrated taxonomy” of melanocytic tumors, within which 9 pathways of cutaneous, mucosal, and uveal melanoma development were recognized.

Nodular melanomas likely occur in several of these pathways, representing lesions in which an intraepidermal or junctional component was present in most instances, but was overrun by an early-developing VGP tumor nodule.33  It has also been proposed that some NMs may arise from melanocytes that have lost tumor suppressor gene function first and then acquired a gain-of-function oncogenic alteration.34  Nodular melanoma often presents as a symmetrical tumor that can be pigmented or nonpigmented. They may be relatively small in diameter despite having reached a thickness that could be associated with a high mortality rate, and they often do not exhibit the “ABCDE” clinical signs and consequently are often not recognized clinically as melanoma.21  Histologically, NM is defined by a VGP without an adjacent RGP.15  They tend to grow rapidly, having a high mitotic rate,35  and may seem to be innocuous to the patient until they have reached a considerable Breslow thickness.36  Because of these distinctive characteristics, NM is discussed in the classification as a separate clinicopathologic entity.

The pathways of cutaneous and mucosal melanoma, with inclusion of simulant and precursor lesions, and their associated genomic aberrations, are listed in Tables 2 and 3.37 

Table 2

Classification of Melanomas and Precursor Lesions on the Basis of Epidemiologic, Clinical, Pathologic, and Genomic Attributes, Pathways I to III

Classification of Melanomas and Precursor Lesions on the Basis of Epidemiologic, Clinical, Pathologic, and Genomic Attributes, Pathways I to III
Classification of Melanomas and Precursor Lesions on the Basis of Epidemiologic, Clinical, Pathologic, and Genomic Attributes, Pathways I to III
Table 3

Classification of Melanomas and Precursor Lesions on the Basis of Epidemiologic, Clinical, Pathologic, and Genomic Attributes: Pathways IV to VIII

Classification of Melanomas and Precursor Lesions on the Basis of Epidemiologic, Clinical, Pathologic, and Genomic Attributes: Pathways IV to VIII
Classification of Melanomas and Precursor Lesions on the Basis of Epidemiologic, Clinical, Pathologic, and Genomic Attributes: Pathways IV to VIII

Pathway I: Low-CSD Melanoma/Superficial Spreading Melanoma

The concept of classifying melanomas based on the degree of CSD of the surrounding skin is based on the observation that melanomas on sun-exposed skin with little solar elastosis have genetic alterations distinctive from those on sun-exposed skin with marked elastosis.24,25  Most primary melanomas in the category originally termed non–cumulative solar damage in these genetic analyses were SSMs but other traditional melanoma subtypes such as NMs and unclassifiable melanomas were present as well. The absence of marked solar elastosis was the most powerful morphologic criterion to predict the genotype of BRAFV600E mutation and was more reproducible among expert pathologists than the traditional designations of SSM, LMM, and NM.38,39  Subsequently, more comprehensive genetic analyses have distinguished melanomas by anatomic site or inferred degree of cumulative sun exposure rather than the traditional McGovern-Clark categories and have confirmed the distinct patterns of genetic alterations within them.40,41  The WHO Melanocytic Tumours Working Group decided to merge these concepts into a low-CSD category that incorporates SSM. The definition of SSM remains the same as originally proposed by Clark et al15  and as represented in prior melanoma classifications.

Epidemiology

This is the most common form of melanoma in Western countries and early studies of “melanoma, not otherwise classified” will have captured data mostly relevant to this subtype.42  Following the landmark publication by Landcaster and Nelson43  in 1957, there was gradual recognition last century that cutaneous melanomas were related to sun exposure. In a seminal study, migrants from the United Kingdom to Australia were found to acquire the higher incidence of melanoma characteristic of the Australian population, but only if they had emigrated during childhood.44  This suggests that childhood sun exposure is crucial in establishing the risk for certain melanomas. However, there is evidence that exposure continuing in adult life also modifies the risk.18  In the low-CSD form of melanoma, prior cumulative sun exposure tends to be low to moderate, that is, insufficient to have caused marked solar elastosis, and typically occurs through “intermittent” exposure on weekends and on vacations. This subtype of melanoma is particularly localized to parts of the body that are exposed to the sun during these activities but not continuously throughout the typical “work week.” Thus, the commonest location for melanoma in men is the back while in women it is the back of the legs or calf region (although melanomas in both sexes occur in both of these locations with lesser frequency).45  This form of melanoma is also related to UV exposure in tanning beds, resulting in a significant though small epidemic of melanomas occurring in younger mostly female individuals.46  Other risk factors for low-CSD melanoma established in case-control studies include the total number of nevi, large size of nevi, and clinically atypical/dysplastic nevi.47  Correspondingly, risk factors for nevi overlap with those for melanomas. In a large case-control study, risk for melanoma was strongly related to number of small nevi, large nondysplastic nevi, and clinically dysplastic nevi. In the absence of dysplastic nevi, increased numbers of small nevi were associated with a 2-fold elevated risk, and increased numbers of both small and large nondysplastic nevi were associated with a 4-fold risk. One clinically dysplastic nevus was associated with a 2-fold risk, while 10 or more conferred a 12-fold increased risk.48  In 2 studies, a single histologically dysplastic nevus was associated with an approximately 4-fold risk,49,50  while lesional size greater than 4.4 mm was associated with a 5-fold risk.51  Although hereditary nevus susceptibility genes, in addition to those associations with melanoma risk, have been described, their functional roles and contributions to melanoma risk are unclear at this time.52 

Clinical Features

Most of the low-CSD melanomas fall into the category that was simultaneously described by McGovern16  as pagetoid melanoma and by Clark et al15  as SSM, as reviewed above. These terms represent prominent histopathologic and clinical features of the lesions, respectively. Like other melanomas that begin with an RGP phase, SSMs in their earliest forms present as patches of pigmentation on the skin that evolve into elevated plaques. Although initially essentially indistinguishable from benign junctional nevi, they gradually develop the distinctive “ABCDE” clinical characteristics.20,53 

Histopathology

The histopathology of SSM is defined primarily by aspects of its RGP, whether or not a VGP is present. In the RGP, there is a predominantly intraepidermal proliferation of large epithelioid melanocytes, arranged along the dermal-epidermal junction and having a high propensity for forming nests. There is also scatter of neoplastic cells into the epidermis termed pagetoid scatter because of its resemblance to scatter of breast cancer cells into the epidermis in Paget disease of the nipple or other sites. The lesions are often heavily pigmented and quite well circumscribed (Figure 1, A through D). In the dermis, there tends to be diffuse fibroplasia and there may be areas of loss of lesional cells consistent with partial (or sometimes complete) regression in the RGP. Some degree of solar elastosis is present in most cases of SSM/low-CSD melanoma but, by definition, is mild to moderate rather than severe. Mild or grade I CSD is defined as the presence of single elastotic fibers in the dermis visible at ×20 magnification. Moderate or grade II CSD is defined as the presence of altered fibers in bunches or fascicles. In contrast, severe or grade III CSD that is characteristic of high-CSD melanomas is defined by the presence of homogeneous clumps of elastotic material that have lost their texture of individual fibrils.54  There is often evidence of an associated nevus in SSMs, including superficial congenital pattern, common acquired, and dysplastic nevi.55 

Figure 1

Superficial spreading/low–cumulative solar damage melanoma. A, A complex lesion with a tumorigenic component in the left half of the image and a plaque component to the right, representing vertical growth phase and radial growth phase, respectively. B, In the radial growth phase, there is pagetoid scatter of uniformly atypical single melanocytes, and in addition there are quite prominent nests. There is 1 nest in the dermis that is smaller than the largest nest in the overlying epidermis. If not for the large vertical growth phase seen in (A), this could qualify the lesion as nontumorigenic invasive radial growth phase melanoma. C, The tumorigenic vertical growth phase is superficially ulcerated, as recognized by the presence of a serum crust and inflammatory reaction. D, In this example, the vertical growth phase is composed of heavily pigmented melanocytes, although pigmentation is highly variable and may be minimal or absent (hematoxylin-eosin, original magnifications ×20 [A], ×200 [B], ×50 [C], and ×400 [D]).

Figure 1

Superficial spreading/low–cumulative solar damage melanoma. A, A complex lesion with a tumorigenic component in the left half of the image and a plaque component to the right, representing vertical growth phase and radial growth phase, respectively. B, In the radial growth phase, there is pagetoid scatter of uniformly atypical single melanocytes, and in addition there are quite prominent nests. There is 1 nest in the dermis that is smaller than the largest nest in the overlying epidermis. If not for the large vertical growth phase seen in (A), this could qualify the lesion as nontumorigenic invasive radial growth phase melanoma. C, The tumorigenic vertical growth phase is superficially ulcerated, as recognized by the presence of a serum crust and inflammatory reaction. D, In this example, the vertical growth phase is composed of heavily pigmented melanocytes, although pigmentation is highly variable and may be minimal or absent (hematoxylin-eosin, original magnifications ×20 [A], ×200 [B], ×50 [C], and ×400 [D]).

Close modal

For practical purposes of classifying melanomas, any melanoma on nonglabrous skin with no, mild, or moderate solar elastosis should be classified as low CSD. For melanomas that arise in a background of grade III solar elastosis but show clear features of SSM such as marked pagetoid scatter, a predominance of large epithelioid melanocytes with powdery melanin pigmentation, or a contiguous melanocytic nevus as a likely precursor (as opposed to an incidental intradermal nevus as occasionally seen on the face), should also be classified as low-CSD/SSM.

Differential Diagnosis, Simulants, and Precursors

Although at the clinical level many other entities including pigmented seborrheic keratoses and basal cell carcinomas can simulate SSM, the most relevant simulants especially at the histologic level are melanocytic nevi, which are benign tumors of melanocytes. These may also act as precursors of some melanomas; however, the vast majority of nevi are stable and will never progress—indeed, the natural history of most nevi including dysplastic nevi appears to be involution.3  Categories of nevi in this “low CSD,” predominantly BRAF-mutated pathway, include common acquired nevi and dysplastic nevi (Table 2). Recently, deep penetrating nevi,56  BAP1-inactivated nevi,57  and a subset of pigmented epithelioid melanocytomas58  have been added to this category because of the presence of driver BRAF mutations. They represent branches of the low-CSD/SSM pathway characterized by specific secondary mutations that result in the formation of histopathologically distinctive lesions. Deep penetrating nevi often present as a combined lesion with a subset of the lesion representing a common acquired nevus. While both morphologically distinct areas harbor an identical MAP-kinase pathway mutation such as BRAFV600E, the pigmented spindle and epithelioid cell proliferation characteristic of deep penetrating nevi has an additional mutation in the WNT pathway, most commonly an activating β-catenin mutation.56 

In another pattern of combined nevus, there is biallelic inactivation of the tumor suppressor BAP-1, leading to a focal clone of partially transformed melanocytes presenting as a nodule of amelanotic enlarged epithelioid cells with vesicular nuclei (showing some resemblance to the cells of Spitz nevi) within a background nevus.57  These BAP1-inactivated spitzoid tumors mostly occur sporadically by somatic inactivation of both BAP1 alleles on chromosome 3. They can also occur in the setting of a cancer susceptibility syndrome, in which patients harbor a germline BAP1 mutation and often develop multiple BAP1-inactivated spitzoid tumors and are predisposed to a variety of cancers, including cutaneous and uveal melanoma.

Pigmented epithelioid melanocytoma is another intermediate lesion that resembles a blue nevus but has characteristic vesicular nuclei with prominent nucleoli. It is caused by the biallelic inactivation of another suppressor gene, PRKAR1A, typically in a conventional nevus with a BRAFV600E mutation.58,59 

Nevi with atypia, especially some dysplastic nevi, may be characterized by noncanonical BRAF mutations (non-V600E),6  or by NRAS60  or other driver mutations, and by the presence of a second genomic abnormality such as a TERT promoter mutation, or hemizygous loss of the CDKN2A gene at 9p21, which codes for the tumor suppressor p16.6 

Genomic Features

The most commonly mutated driver oncogene in SSM/low-CSD melanoma is BRAF.24,38  The most common mutation results in an amino acid substitution from a valine (V) to a glutamic acid (E) at position 600, p.V600E.61  This was the first mutation in melanoma to be targeted with inhibitory molecules that are designed to block the active site of this protein.62  The same mutation also occurs in most banal nevi.6  The genomic evolution of melanoma has been studied by Zeng et al,64  who analyzed melanomas with an adjacent nevus, melanoma in situ, or intermediate lesion. The point-mutation burden increased from benign through intermediate lesions to melanoma, with a strong UV mutation signature. Most intermediate lesions and melanomas in situ had TERT promoter mutations in addition to the initiating BRAF or NRAS mutation. Biallelic inactivation of CDKN2A marks the transition to invasive melanomas in most cases.63,64  PTEN and TP53 mutations were present only in advanced primary melanomas. Copy-number alterations emerged in intermediate and in situ lesions and continued to accumulate during progression to invasive and metastatic tumors.63,64  Tumor heterogeneity was observed in the form of genetically distinct subpopulations as melanomas progressed.6,63 

Pathway II: High-CSD Melanoma/Lentigo Maligna Melanoma

Epidemiology

This form of melanoma is less common than SSM/low-CSD melanoma but its incidence has been increasing especially in very heavily sun-exposed populations including outdoor workers but also some recreational “sun worshipers.” By definition, these melanomas arise in skin with severe or grade III CSD.55 

Clinical Features

In comparison with SSM, lentigo maligna melanoma tends to have a more poorly circumscribed border both clinically and histologically,38  with microscopic melanoma sometimes extending a considerable distance beyond the visible clinical border. This has been associated with increased propensity for local recurrence65  and has resulted in recommendations for LMM to be treated by excisions with wider clinical margins or comprehensive marginal evaluation such as by the Mohs technique, especially when on the face.66  As in all of the forms of RGP melanoma, the lesions evolve from patch to plaque stages and eventually most lesions will fulfill the ABCDE criteria. Tumorigenic VGP can evolve within these lesions at any time although the pace of progression of LMM seems to be slower than that of SSM. In some cases, the VGP is desmoplastic, as will be discussed in the next section. Pigmentation is less than in SSM and some LMM lesions are almost or completely amelanotic, which can result in their initially being misdiagnosed as an inflammatory process and also contributes to the problem of margin definition.

Histopathology

By definition, LMMs/high-CSD melanomas must demonstrate grade III solar elastosis. As in all melanomas, the characteristic histologic features are best appreciated when evaluating their RGP when this is present. The features are usually best evaluated near the periphery of the lesion because they may evolve toward a “final common pathway” of large cells with pagetoid scatter near their centers, overlapping with the pattern of SSM.67  Severe (or moderate to severe) solar elastosis is a requirement for diagnosis of high-CSD melanoma but alone is not sufficient (occasionally low-CSD melanomas/SSMs may occur in skin with high CSD). Compared to SSM, high-CSD melanomas have less nesting and a greater tendency to lentiginous (basal) proliferation of single cells along the dermal-epidermal junction (Figure 2, A through C). This lentiginous pattern, important in diagnosis of benign as well as malignant melanocytic proliferations, appears to be associated with driver mutations of distinct genes, such as NRAS and occasionally KIT.38  In contrast to solar and other lentigines, the rete ridges tend to be effaced rather than elongated, the epidermis is thinned, and the proliferation is at least focally continuous rather than intermittent. There may be a few nests near the tips and sides of elongated rete ridges, sometimes bridging between adjacent rete, in a pattern simulating a dysplastic nevus; however, in contrast to dysplastic nevi this pattern is focal within the lesion rather than symmetrically present around a central dermal nevic component, and is nonuniform across the lesion.68  There may be apparently skipped regions, and there may be evidence of RGP regression in the form of dermal fibroplasia and absence of lesional cells in the dermis and in the epidermis. These melanomas are typically not associated with a precursor nevus in contrast to low-CSD melanomas,69,70  and when nevus remnant cells are present the association may be incidental. Lesions that lack an RGP (ie, nodular melanomas) that occur in high-CSD skin could perhaps be classified as “high-CSD/nodular melanomas,” although this was not directly addressed in the 4th edition of the WHO Classification of Skin Tumours.71 

Figure 2

Lentigo maligna/high–cumulative solar damage melanoma. A, A broad lesion occurring in skin with severe chronic solar damage (solar elastosis in which fibers have to a large extent lost their fibrillary texture with formation of homogeneous material). B, In the radial growth phase, there are single cells predominating; however, as seen in this image a few nests are characteristically present, occasionally simulating a nevus, though generally present only focally within the broad radial growth phase. C, There is an invasive component composed of somewhat nevoid melanocytes; however, these resemble the melanoma cells in the junctional component and have little evidence of maturation or dispersion at the base, as would be characteristically seen in nevus cells (hematoxylin-eosin, original magnifications ×5 [A], ×200 [B], and ×100 [C]).

Figure 2

Lentigo maligna/high–cumulative solar damage melanoma. A, A broad lesion occurring in skin with severe chronic solar damage (solar elastosis in which fibers have to a large extent lost their fibrillary texture with formation of homogeneous material). B, In the radial growth phase, there are single cells predominating; however, as seen in this image a few nests are characteristically present, occasionally simulating a nevus, though generally present only focally within the broad radial growth phase. C, There is an invasive component composed of somewhat nevoid melanocytes; however, these resemble the melanoma cells in the junctional component and have little evidence of maturation or dispersion at the base, as would be characteristically seen in nevus cells (hematoxylin-eosin, original magnifications ×5 [A], ×200 [B], and ×100 [C]).

Close modal

Differential Diagnosis/Simulants and Precursors

Histopathologic differential diagnostic considerations include junctional and compound “banal” nevi and lentigines. Banal nevi are less broad than LMM, with most being less than 4 mm in diameter. Larger lesions are often dysplastic nevi, which may have cytologic atypia in addition to architectural disorder that may overlap at least focally with LMM. However, nevi in general should have a predominantly nested junctional component, with nests evenly distributed across the interface.72  The presence of focal bridging nests should not rule out LMM.68  A dermal component of a nevus should be centrally placed and have evidence of maturation. The dermal component of LMM may seem mature also (Figure 2, C); however, the dermal cells are not symmetrically distributed and may be multifocal, and they resemble the cells in the overlying in situ component. Lentiginous nevi may have a prominent component of single cells at the junction; the proliferation should be discontinuous and there should be less atypia than in LMM. Lesions termed lentiginous melanoma,73  and nevoid (variant of) lentigo maligna74  may overlap considerably with lentiginous nevi, and doubtful cases may be assigned a descriptive term such as intraepidermal atypical proliferation of uncertain significance or superficial atypical proliferation of uncertain significance, and managed according to the differential diagnosis. The diagnosis of “junctional nevus” or of “dysplastic nevus” on sun-damaged skin of the face in an older subject should be made with great caution, if at all.75  In the case of solar lentigines, there may be almost complete overlap with LMM in situ, when there is atypia and a tendency to confluent lentiginous proliferation at the junction, and in addition, changes of a solar lentigo may be seen in contiguity with an LMM, and could cause sampling confusion.76  It is not known whether these lesions are evolving precursors or lesions overrun by a developing in situ melanoma. Again, descriptive terminology and complete excision are appropriate forms of management.

Genomic Features

Driver mutations differ in LMM from those in SSM and include NF1, BRAFV600K, or other non-V600E mutations, NRAS,38  and to a lesser extent KIT.7,77,78  NF1 is a classical tumor suppressor gene and inactivation of both alleles is required to drive proliferation. When intact, it catalyzes the hydrolysis of GTP by RAS family members, accelerating their transition to the off state. When NF1 is inactivated, RAS stays longer in its activated state, resulting in a more sustained activation of the MAP-kinase pathway, thus driving proliferation.41  Because of the differences in the mutation patterns, the options for targeted therapy for metastatic LMM differ from those for SSM. These melanomas have a very high mutation burden, with predominant UV signature mutations.79  This high mutation burden may correlate with better responsiveness to checkpoint inhibitor immunotherapy.80 

Pathway III: Desmoplastic Melanoma

Epidemiology

Desmoplastic melanoma (DM) accounts for approximately 1% of melanomas in the United States. It most commonly arises on skin with high CSD. These may be a subtype of pathway II, at least in the high-CSD cases, but were thought to have sufficiently distinctive features to be classified independently in the 2018 classification, and are not necessarily associated with an RGP/in situ component. The desmoplastic component of these tumors presents as a spindle cell VGP with individual cells separated by collagen fibers, a “desmoplastic” pattern of growth, which is to be distinguished from cells lying in contiguity with one another. A morphologically similar desmoplastic pattern of growth may also be seen in some areas of high-CSD/LMM, and also in acral and mucosal lentiginous melanomas, which have little or no CSD.

Clinical Features

Desmoplastic melanoma may present as a firm scarlike tumor. The lesions are commonly amelanotic or sparsely pigmented, and the differential diagnosis of melanoma is not always apparent to the clinician.81  In other cases, there may be a preexisting pigmented patch within which a tumor develops. The lesions are typically endophytic rather than forming a nodule.

Histopathology

The histopathology has been recently reviewed.82  In most cases, there is an in situ/invasive RGP component, with general characteristics of LMM. Pigment is commonly sparse or absent. In some cases, there is an inconspicuous junctional proliferation that does not meet criteria for melanoma in situ, and in a small number of cases, there is no junctional component at all.83  These lesions were described as “nerve-centered dermal DM,”84  with the suggestion that they arise from dermal nerves; however, in many instances such a connection is not readily evident. The tumors in the dermis are composed of spindle cells that may have an undulating or wavy fiber pattern reminiscent of schwannian differentiation (Figure 3, A through C). “Pure” and “mixed” forms of DM have been described.85  In the pure component, the lesional cells are individually separated by delicate collagen fibers, which appear to have been synthesized by the tumor. In the mixed tumors, there is a component where the cells lie in contiguity with one another, which is an epithelial pattern of growth. In the mixed or epithelial areas, there may be mitoses and pigment may be present; however, these are generally absent in the pure DM components. The desmoplastic component is typically highly infiltrative and will extend down the septa of the panniculus in a subtle pattern that may involve specimen margins in an inconspicuous manner. A characteristic feature is the presence of nodular clusters of lymphocytes, which may correlate with the high mutation burden expressed in these tumors.86  By IHC, the cells of the “pure” component of DM are reactive with antibodies against pS100 and Sox10 but not with the more specific melanocytic markers such as HMB-45 and Melan-A/Mart1.87  However, in “mixed” DM, which has an epithelioid as well as a desmoplastic component and has a worse prognosis, there may be staining of the epithelioid component with these markers and also staining of the in situ component.

Figure 3

Desmoplastic melanoma, high cumulative solar damage. A and B, In this punch biopsy, there is a cellular tumor in the superficial dermis; however, the architecture of the reticular dermis is subtly altered and there are prominent nodular clusters of lymphocytes, close to the presence of involvement by desmoplastic vertical growth phase (H&E, 10x and 50x). C, In this deep dermal component, there are subtle spindle cells placed between altered collagen fibers, extending to the periphery of the specimen. A nodular cluster of lymphocytes is also illustrated (hematoxylin-eosin, original magnifications ×10 [A], ×50 [B], and ×200 [C]).

Figure 4. Atypical Spitz tumor/Spitz melanoma. No cumulative solar damage. A, In this lesion from the scalp of a 4-year-old child, there is a tumor that extends into the deep reticular dermis with a subcutaneous “satellite” nodule. B, The tumor is composed of large epithelioid melanocytes with abundant amphophilic cytoplasm and large nuclei with generally regular nuclear membranes, pale chromatin, and prominent nucleoli. These are pagetoid cytologic attributes. The mitotic rate was 4/mm2, which would be a high rate in an adult but borderline in a child. C, Tumor cells do not mature or disperse well at the base of the lesion and there is an associated lymphocytic response. This lesion was classified as of uncertain malignant potential. Genomic studies were not done. In a child, its behavior will likely be benign, and no adverse effects have been reported in several years of follow-up (hematoxylin-eosin, original magnifications ×5 [A], ×200 [B], and ×400 [C]).

Figure 3

Desmoplastic melanoma, high cumulative solar damage. A and B, In this punch biopsy, there is a cellular tumor in the superficial dermis; however, the architecture of the reticular dermis is subtly altered and there are prominent nodular clusters of lymphocytes, close to the presence of involvement by desmoplastic vertical growth phase (H&E, 10x and 50x). C, In this deep dermal component, there are subtle spindle cells placed between altered collagen fibers, extending to the periphery of the specimen. A nodular cluster of lymphocytes is also illustrated (hematoxylin-eosin, original magnifications ×10 [A], ×50 [B], and ×200 [C]).

Figure 4. Atypical Spitz tumor/Spitz melanoma. No cumulative solar damage. A, In this lesion from the scalp of a 4-year-old child, there is a tumor that extends into the deep reticular dermis with a subcutaneous “satellite” nodule. B, The tumor is composed of large epithelioid melanocytes with abundant amphophilic cytoplasm and large nuclei with generally regular nuclear membranes, pale chromatin, and prominent nucleoli. These are pagetoid cytologic attributes. The mitotic rate was 4/mm2, which would be a high rate in an adult but borderline in a child. C, Tumor cells do not mature or disperse well at the base of the lesion and there is an associated lymphocytic response. This lesion was classified as of uncertain malignant potential. Genomic studies were not done. In a child, its behavior will likely be benign, and no adverse effects have been reported in several years of follow-up (hematoxylin-eosin, original magnifications ×5 [A], ×200 [B], and ×400 [C]).

Close modal

Differential Diagnosis/Simulants and Precursors

Simulants of DM include low-grade spindle cell tumors and reactive conditions. A subset of nevi, called desmoplastic nevi, has a delicate fibrous stroma that can resemble that of DM.88  Many of these nevi are composed of large spindle and/or epithelioid cells with amphophilic hyaline cytoplasm and large ovoid nuclei with regular nuclear membranes, pale uniform chromatin, and prominent nucleoli. These could be regarded as atypical cells; however, these are spitzoid attributes and not characteristic of DM, and these lesions have been regarded as desmoplastic Spitz nevi, though genomic corroboration of this assignment is lacking, and this category has also been regarded as a distinct entity.89  Other desmoplastic nevi may be composed of smaller nevoid melanocytes, lacking atypia and mitotic activity, embedded in fibrotic stroma. Some of these may be neurotized nevi, which may simulate DM.90  However, DMs may also lack or only subtly express atypia and mitotic activity. Desmoplastic melanomas generally extend deeper than desmoplastic nevi, although superficial examples of DM may occur. The presence of nodular clusters of lymphocytes is characteristic of DMs but not of nevi. If an in situ component of a melanoma is present, usually of the lentigo maligna type (or of another lentiginous melanoma), the diagnosis of DM would be strongly favored. Reactivity for Melan-A and HMB-45 is most unusual in the desmoplastic component of DM (though not the epithelioid components of mixed DM), and this would strongly favor a nevus.88  Low-grade spindle cell proliferations such as atypical leiomyomatous tumor/leiomyosarcoma, dermatofibroma, and dermatofibrosarcoma protuberans may sometimes raise the differential diagnosis of DM, and can be distinguished by their specific morphology aided if necessary by IHC.71,91  Neurofibromas may express the same IHC markers as DM and have to be distinguished by consideration of the morphologic features reviewed above.92  Finally, mature and hyperplastic scars may be difficult to distinguish from DM, and conversely occasional examples of DM may mimic a scar. Reactivity for pS100 and Sox10 should distinguish these lesions; however, the staining of occasional cells within scars should not be overinterpreted as evidence of melanoma.93 

Genomic Features

High-CSD–associated DMs have an extremely high mutation burden with a very strong UV signature. Inactivating mutations of NF1 (neurofibromin),94  promoter mutations of NFKBIE, and diverse activating mutations in the MAP kinase pathway are observed.95  As previously mentioned, when NF1 is inactivated, RAS stays longer in its activated state, resulting in a more sustained activation of the MAP-kinase pathway, thus driving proliferation.41  Oncogenic mutations commonly found in other melanomas, in particular canonical mutations in BRAF and NRAS, are generally absent. Other genetic alterations known to activate the MAPK and PI3K signaling cascades have been identified, affecting NF1, CBL, ERBB2, MAP2K1, MAP3K1, BRAF, EGFR, PTPN11, MET, RAC1, SOS2, NRAS, and PIK3CA. These are not necessarily mutually exclusive. Some are candidates for targeted therapies.95 

These genomic features are quite distinctive and likely reflect a form of melanoma that evolves by the slow accumulation of weakly oncogenic mutations, an evolutionary trajectory distinct from that of most other melanoma subtypes, which begin with initiating mutations in strong oncogenes such as BRAF and NRAS.95 

Pathway IV: Spitz Melanoma

Definition and Epidemiology

In the past, melanomas have been classified as spitzoid on the basis of cytomorphologic features such as a predominance of large epithelioid cells. However, genomic analyses have revealed that most cases with such morphologic features have genomic characteristics of low-CSD melanomas, with frequent BRAFV600E mutations.96  In the revised WHO classification, we defined Spitz melanoma (SM) as the malignant counterpart of Spitz nevi (SN), defined morphologically and genomically. The spectrum from SN to SM is morphologically characterized by distinctive large spindle and/or epithelioid melanocytes and genetically by a different set of driver mutations that include HRAS, and fusion kinases involving ALK, ROS1, NTRK1, NTRK3, MET, RET, BRAF, and MAP3K8.30,97  Lesions with intermediate genetic and/or histopathologic characteristics are termed atypical Spitz tumors (ASTs). Spitz nevi occur most commonly in childhood, while ASTs and SMs are probably more common in older age groups, although conclusive data are lacking, and there may be confounding data because of the inclusion of “spitzoid melanomas,” most of which are likely low-CSD melanomas, genetically and biologically. Risk factors for the development of SN and melanomas are unknown.

Clinical Features

Lesions ultimately diagnosed as SM tend to differ from SN in being larger, sometimes ulcerated, and having a history of continuous progressive growth and change. Spitz nevi typically present as amelanotic papules or nodules, with symmetrical, well-circumscribed raised borders, and a shiny stretched epidermis covering the lesion. Occasional examples, especially in children, are ulcerated. There is typically a history of appearance and short-lived growth of the lesion, followed by a period of stability. Spitz melanoma would not be expected to undergo this cessation of growth.

The prognosis of SM is probably not accurately predictable by using prognostic attributes developed for usual melanomas. In particular sentinel node staging does not appear to be predictive of survival, even when positive. No study has reported a survival benefit for patients with AST undergoing sentinel lymph node biopsy or completion lymphadenectomy, and these procedures, although reasonable to consider in some cases, are not considered to be standard of care for AST or for SM.98 

Histopathology

The lesions are defined by the presence of large spindle and/or epithelioid melanocytes. In SN, these have abundant amphophilic hyaline cytoplasm and large nuclei with regular nuclear membranes, pale chromatin, and prominent nucleoli. The lesions usually have a junctional component composed of nests of these spitzoid cells, often with prominent clefting artifact with adjacent keratinocytes that are often hyperplastic. Globoid eosinophilic “Kamino bodies” are characteristically present at the interface.99  The cells protrude into the papillary dermis and often extend through it into the reticular dermis, having a tendency to “maturation” toward a smaller cell type at the base, with dispersion of single cells into reticular dermis collagen. The diagnosis of SM is difficult, as the criteria all depend on thresholds of differences from SN that are difficult to set (Figure 4, A through C). Proposed criteria (by no means all universally accepted) include older age, large size, asymmetry, poor circumscription, ulceration,100  and “consumption” of the epidermis,101  failure of maturation of the dermal component,102  increased mitotic rate with proposed thresholds for mitoses per square millimeter of fewer than 3 in adults103  or 6 or more in children,100  mitoses near the base,104  and the presence of a lymphocytic infiltrate.104,105  In a cohort follow-up study, the histologic features that most correlated with disease progression were frequent mitoses, deep mitoses, asymmetry, high-grade cytologic atypia, and ulceration.105  By IHC, SM may exhibit loss of staining with Melan-A/Mart1, loss of stratification with irregular staining for HMB-45, high Ki-67 proliferation rate106  (with a threshold proposed of >20% in a “hotspot”), and loss of staining for p16.107  The latter can indicate homozygous loss of chromosome band 9p21, which has been found to be more common at the malignant end of the spectrum. This locus may be important not only because of p16, but also because it is the locus of 2 other tumor suppressors, p14 and p15, which have relevance to melanoma progression.108  In one study, TERT promoter mutations were highly associated with fatal outcomes.109  The sensitivity, specificity, and predictive value of these features and tests have in general not been tested and this is difficult because appropriate gold standards do not exist. Lesions with some of these attributes, but insufficient for a diagnosis of SM, may be classified as AST, or designated as melanocytic tumor of uncertain malignant potential, with an appropriate differential diagnosis that may be used to plan rational therapy.

In addition to IHC as discussed above, ancillary genomic testing, including comparative genomic hybridization, fluorescence in situ hybridization (FISH), and gene expression profiling can be used to contribute to establishing the diagnosis of SM. In a seminal study, Gerami et al110  demonstrated that loss of chromosome band 9p21 (assessed by FISH) was associated with rare lethal behavior in a large group of atypical spitzoid lesions. Although these lesions were spitzoid, they had not been characterized genomically and may not have been true SM.

Imaging mass spectrometry analysis has also been proposed to differentiate SN from SM in formalin-fixed, paraffin-embedded tissue on the basis of proteomic differences.111 

Differential Diagnosis/Simulants and Precursors

The major differential diagnoses of SM are SN and AST. It is also important to distinguish true SMs from “spitzoid melanomas,” which lack the characteristic Spitz tumor genomic profiles. These lesions with “spitzoid” morphologic characteristics mostly represent examples of low-CSD NM or from one of the other pathways. Some of them can be distinguished by use of an immunohistochemical test with the anti–BRAF V600E antibody (VE1), which, if positive, excludes an SM.107 

The distinction between SN and SM has been discussed above. In a recent study of classical SN compared to melanomas (not necessarily spitzoid), statistically significant differences were found between SN and melanoma for the following features: pagetoid spread, atypia, maturation, elastosis, Kamino bodies, p16 expression, and the staining pattern of HMB-45. FISH testing supported the diagnosis in 36 of 37 cases.107  This study is limited in that it did not compare ASTs with Spitz or spitzoid melanomas, which is a crucial and difficult distinction. In another study of 18 SMs, the most useful parameters for the differential diagnosis were cell density, mitoses, zonation, infiltration pattern, and consumption of the epidermis.102 

BAP1-Inactivated Spitzoid Tumors

A subset of spitzoid lesions is associated with loss of expression of the tumor suppressor BAP1.112  These tumors (now classified in the low-CSD pathway because they characteristically express the BRAFV600E mutant protein) are predominantly intradermal, with occasional junctional involvement. The lesional cells have varying degrees of atypia ranging from nevoid cells with minimal atypia to very large, epithelioid cells with abundant amphophilic cytoplasm and well-defined cytoplasmic borders, and vesicular nuclei with prominent nucleoli that may be pleomorphic. Some tumors have marked atypical features, including nuclear pleomorphism, high cellularity, and increased mitotic activity. These often cannot be confidently classified as benign or malignant on histologic grounds, suggesting a spectrum ranging from clearly benign to potentially malignant. The cytologic characteristics resemble those seen of cells in Spitz tumors, but the lesions lack many histologic features of SN, such as epidermal hyperplasia, hypergranulosis, clefting around junctional nests, and Kamino bodies. Some tumors have an adjacent component of smaller nevus cells, as seen in common acquired and congenital pattern nevi, and are classified as combined nevi. The lesions can be diagnosed as BAP-1 deficient by using an antibody against this antigen, which demonstrates loss of expression in the nuclei of the spitzoid cells, but not in those of the background nevi. The background lesion and the cellular nodule characteristically express the BRAFV600E mutation,112  indicating that they are best placed in the “low CSD” melanoma category rather than in the category of Spitz tumor, despite morphologic overlap. These lesions can occur in the context of the BAP1 tumor susceptibility syndrome, in which patients have already inherited an inactivating mutation of 1 BAP1 allele and lost the remaining allele owing to a somatic event.113  The more common “sporadic” BAP1-inactivated Spitz tumors have lost both BAP1 alleles owing to somatic alterations. Counting the initiating BRAFV600E mutation (or other driver mutation) and the BAP1 inactivation, these tumors thus harbor 2 (syndromic) or 3 (sporadic) somatic mutations, placing them in the “intermediate” category of tumor progression. Nevertheless, most behave in a clinically benign manner.

Genomic Features

The genomic alterations of the various forms of Spitz tumors are distinctive, with mutually exclusive, constitutively active kinase fusions in ROS1, NTRK1, NTRK3, ALK, BRAF, MET, and RET genes having been identified to date.27,28,114  Another subset of Spitz tumors has a point mutation in HRAS, typically accompanied by a gain of chromosome arm 11p, where HRAS resides.115  TERT promoter mutations and loss of the chromosome 9p21 region, which contains the tumor suppressor CDNK2A (and also CDNK2B), affecting P16 and P14ARF, or P15, respectively, have been identified in a few of the rare aggressive and occasionally lethal tumors that represent true SM109,116,117 ; however, none of these changes alone is specifically diagnostic of malignancy.107,118,119  The BAP-1–deficient lesion has been described in the previous section.

Pathway V: Acral Melanoma

Epidemiology

Acral melanoma (AM) refers to melanoma occurring in the glabrous, that is, non–hair-bearing skin of the volar aspects of the fingers and toes, palms and soles, and nail beds. Melanomas occurring on the dorsal aspects of these sites may represent CSD-related melanomas. Glabrous skin lacks hairs and has a thick stratum corneum, which acts as a barrier to penetration of UV into the underlying epithelium and dermis by scattering the light. Acral melanomas occur with approximately similar frequency in most ethnic groups around the world, and in populations not susceptible to CSD melanoma (such as in persons of Asian and African descent and in other populations of color); this is the most frequent subtype mainly owing to a reduced incidence of low- and high-CSD melanomas in these populations.120  The etiology of these melanomas is unclear. UV radiation does not play a significant role and it has long been suspected that these lesions might be induced by trauma. And 2 recent independent studies121,122  have shown that AMs commonly occur in regions of physical stress, such as flexure lines and the heel region, perhaps on the basis of repetitive motion/trauma and injury preferentially occurring at these sites. Prognosis is typically poor for AM, undoubtedly owing to a tendency for diagnosis at an advanced stage, but also perhaps to substantive differences from other subtypes.123 

Clinical Features

As in SSM, LMM, and other melanomas with an RGP, AMs begin with a patch lesion that enlarges more or less radially.123125  These lesions may form a plaque as they begin to involve the dermis and cause epidermal thickening; however, the thick stratum corneum often results in a lesion that remains flat in relation to the surrounding skin. Usual ABCDE characteristics apply in these lesions. When VGP ensues, the lesions may become ulcerated and a nodule may protrude through the ulcerated stratum corneum and form a protuberant VGP.

Histopathology

Acral melanomas most commonly present with a lentiginous pattern of proliferation (Figure 5, A through C), and have been termed acral lentiginous melanomas (ALMs)124,125 ; however, pagetoid melanomas also occur in these sites. There is evidence that these pagetoid melanomas may resemble SSM genomically and likely belong to the low-CSD melanoma pathway.126,127  The ALMs are notoriously poorly circumscribed—the last cells at the periphery of the lesions are single cells rather than nests—and there is evidence that genomic abnormalities are present in morphologically normal melanocytes beyond the periphery of the histologically recognizable RGP, constituting a “field effect.”128  This perhaps, likely in addition to compromise of therapy in order to minimize functional consequences of wide excisions, may contribute to the well-known propensity of these lesions to recur locally. The criteria for diagnosis of ALM may overlap with those of a subset of acral nevi and are discussed more extensively in the next paragraph. As mentioned above, the VGP may be composed of spindle cells with or without a desmoplastic pattern of growth, which likely differs from DM in high-CSD skin, for example, in having a lower tumor mutation burden. These melanomas are more likely to be associated with neurotropism,129  and it is not uncommon for AMs, especially subungual ones,130  to be seen invading into bone, perhaps because the bone is superficially located in these sites.

Figure 5

Acral lentiginous melanoma in situ. No cumulative solar damage. A, In this melanoma reexcision specimen, there is obvious cellular proliferation at the interface especially toward the left of the specimen. B, In this region, there are a few nests and there are single cells along the junction (a lentiginous pattern), associated with a focally prominent infiltrative lymphocytic response. C, At the periphery of the specimen, there is a much less cellular proliferation, which is also amelanotic, constituting subtle involvement of the resection margin, an important consideration in the evaluation of resections of lentiginous melanomas (hematoxylin-eosin, original magnifications ×50 [A], ×200 [B], and ×100 [C]).

Figure 6 Mucosal melanoma. No cumulative solar damage. A, In this resection of an anal lesion, there are 2 separate bulky vertical growth phase nodules, associated with a radial growth phase component. B, The radial growth phase component is predominantly lentiginous, with invasion of the lamina propria by cells that differ from those of the vertical growth phase nodules. C, In a vertical growth phase nodule, superficially ulcerated, heavily pigmented spindle cells predominate, with numerous associated melanophages (hematoxylin-eosin, original magnifications ×5 [A], ×100 [B], and ×200 [C]).

Figure 5

Acral lentiginous melanoma in situ. No cumulative solar damage. A, In this melanoma reexcision specimen, there is obvious cellular proliferation at the interface especially toward the left of the specimen. B, In this region, there are a few nests and there are single cells along the junction (a lentiginous pattern), associated with a focally prominent infiltrative lymphocytic response. C, At the periphery of the specimen, there is a much less cellular proliferation, which is also amelanotic, constituting subtle involvement of the resection margin, an important consideration in the evaluation of resections of lentiginous melanomas (hematoxylin-eosin, original magnifications ×50 [A], ×200 [B], and ×100 [C]).

Figure 6 Mucosal melanoma. No cumulative solar damage. A, In this resection of an anal lesion, there are 2 separate bulky vertical growth phase nodules, associated with a radial growth phase component. B, The radial growth phase component is predominantly lentiginous, with invasion of the lamina propria by cells that differ from those of the vertical growth phase nodules. C, In a vertical growth phase nodule, superficially ulcerated, heavily pigmented spindle cells predominate, with numerous associated melanophages (hematoxylin-eosin, original magnifications ×5 [A], ×100 [B], and ×200 [C]).

Close modal

Differential Diagnosis/Simulants and Precursors

The differential diagnosis of AM includes primarily acral nevi. Acral nevi are associated with ethnicity, pigmentation, age, and cutaneous melanoma risk factors including other nevi and atypical nevi.131  In a consecutive series of 165 plantar nevi, a group of 36 distinctive nevi were designated “acral-lentiginous nevi.”132  Compared to most acral nevi, these were characterized by “elongation of rete ridges, contiguous proliferation of melanocytes at the dermoepidermal junction, presence of single scattered melanocytes, or less commonly small clusters, within the upper epidermis, poor or absent lateral circumscription, melanocytes with abundant pale cytoplasm and round to oval, sometimes hyperchromatic, nuclei and prominent: nucleoli present at the dermoepidermal junction.”132  These are features shared with many melanomas, especially subtle early lesions or changes at the periphery of established lesions. Anastomosing rete ridges, cytologic atypia, and well-formed lamellar fibroplasia as seen in dysplastic nevi were absent. Criteria that distinguish these nevi from melanoma were “lack of pagetoid lateral spread, absence of mitotic activity in the deep dermal component, and the evidence of dermal nevocytic differentiation.” To these criteria we would add smaller size and greater symmetry of the nevi compared to the melanomas.

Dermoscopically, “parallel ridge” and “parallel furrow” patterns are recognized in ALM and in acral nevi, following the dermatoglyphics. The sensitivity and specificity of the parallel ridge pattern in diagnosing early acral melanoma are said to be 86% and 99%.133,134  Histologically, if melanin granules in the cornified layer are detected as melanin columns regularly distributed under the surface furrows (which can be enhanced with Fontana-Masson staining), the lesion is “strongly suggested to be a benign acral nevus.”134  This finding depends on sectioning of the specimen perpendicular to the ridge and furrow pattern.

The genetic events in acral nevi parallel those of AMs, including frequent copy number variations,126  differing in this regard from nevi of other sites. In a study by FISH, no abnormalities were seen in 36 acral nevi, differing from the findings in 44 AMs, for which the sensitivity of diagnosis was 88.6%.135 

Genomic Features

Acral melanomas have a relatively low burden of point mutations and a high incidence of copy number variation with multiple amplifications of genes such as CCND1 (cyclin D1) and KIT.25,136,137  Somatic TERT translocations, copy gains, and missense and promoter mutations, or germline events, were recently described in 41% of patients.138  Mutually exclusive mutations of BRAF, NRAS, and KIT are seen in a subset of cases,126,139  and also kinase fusions have been identified.31  Some of these events may represent examples of melanomas of other pathways occurring in acral sites.

Pathway VI: Mucosal Melanoma

Epidemiology

Defined as melanoma occurring in a mucous membrane, these lesions occur most commonly in genital sites, in the oral and nasal cavities, and the conjunctiva, and rarely other mucosae.140  These lesions occur with about equal frequency in all races and therefore form a substantial fraction of the melanomas that occur outside of the high-risk regions populated by whites. Risk factors are largely unknown, as there is no known association with sun exposure141  or evidence for a pathogenic role of chemical carcinogens or viruses.

Clinical Features

Mucosal melanomas may evolve through an RGP that presents “ABCDE” features and may be recognized clinically in visible regions such as the vulva, the oral cavity, and the conjunctiva.142  Lesions occurring in nasal sinuses and occasionally in visceral organs are almost never recognized when entirely in the RGP. Because of the difficulties in visualizing these lesions, they commonly present as a bulky tumor that invades and destroys surrounding tissues, sometimes presenting with bleeding and sometimes with pain or discomfort. In a recent study, vulvar and vaginal melanomas had similar molecular characteristics,143  even though vulvar lesions usually involve skin, indicating that these are closely related and differ from nongynecologic mucosal melanoma (see Genomic Features).

Histopathology

The RGP of mucosal melanomas typically presents a lentiginous pattern of growth of single cells tending to become confluent along the interface region of usually squamous mucous membranes,142  and these lesions have been called mucosal lentiginous melanomas.144  There is typically no evidence of solar elastosis,140  except in exposed sites like the conjunctiva, where solar damage may be etiologic. However, genetic analyses of conjunctival melanomas have revealed that these are related to melanomas from other pathways (ie, a mixture of high-CSD and low-CSD melanomas).145  There may be a tendency for nesting and for pagetoid scatter into the epithelium but these tend to be relatively limited compared to SSM and occur when the lesion is more advanced. When VGP eventuates, it forms a tumor comparable to that in other pathways of melanoma (Figure 6, A through C). A desmoplastic pattern of VGP is sometimes seen; this likely differs in many respects including tumor mutation burden from DM in high-CSD skin.

Differential Diagnosis

The major simulators of superficial mucosal melanomas are mucosal melanosis, mucosal lentigines, and atypical mucosal nevi (the latter also simulate tumorigenic melanomas). Occasionally, a nonmelanocytic tumor near a mucocutaneous junction may contain melanin pigment produced by reactive melanocytes, and in addition, metastatic melanomas to mucosal surfaces need to be distinguished from tumorigenic primary melanomas.

Mucosal lentigo, as in other sites, is defined as a patch of hyperpigmentation of basal keratinocytes, with an increased number of melanocytes.146  Clinically, such lentigines and other macular hyperpigmentations including melanosis and melanoacanthoma may be referred as melanotic macules, a histologically nonspecific term.146  There is overlap clinically and histologically between mucosal lentigines and mucosal melanosis, in which there is hyperpigmentation but the number of melanocytes is not increased, and with mucosal melanoma in situ, in which there is melanocytic atypia and usually at least focal contiguous proliferation of melanocytes along the junction. By definition, nests of melanocytes are absent in a lentigo, and if they were present the differential diagnosis would be between a (lentiginous) nevus and melanoma. Extensive continuous and contiguous proliferation of atypical cells, as well as the presence of some nests, raises concern for melanoma in situ. The literature on mucosal lentigines is scant. In one study, it was noted that melanoma in situ and mucosal melanosis were indistinguishable clinically in a patient with oral mucosal melanoma.147  It is important to be aware that mucosal lentigines, with varying degrees of atypia, may be seen in the same sites as mucosal melanomas, including especially oral mucosa and the mucocutaneous surfaces of the genitalia, especially the vulva, often presenting considerable difficulties of differential diagnosis and management.148 

Genomic Features

The somatic mutation burden is lower than that for CSD melanomas and there are more numerous structural variations.149  KIT136,149,150  and NRAS151  mutations have been described in a proportion of tumors, but BRAF mutations are uncommon,149  although oncogenic BRAF fusions have recently been identified.151  In contrast, conjunctival melanomas are probably a mixture of high-CSD melanoma, having evidence of solar elastosis and genomic changes indicative of high UV exposure, and of low-CSD melanomas.152,153 

Pathway VII: Melanoma Arising in a Congenital Nevus

Epidemiology

Congenital nevi, defined as melanocytic nevi present at birth, occur in about 1% of newborns.154  Most of these are small lesions clinically not distinguishable from acquired nevi that develop in later life. The congenital nevi are divided into 3 subsets: giant or garment nevi that cover whole regions of the body and are usually not able to be excised; intermediate congenital nevi susceptible to surgical excision; and small congenital nevi defined as less than 2.5 cm in diameter (still considerably larger than most acquired nevi). Melanomas that occur in giant congenital nevi tend to occur during childhood155  and apparently with a lesser frequency throughout life. Estimates of the lifetime incidence of melanoma in large congenital nevi range widely from 1% to 30%. In a comprehensive review, prospective studies of academic referral centers showed significantly lower average rates of 2% to 5%, in which the average follow-up period varied from 4.5 to 7.3 years.156  In a registry study, patients with a giant nevus had a 51.6-fold higher risk of developing a melanoma when compared with the general population rates.157  Intermediate and smaller nevi have been less well studied because of difficulties of ascertainment, definition, and follow-up, but the risk of melanoma arising within any individual lesion is much lower than for giant nevi.

Clinical Features

Melanomas may develop in the junctional component or in the dermal component of congenital nevi.156  Lesions that develop in a junctional component with an RGP may have characteristics similar to those of low-CSD melanomas, while lesions that develop in the dermal component have distinctive characteristics. The developing melanoma may be clinically masked by the background pigmented, and often hairy, nevus. These need to be distinguished from the phenomenon of cellular and proliferative nodules in congenital nevi, which are benign lesions typically arising during the first year of life, usually but not always requiring biopsy.158 

Histopathology

Melanomas that develop in the junctional component of congenital nevi usually resemble SSM or occasionally LMM histologically.156  They may progress from in situ to superficially invasive RGP and to VGP nodules. The background congenital nevus is present in contiguity with the melanoma. Melanomas that develop in the dermal component of a nevus present difficulties of accurate diagnosis (Figure 7, A through D). They may present as nodules and tumor masses, which may be composed of epithelioid, spindled, or “small round blue” cells, and may exhibit features of various sarcoma types, such as malignant schwannoma, rhabdomyosarcoma, and liposarcoma.156,159  Distinction between authentic dermal melanomas and atypical proliferative nodules may be very difficult; however, genomic analysis can be of assistance (see below).

Figure 7

Melanoma in a congenital melanocytic nevus. No cumulative solar damage. A, There is a bulky nodule that presented as a rapidly growing tumor of the back in a giant congenital nevus in a 4-year-old child. The tumor was not present at birth. B, The nodule contrasts with the background nevus, although there is a subtle tendency to blending between the 2 components. C, The nodule is composed of “small round blue cells,” which are quite uniform. D, There is nuclear molding and there are numerous mitoses. Genomic studies could be helpful in considering a diagnosis of proliferative nodule versus melanoma in such an instance. This lesion behaved aggressively, with liver and bone metastases, and ultimately caused the death of the patient within about a year of its presentation (hematoxylin-eosin, original magnifications ×5 [A], ×100 [B], ×200 [C], and ×400 [D]).

Figure 7

Melanoma in a congenital melanocytic nevus. No cumulative solar damage. A, There is a bulky nodule that presented as a rapidly growing tumor of the back in a giant congenital nevus in a 4-year-old child. The tumor was not present at birth. B, The nodule contrasts with the background nevus, although there is a subtle tendency to blending between the 2 components. C, The nodule is composed of “small round blue cells,” which are quite uniform. D, There is nuclear molding and there are numerous mitoses. Genomic studies could be helpful in considering a diagnosis of proliferative nodule versus melanoma in such an instance. This lesion behaved aggressively, with liver and bone metastases, and ultimately caused the death of the patient within about a year of its presentation (hematoxylin-eosin, original magnifications ×5 [A], ×100 [B], ×200 [C], and ×400 [D]).

Close modal

Differential Diagnosis/Simulants and Precursors

Nodular malignant tumors that occur in congenital nevi need to be distinguished from cellular and proliferative nodules, which occur quite frequently in giant congenital nevi, typically in childhood.160  These nodules are composed of cells that are usually larger than those in the background nevus. There may be a tendency for blending with the background nevus, or there may be a sharp demarcation. These lesions may have a few or even many mitoses, or there may be no mitoses at all, and such amitotic lesions may be termed cellular rather than proliferative nodules.158  Unusual differentiation that may occur in melanoma nodules in congenital nevus does not typically occur in the cellular nodules. Characteristics that can differentiate the lesions from melanoma include lack of high-grade uniform cellular atypia; lack of ulceration and of necrosis within the nodule; rarity of mitoses; evidence of maturation in the form of blending or transitional forms between the cells in the nodule and the adjacent nevus cells; lack of pagetoid spread into the overlying epidermis; and no destructive expansile/infiltrative growth.158  Genomic studies can be helpful in the distinction (see below).

Genomic Features

NRAS mutations are the most common drivers in large and intermediate congenital nevi,161,162  and in the melanomas that arise in them.163  Lesions known as congenital pattern nevi, which are generally less than 1 cm in diameter and have lesional cells extending into the reticular dermis and/or are around or within skin appendages but were not present at birth, usually have BRAF mutations and probably represent a subset of acquired nevi.161,162  In subsets of melanoma, including those arising in giant congenital nevi, TERT expression may be upregulated epigenetically by a methylation-dependent mechanism,164  while the tumors retain the wild-type genotype.163  Proliferative nodules, in contrast to melanomas, tend to have whole chromosome number aberrations.160  In a recent study of 2 lethal melanoma nodules compared to 22 proliferative/cellular nodules, the lethal melanomas both featured expansile nodules of epithelioid melanocytes with high mitotic counts (5–20 mitoses/mm2), and an ulcerated overlying epidermis. At the genomic level, the proliferative nodules had mostly whole chromosomal copy number aberrations, in some cases accompanied by rare partial chromosomal aberrations, whereas lethal melanomas had highly elevated copy number aberrations involving 6p25 without gains of the long arm of chromosome 6,160  and/or homozygous loss of 9p21,165  suggesting that these quite dramatic differences may be reliably predictive of behavior even in atypical cases; however, direct evidence for this is currently lacking.

Pathway VIII: Melanoma Arising in Blue Nevus

Epidemiology

Blue nevi (BN) are relatively uncommon and risk factors for their occurrence are unknown.

Clinical Features

Several categories of BN are recognized, most importantly the common lesions variously termed banal, Jadassohn, usual, or dendritic BN, and the less common cellular blue nevi (CBN). Other subtypes include epithelioid BN and plaque-type BN,166  and hypopigmented and sclerosing BN.167  Typical BN are composed of a relatively sparsely distributed population of pigmented spindle cells with thin dendritic cytoplasmic processes located among sometimes thickened reticular dermis collagen fiber bundles. Cellular blue nevi in addition have areas of confluence of cells that may have clear cytoplasm and form nests. In a characteristic “mixed-biphasic” pattern, fascicles of spindle cells extend between nests of cells with partially clear cytoplasm.168  Lesions often have a bulbous expansion at the base extending into the subcutis. Melanoma arising in blue nevus (MBN) is a term that is preferred to the previously commonly used “malignant blue nevus,” because the melanomas occur as a new population of cells usually developing in the background of a CBN, which itself often occurs in a background of a more banal BN. In a recent review of 91 cases, the mean age at diagnosis was 45 years, with a slight male predominance. Metastases were reported in 55% (n = 50), of which 16 had metastases at the time of diagnosis, 16 developed metastases within the first year, and 18 within 5 years of initial diagnosis. The mean Breslow thickness was 6.8 mm at the time of diagnosis (n = 39).169 

Histopathology

MBN presents as a tumorigenic proliferation within a background lesion, usually a CBN, and is usually diagnosed relatively late because early changes are perhaps obscured by the presence of the precursor lesions (Figure 8, A through C). Ulceration may occur; however, often the lesions are deep-seated and are recognized only because of an increase in size of a long-standing preexisting lesion. These melanomas are characterized by tumorigenic proliferation of uniformly large cells with marked anaplastic cytologic atypia, frequent mitoses, and usually the presence of necrosis or ulceration.166  The diagnosis is therefore usually not in doubt; however, there is probably some morphologic overlap of potentially aggressive cases with that of atypical CBN, leaving room for doubt regarding the diagnosis in some cases.170  Such lesions probably represent intermediate forms of progression from CBN to melanoma. Genomic studies can assist in these distinctions (see Genomic Features below).

Figure 8

Melanoma arising in blue nevus. No cumulative solar damage. A, There is a bulky nodule in the subcutis. B, In the nodule, there is a highly cellular proliferation of uniformly atypical melanocytes with frequent mitoses, histologically diagnostic of malignancy. C, In the background, there are “mixed-biphasic” changes of a cellular blue nevus (hematoxylin-eosin, original magnifications ×5 [A], ×200 [B], and ×50 [C]).

Figure 8

Melanoma arising in blue nevus. No cumulative solar damage. A, There is a bulky nodule in the subcutis. B, In the nodule, there is a highly cellular proliferation of uniformly atypical melanocytes with frequent mitoses, histologically diagnostic of malignancy. C, In the background, there are “mixed-biphasic” changes of a cellular blue nevus (hematoxylin-eosin, original magnifications ×5 [A], ×200 [B], and ×50 [C]).

Close modal

Differential Diagnosis/Simulants and Precursors

The differential diagnosis of MBN includes BN/CBN, and also melanomas that simulate MBN but can be distinguished by lacking the defining features of a background BN and, more recently, by the characteristic genomic abnormalities.171  Metastatic melanoma can simulate MBN and can be distinguished by the history and clinical workup, and the lack of a background lesion, perhaps supported by genomic studies.172  A subset of primary melanomas not originally diagnosed as MBN also contains the characteristic genomic changes, and may perhaps appropriately be reclassified.171 

Genomic Features

In a study using comparative genomic hybridization of CBN and malignant BN, the number of chromosomal aberrations (3 or fewer versus more per lesion) correlated with cytologic atypia, a high mitotic rate, the presence of necrosis, and with a diagnosis of malignancy and with aggressive behavior.173  The genetic alterations in melanomas in BN are distinctive and overlap extensively with those of uveal melanoma.174,175  Driver mutations occur in the G protein signaling pathway, most often in the genes GNAQ and GNA11, and infrequently in PLCB4 or CYSLTR2 (in both BN and associated melanoma). EIF1AX, SF3B1, and BAP1 mutations (characteristically seen in uveal melanomas) are also present in a subset of MBN cases, with BAP1 and SF3B1 R625 mutations being present only in clearly malignant tumors. In biphasic lesions with a BN and MBN component, the secondary alterations in BAP1, SF3B1, or EIF1AX are confined to the MBN component, indicating that they are responsible for the malignant transformation. Testing for these mutations, particularly BAP1 IHC, can be a useful diagnostic adjunct to confirm malignancy where there is diagnostic doubt.6,171,176  Copy number aberrations are more common and often complex in melanomas in BN compared with CBN and atypical CBN. Gains and losses of entire chromosomal arms have also been identified including gains of 1q, 4p, 6p, and 8q, and losses of 1p and 4q.173,177 

Nodular Melanoma

Epidemiology

Nodular melanomas most likely can occur in any of the pathways discussed above, and therefore their epidemiologic and genomic features are likely to be heterogeneous.

Clinical Features

Nodular melanomas have a papular or nodular configuration on clinical evaluation.178  They may be pigmented and the pigment may be homogeneous or heterogeneous; however, they are commonly amelanotic, presenting as a pink papulonodular lesion. Because they are tumorigenic from close to their initiation, NMs present as rapidly growing lesions. Nodular melanomas have a worse prognosis, on the average, than other melanomas, but this difference disappears, perhaps not completely,179  when multivariable analyses are done.

Histopathology

Despite being relatively small in diameter, NMs can have a significant Breslow depth. However, they share the common feature of being tumorigenic proliferations of uniformly atypical mitotically active neoplastic melanocytes (Figure 9, A through E). The tumors are commonly ulcerated. They are characteristically elevated above the epidermis, indicating accretive growth in an upward direction. A few lesions have a predominantly tumorigenic configuration but may have a few atypical melanocytes in the epidermis. By convention, if these extend beyond 3 rete ridges, they may be considered to represent a preexisting RGP, which can then potentially be classified into one of the other pathways. Otherwise, lesional cells that involve the epidermis in these lesions are quite likely to have been derived from the expanding dermal nodule.

Figure 9

Nodular melanoma (from Cochran et al,185  with permission. International Agency for Research on Cancer. World Health Organization. Elder DE, Massi D, Scolyer RA, Willemze R, eds. WHO Classification of Skin Tumours. 4th ed. Lyon, France: IARC; 2018). A, There is a nodular tumor elevating the epidermis with a collaret on the right-hand side and with a remnant of a nevus on the left. B, The precursor nevus cells contrast with those of the nodule. C, The lesional cells are large, with uniformly atypical nuclei and frequent mitoses. D, There is only slight evidence of maturation to a smaller cell type at the base. E, On the right-hand side there is a collaret and there is no associated in situ or invasive radial growth phase component (hematoxylin-eosin, original magnifications ×5 [A], ×50 [B and E], and ×200 [C and D]).

Figure 9

Nodular melanoma (from Cochran et al,185  with permission. International Agency for Research on Cancer. World Health Organization. Elder DE, Massi D, Scolyer RA, Willemze R, eds. WHO Classification of Skin Tumours. 4th ed. Lyon, France: IARC; 2018). A, There is a nodular tumor elevating the epidermis with a collaret on the right-hand side and with a remnant of a nevus on the left. B, The precursor nevus cells contrast with those of the nodule. C, The lesional cells are large, with uniformly atypical nuclei and frequent mitoses. D, There is only slight evidence of maturation to a smaller cell type at the base. E, On the right-hand side there is a collaret and there is no associated in situ or invasive radial growth phase component (hematoxylin-eosin, original magnifications ×5 [A], ×50 [B and E], and ×200 [C and D]).

Close modal

Differential Diagnosis/Simulants and Precursors

Nodular melanomas must be differentiated from other pink (or variegated) papules, including lesions such as dermatofibromas, nevi, neurothekeomas, neurofibromas, and skin appendage tumors. A difficult problem is posed by superficial metastases of melanoma to the skin as these may be epidermotropic, involving the overlying epidermis and thus closely resembling a primary melanoma. In a recent study, features significantly associated with epidermotropic metastatic melanoma included “a tumor size of less than 2 mm, an absence of tumor-infiltrating lymphocytes and plasma cells, monomorphism, and involvement of adnexal epithelium.”180  The presence of lymphovascular invasion may also be a differentiating feature.181  Lack of extension of the epidermal component beyond the borders of the dermal component was once emphasized, but there are occasional exceptions to this and other “rules.”182  Features associated with primary NM included “a polypoid (exophytic) configuration, prominent tumor-infiltrating plasma cells (TIPs), a tumor size greater than 10 mm, ulceration, epidermal collarets, a higher mitotic rate, necrosis, multiple phenotypes, significant pleomorphism, and lichenoid inflammation.”180  In multivariate analysis, a logistic regression model including large tumor size, ulceration, prominent TIPs, lichenoid inflammation, and epidermal collarets was highly predictive of primary NM.180  Rare epidermotropic melanomas may have an “epidermal-only” or “epidermal-predominant” pattern closely simulating in situ or microinvasive melanoma.180,183  Other epidermotropic metastatic melanomas and superficial dermal metastatic melanomas are well differentiated and may simulate nevi (nevoid or differentiated epidermotropic metastatic melanoma or epidermotropic metastatic melanomas with maturation).184  Genomic studies could be helpful in this distinction.

Genomic Features

Studies of hotspot mutations in BRAF and NRAS, and genome-wide copy number analyses have indicated that NMs share the genetic alterations of other melanomas arising in similar settings (CSD or anatomic site).25,38 

We have provided a summary of a classification of melanoma that builds on previous work and distinguishes 9 distinct types of melanoma development based on their epidemiology, clinical and histologic morphology, and genomic characteristics (including uveal melanoma, which is not discussed in detail here). Wherever appropriate based on currently known data, each melanoma subtype is placed in a position at the end of an evolutionary lineage (or “pathway”) that is founded in its respective precursor lesion. Each precursor subtype has a variable, usually low, risk of progression through stages of evolution culminating in an invasive and tumorigenic melanoma, which in turn has a variable risk of metastasizing and causing death based on continuing evolution.

1
Elder
DE
,
Barnhill
RL
,
Bastian
BC
, et al.
Melanocytic tumour classification and the pathway concept of melanoma pathogenesis
.
In
:
Elder
DE
,
Massi
D
,
Scolyer
RA
,
Willemze
R
,
eds
.
WHO Classification of Skin Tumours. 4th ed
.
Lyon, France
:
IARC;
2018
:
66
71
.
World Health Organization Classification of Tumours; vol 11
.
2
Elder
DE
.
Precursors to melanoma and their mimics: nevi of special sites
.
Mod Pathol
.
2006
;
19
(
suppl 2
):
S4
S20
.
3
Halpern
AC
,
Guerry
D
,
Elder
DE
,
Trock
B
,
Synnestvedt
M
,
Humphreys
T.
Natural history of dysplastic nevi
.
J Am Acad Dermatol
.
1993
;
29
(
1
):
51
57
.
4
Bevona
C
,
Goggins
W
,
Quinn
T
,
Fullerton
J
,
Tsao
H.
Cutaneous melanomas associated with nevi
.
Arch Dermatol
.
2003
;
139
(
12
):
1620
1624
.
5
Tsao
H
,
Bevona
C
,
Goggins
W
,
Quinn
T.
The transformation rate of moles (melanocytic nevi) into cutaneous melanoma: a population-based estimate
.
Arch Dermatol
.
2003
;
139
(
3
):
282
288
.
6
Shain
AH
,
Yeh
I
,
Kovalyshyn
I
, et al.
The genetic evolution of melanoma from precursor lesions
.
N Engl J Med
.
2015
;
373
(
20
):
1926
1936
.
7
Bastian
BC
.
The molecular pathology of melanoma: an integrated taxonomy of melanocytic neoplasia
.
Annu Rev Pathol
.
2014
;
9
:
239
271
.
8
McGovern
VJ
.
The classification of melanoma
.
Minn Med
.
1971
;
54
:
426
428
.
9
Clark
WHJ
.
A classification of malignant melanoma in man correlated with histogenesis and biologic behavior
.
In
:
Montagna
W
,
Hu
F
,
eds
.
Advances in the Biology of the Skin Volume VIII
.
New York
:
Pergamon Press;
1967
:
621
647
.
10
Gimotty
PA
,
Van
BP
,
Elder
DE
, et al.
Biologic and prognostic significance of dermal Ki67 expression, mitoses, and tumorigenicity in thin invasive cutaneous melanoma
.
J Clin Oncol
.
2005
;
23
(
31
):
8048
8056
.
11
Elder
DE
,
Guerry
D IV
,
Epstein
MN
, et al.
Invasive malignant melanomas lacking competence for metastasis
.
Am J Dermatopathol
.
1984
;
6
(
suppl
):
55
61
.
12
Guerry
D IV
,
Synnestvedt
M
,
Elder
DE
,
Schultz
D.
Lessons from tumor progression: the invasive radial growth phase of melanoma is common, incapable of metastasis, and indolent
.
J Invest Dermatol
.
1993
;
100
:
342S
345S
.
13
Gershenwald
JE
,
Scolyer
RA
,
Hess
KR
, et al.
Melanoma staging: evidence-based changes in the American Joint Committee on Cancer eighth edition cancer staging manual
.
CA Cancer J Clin
.
2017
;
67
(
6
):
472
492
.
14
Clark
WHJ
,
Elder
DE
,
Guerry
DI
, et al.
Model predicting survival in stage I melanoma based on tumor progression
.
J Natl Cancer Inst
.
1989
;
81
:
1893
1904
.
15
Clark
WHJ
,
From
L
,
Bernardino
EA
,
Mihm
MCJ
.
The histogenesis and biologic behavior of primary human malignant melanomas of the skin
.
Cancer Res
.
1969
;
29
:
705
727
.
16
McGovern
VJ
.
The classification of melanoma and its relationship with prognosis
.
Pathology
.
1970
;
2
:
85
98
.
17
Armstrong
BK
,
Cust
AE
.
Sun exposure and skin cancer, and the puzzle of cutaneous melanoma: a perspective on Fears et al. Mathematical models of age and ultraviolet effects on the incidence of skin cancer among whites in the United States. American Journal of Epidemiology. 1977; 105: 420–427
.
Cancer Epidemiol.
2017
;
48
:
147
-
156
.
18
Arnold
M
,
de Vries
E
,
Whiteman
DC
, et al.
Global burden of cutaneous melanoma attributable to ultraviolet radiation in 2012
.
Int J Cancer
.
2018
;
143
(
6
):
1305
1314
.
19
Friedman
RJ
,
Rigel
DS
,
Kopf
AW
.
Early detection of malignant melanoma: the role of physician examination and self-examination of the skin
.
CA A Cancer J Clin
.
1985
;
35
:
130
151
.
20
Rigel
DS
,
Friedman
RJ
,
Kopf
AW
,
Polsky
D.
ABCDE—an evolving concept in the early detection of melanoma
.
Arch Dermatol
.
2005
;
141
(
8
):
1032
1034
.
21
Lin
MJ
,
Mar
V
,
McLean
C
,
Wolfe
R
,
Kelly
JW
.
Diagnostic accuracy of malignant melanoma according to subtype
.
Australas J Dermatol
.
2014
;
55
(
1
):
35
42
.
22
Grob
JJ
,
Bonerandi
JJ
.
The ‘ugly duckling' sign: Identification of the common characteristics at nevi in an individual as a basis for melanoma screening
.
Arch Dermatol
.
1998
;
134
:
103
104
.
23
Whiteman
DC
,
Watt
P
,
Purdie
DM
,
Hughes
MC
,
Hayward
NK
,
Green
AC
.
Melanocytic nevi, solar keratoses, and divergent pathways to cutaneous melanoma
.
J Natl Cancer Inst
.
2003
;
95
(
11
):
806
812
.
24
Maldonado
JL
,
Fridlyand
J
,
Patel
H
, et al.
Determinants of BRAF mutations in primary melanomas
.
J Natl Cancer Inst
.
2003
;
95
(
24
):
1878
1890
.
25
Curtin
JA
,
Fridlyand
J
,
Kageshita
T
, et al.
Distinct sets of genetic alterations in melanoma
.
N Engl J Med
.
2005
;
353
(
20
):
2135
2147
.
26
Stephens
P
,
Wiesner
T
,
He
J
, et al.
Next-generation sequencing of genomic and cDNA to identify a high frequency of kinase fusions involving ROS1, ALK, RET, NTRK1, and BRAF in Spitz tumors
.
J Clin Oncol
.
2013
;
31
(
15
):
9002
.
27
Yeh
I
,
Botton
T
,
Talevich
E
, et al.
Activating MET kinase rearrangements in melanoma and Spitz tumours
.
Nat Commun
.
2015
;
6
:
7174
.
28
Yeh
I
,
Tee
MK
,
Botton
T
, et al.
NTRK3 kinase fusions in Spitz tumours
.
J Pathol
.
2016
;
240
(
3
):
282
290
.
29
Bahrami
A
,
Lee
S
,
Wu
G
, et al.
Pigment-synthesizing melanocytic neoplasm with protein kinase C alpha (PRKCA) fusion
.
JAMA Dermatol
.
2016
;
152
(
3
):
318
322
.
30
Newman
S
,
Fan
L
,
Pribnow
A
, et al.
Clinical genome sequencing uncovers potentially targetable truncations and fusions of MAP3K8 in spitzoid and other melanomas
.
Nature Med
.
2019
;
25
(
4
):
597
602
.
31
Turner
J
,
Couts
K
,
Sheren
J
, et al.
Kinase gene fusions in defined subsets of melanoma
.
Pigment Cell Melanoma Res
.
2017
;
30
(
1
):
53
62
.
32
Elder
DE
,
Xu
X.
The approach to the patient with a difficult melanocytic lesion
.
Pathology
.
2004
;
36
(
5
):
428
434
.
33
Heenan
PJ
,
Holman
CD
.
Nodular malignant melanoma: a distinct entity or a common end stage?
Am J Dermatopathol
.
1982
;
4
:
477
478
.
34
Shain
AH
,
Bastian
BC
.
From melanocytes to melanomas
.
Nat Rev Cancer
.
2016
;
16
(
6
):
345
358
.
35
Shen
S
,
Wolfe
R
,
McLean
CA
,
Haskett
M
,
Kelly
JW
.
Characteristics and associations of high-mitotic-rate melanoma
.
JAMA Dermatol
.
2014
;
150
(
10
):
1048
1055
.
36
Chamberlain
AJ
,
Fritschi
L
,
Kelly
JW
.
Nodular melanoma: patients' perceptions of presenting features and implications for earlier detection
.
J Am Acad Dermatol
.
2003
;
48
(
5
):
694
701
.
37
Bastian
BC
,
de la Fouchardiere
A
,
Elder
DE
, et al.
Genomic landscape of melanoma
.
In
:
Elder
DE
,
Massi
D
,
Scolyer
RA
,
Willemze
R
,
eds
.
WHO Classification of Skin Tumours. 4th ed
.
Lyon, France
:
IARC;
2018
:
72
75
.
World Health Organization Classification of Tumours; vol 11
.
38
Viros
A
,
Fridlyand
J
,
Bauer
J
, et al.
Improving melanoma classification by integrating genetic and morphologic features
.
Plos Med
.
2008
;
5
(
6
):
e120
.
39
Broekaert
SM
,
Roy
R
,
Okamoto
I
, et al.
Genetic and morphologic features for melanoma classification
.
Pigment Cell Melanoma Res
.
2010
;
23
(
6
):
763
770
.
40
Hodis
E
,
Watson
IR
,
Kryukov
GV
, et al.
A landscape of driver mutations in melanoma
.
Cell
.
2012
;
150
(
2
):
251
263
.
41
Krauthammer
M
,
Kong
Y
,
Bacchiocchi
A
, et al.
Exome sequencing identifies recurrent mutations in NF1 and RASopathy genes in sun-exposed melanomas
.
Nat Genet
.
2015
;
47
(
9
):
996
1002
.
42
Emmett
EA
.
Ultraviolet radiation as a cause of skin tumors
.
CRC Crit Rev Toxicol
.
1973
;
2
(
2
):
211
255
.
43
Lancaster
HO
,
Nelson
J.
Sunlight as a cause of melanoma; a clinical survey
.
Med J Aust
.
1957
;
6
(
44
):
452
456
.
44
Holman
CD
,
Mulroney
CD
,
Armstrong
BK
.
Epidemiology of pre-invasive and invasive malignant melanoma in Western Australia
.
Int J Cancer
.
1980
;
25
:
317
323
.
45
Elder
DE
.
Skin cancer: melanoma and other specific nonmelanoma skin cancers
.
Cancer
.
1995
;
75
(
suppl
):
245
256
.
46
Burbidge
TE
,
Bastian
BC
,
Guo
D
, et al.
Association of indoor tanning exposure with age at melanoma diagnosis and BRAF V600E mutations
.
J Natl Cancer Inst
.
2019
;
111
(
11
):
1228
1231
.
47
Gandini
S
,
Sera
F
,
Cattaruzza
MS
, et al.
Meta-analysis of risk factors for cutaneous melanoma: I, common and atypical naevi
.
Eur J Cancer
.
2005
;
41
(
1
):
28
44
.
48
Tucker
MA
,
Halpern
A
,
Holly
EA
, et al.
Clinically recognized dysplastic nevi: a central risk factor for cutaneous melanoma
.
JAMA
.
1997
;
277
:
1439
1444
.
49
Arumi-Uria
M
,
McNutt
NS
,
Finnerty
B.
Grading of atypia in nevi: correlation with melanoma risk
.
Mod Pathol
.
2003
;
16
(
8
):
764
771
.
50
Shors
AR
,
Kim
S
,
White
E
, et al.
Dysplastic naevi with moderate to severe histological dysplasia: a risk factor for melanoma
.
Br J Dermatol
.
2006
;
155
(
5
):
988
993
.
51
Xiong
MY
,
Rabkin
MS
,
Piepkorn
MW
, et al.
Diameter of dysplastic nevi is a more robust biomarker of increased melanoma risk than degree of histologic dysplasia: a case-control study
.
J Am Acad Dermatol
.
2014
;
71
(
6
):
1257
1258
.
52
Law
MH
,
Macgregor
S
,
Hayward
NK
.
Melanoma genetics: recent findings take us beyond well-traveled pathways
.
J Invest Dermatol
.
2012
;
132
(
7
):
1763
1774
.
53
Rigel
DS
,
Friedman
RJ
.
The rationale of the ABCDs of early melanoma
.
J Am Acad Dermatol
.
1993
;
29
:
1060
1061
.
54
Landi
MT
,
Bauer
J
,
Pfeiffer
RM
, et al.
MC1R germline variants confer risk for BRAF-mutant melanoma
.
Science
.
2006
;
313
(
5786
):
521
522
.
55
Lee
EY
,
Williamson
R
,
Watt
P
,
Hughes
MC
,
Green
AC
,
Whiteman
DC
.
Sun exposure and host phenotype as predictors of cutaneous melanoma associated with neval remnants or dermal elastosis
.
Int J Cancer
.
2006
;
119
(
3
):
636
642
.
56
Yeh
I
,
Lang
UE
,
Durieux
E
, et al.
Combined activation of MAP kinase pathway and beta-catenin signaling cause deep penetrating nevi
.
Nat Commun
.
2017
;
8
(
1
):
644
.
57
Wiesner
T
,
Murali
R
,
Fried
I
, et al.
A distinct subset of atypical spitz tumors is characterized by BRAF mutation and loss of BAP1 expression
.
Am J Surg Pathol
.
2012
;
36
(
6
):
818
830
.
58
Cohen
JN
,
Joseph
NM
,
North
JP
,
Onodera
C
,
Zembowicz
A
,
LeBoit
PE
.
Genomic analysis of pigmented epithelioid melanocytomas reveals recurrent alterations in PRKAR1A, and PRKCA genes
.
Am J Surg Pathol
.
2017
;
41
(
10
):
1333
1346
.
59
Zembowicz
A
,
Carney
JA
,
Mihm
MC
.
Pigmented epithelioid melanocytoma: a low-grade melanocytic tumor with metastatic potential indistinguishable from animal-type melanoma and epithelioid blue nevus
.
Am J Surg Pathol
.
2004
;
28
(
1
):
31
40
.
60
Parekh
V
,
Sobanko
J
,
Miller
CJ
, et al.
NRAS Q61R and BRAF G466A mutations in atypical melanocytic lesions newly arising in advanced melanoma patients treated with vemurafenib
.
J Cutan Pathol
.
2019
;
46
(
3
):
190
194
.
61
Richtig
G
,
Hoeller
C
,
Kashofer
K
, et al.
Beyond the BRAF(V)(600E) hotspot: biology and clinical implications of rare BRAF gene mutations in melanoma patients
.
Br J Dermatol
.
2017
;
177
(
4
):
936
944
.
62
Dankner
M
,
Rose
AAN
,
Rajkumar
S
,
Siegel
PM
,
Watson
IR
.
Classifying BRAF alterations in cancer: new rational therapeutic strategies for actionable mutations
.
Oncogene
.
2018
;
37
(
24
):
3183
3199
.
63
Shain
AH
,
Joseph
NM
,
Yu
R
, et al.
Genomic and transcriptomic analysis reveals incremental disruption of key signaling pathways during melanoma evolution
.
Cancer Cell
.
2018
;
34
(
1
):
45
55.e44
.
64
Zeng
H
,
Jorapur
A
,
Shain
AH
, et al.
Bi-allelic loss of CDKN2A initiates melanoma invasion via BRN2 activation
.
Cancer Cell
.
2018
;
34
(
1
):
56
68.e59
.
65
Mackenzie Ross
AD
,
Haydu
LE
,
Quinn
MJ
, et al.
The association between excision margins and local recurrence in 11,290 thin (T1) primary cutaneous melanomas: a case-control study
.
Ann Surg Oncol
.
2016
;
23
(
4
):
1082
1089
.
66
Etzkorn
JR
,
Jew
OS
,
Shin
TM
,
Sobanko
JF
,
Neal
DE
,
Miller
CJ
.
Mohs micrographic surgery with melanoma antigen recognized by T cells 1 (MART-1) immunostaining for atypical intraepidermal melanocytic proliferation
.
J Am Acad Dermatol
.
2018
;
79
(
6
):
1109
1116.e1101
.
67
Clark
WHJ
,
Elder
DE
,
Van Horn
M.
The biologic forms of malignant melanoma
.
Hum Pathol
.
1986
;
5
:
443
450
.
68
Farrahi
F
,
Egbert
BM
,
Swetter
SM
.
Histologic similarities between lentigo maligna and dysplastic nevus: importance of clinicopathologic distinction
.
J Cutan Pathol
.
2005
;
32
(
6
):
405
412
.
69
Shitara
D
,
Nascimento
MM
,
Puig
S
, et al.
Nevus-associated melanomas: clinicopathologic features
.
Am J Clin Pathol
.
2014
;
142
(
4
):
485
491
.
70
Kvaskoff
M
,
Pandeya
N
,
Green
AC
, et al.
Solar elastosis and cutaneous melanoma: a site-specific analysis
.
Int J Cancer
.
2015
;
136
(
12
):
2900
2911
.
71
Elder
DE
,
Massi
D
,
Scolyer
RA
,
Willemze
R.
WHO Classification of Skin Tumours. 4th ed
.
Lyon, France
:
IARC;
2018
.
World Health Organization Classification of Tumours; vol 11
.
72
Price
NM
,
Rywlin
AM
,
Ackerman
AB
.
Histologic criteria for the diagnosis of superficial spreading melanoma: formulated on the basis of proven metastastic lesions
.
Cancer
.
1976
;
38
:
2434
2441
.
73
King
R
,
Page
RN
,
Googe
PB
,
Mihm
MC
.
Lentiginous melanoma: a histologic pattern of melanoma to be distinguished from lentiginous nevus
.
Mod Pathol
.
2005
;
18
(
10
):
1397
1401
.
74
Kossard
S
,
Wilkinson
B.
Small cell (naevoid) melanoma: a clinicopathologic study of 131 cases
.
Australas J Dermatol
.
1997
;
38
(
suppl 1
):
S54
S58
.
75
Zalaudek
I
,
Cota
C
,
Ferrara
G
, et al.
Flat pigmented macules on sun-damaged skin of the head/neck: junctional nevus, atypical lentiginous nevus, or melanoma in situ?
Clin Dermatol
.
2014
;
32
(
1
):
88
93
.
76
Dalton
SR
,
Gardner
TL
,
Libow
LF
,
Elston
DM
.
Contiguous lesions in lentigo maligna
.
J Am Acad Dermatol
.
2005
;
52
(
5
):
859
862
.
77
Hodi
FS
,
Corless
CL
,
Giobbie-Hurder
A
, et al.
Imatinib for melanomas harboring mutationally activated or amplified KIT arising on mucosal, acral, and chronically sun-damaged skin
.
J Clin Oncol
.
2013
;
31
(
26
):
3182
3190
.
78
Andersen
LB
,
Fountain
JW
,
Gutmann
DH
, et al.
Mutations in the neurofibromatosis 1 gene in sporadic malignant melanoma cell lines
.
Nature Genet
.
1993
;
3
:
118
121
.
79
Mar
VJ
,
Wong
SQ
,
Li
J
, et al.
BRAF/NRAS wild-type melanomas have a high mutation load correlating with histological and molecular signatures of UV damage
.
Clin Cancer Res
.
2013
;
19
(
7
):
4589
4598
.
80
Goodman
AM
,
Kato
S
,
Bazhenova
L
, et al.
Tumor mutational burden as an independent predictor of response to immunotherapy in diverse cancers
.
Mol Cancer Ther
.
2017
;
16
(
11
):
2598
2608
.
81
Lens
MB
,
Newton-Bishop
JA
,
Boon
AP
.
Desmoplastic malignant melanoma: a systematic review
.
Br J Dermatol
.
2005
;
152
(
4
):
673
678
.
82
Chen
LL
,
Jaimes
N
,
Barker
CA
,
Busam
KJ
,
Marghoob
AA
.
Desmoplastic melanoma: a review
.
J Am Acad Dermatol
.
2013
;
68
(
5
):
825
833
.
83
Reed
RJ
,
Leonard
DD
.
Neurotropic melanoma: a variant of desmoplastic melanoma
.
Am J Surg Pathol
.
1979
;
3
:
301
311
.
84
Jain
S
,
Allen
PW
.
Desmoplastic malignant melanoma and its variants: a study of 45 cases
.
Am J Surg Pathol
.
1989
;
13
:
358
373
.
85
Hawkins
WG
,
Busam
KJ
,
Ben-Porat
L
, et al.
Desmoplastic melanoma: a pathologically and clinically distinct form of cutaneous melanoma
.
Ann Surg Oncol
.
2005
;
12
(
3
):
207
213
.
86
Eroglu
Z
,
Zaretsky
JM
,
Hu-Lieskovan
S
, et al.
High response rate to PD-1 blockade in desmoplastic melanomas
.
Nature
.
2018
;
553
(
7688
):
347
350
.
87
Plaza
JA
,
Bonneau
P
,
Prieto
V
, et al.
Desmoplastic melanoma: an updated immunohistochemical analysis of 40 cases with a proposal for an additional panel of stains for diagnosis
.
J Cutan Pathol
.
2016
;
43
(
4
):
313
323
.
88
Sidiropoulos
M
,
Sholl
LM
,
Obregon
R
,
Guitart
J
,
Gerami
P.
Desmoplastic nevus of chronically sun-damaged skin: an entity to be distinguished from desmoplastic melanoma
.
Am J Dermatopathol
.
2014
;
36
(
8
):
629
634
.
89
Barr
RJ
,
Morales
RV
,
Graham
JH
.
Desmoplastic nevus: a distinct histologic variant of mixed spindle cell and epithelioid cell nevus
.
Cancer
.
1980
;
46
:
557
564
.
90
Ferreira
I
,
Kind
P
,
Van Den Berghe
I
, et al.
Melanocytic naevi with perineurial differentiation: a distinctive variant of neurotised naevi and a diagnostic pitfall with desmoplastic melanoma
.
Histopathology
.
2018
;
72
(
4
):
679
684
.
91
Hollmig
ST
,
Sachdev
R
,
Cockerell
CJ
,
Posten
W
,
Chiang
M
,
Kim
J.
Spindle cell neoplasms encountered in dermatologic surgery: a review
.
Dermatol Surg
.
2012
;
38
(
6
):
825
850
.
92
Machado
I
,
Llombart
B
,
Cruz
J
, et al.
Desmoplastic melanoma may mimic a cutaneous peripheral nerve sheath tumor: report of 3 challenging cases
.
J Cutan Pathol
.
2017
;
44
(
7
):
632
638
.
93
Ramos-Herberth
FI
,
Karamchandani
J
,
Kim
J
,
Dadras
SS
.
SOX10 immunostaining distinguishes desmoplastic melanoma from excision scar
.
J Cutan Pathol
.
2010
;
37
(
9
):
944
952
.
94
Wiesner
T
,
Kiuru
M
,
Scott
SN
, et al.
NF1 mutations are common in desmoplastic melanoma
.
Am J Surg Pathol
.
2015
;
39
(
10
):
1357
1362
.
95
Shain
AH
,
Garrido
M
,
Botton
T
, et al.
Exome sequencing of desmoplastic melanoma identifies recurrent NFKBIE promoter mutations and diverse activating mutations in the MAPK pathway
.
Nat Genet
.
2015
;
47
(
10
):
1194
1199
.
96
Lazova
R
,
Pornputtapong
N
,
Halaban
R
, et al.
Spitz nevi and Spitzoid melanomas: exome sequencing and comparison with conventional melanocytic nevi and melanomas
.
Mod Pathol
.
2017
;
30
(
5
):
640
649
.
97
Paniago-Pereira
C
,
Maize
JC
,
Ackerman
AB
.
Nevus of large spindle and/or epithelioid cells (Spitz's nevus)
.
Arch Dermatol
.
1978
;
114
(
12
):
1811
1823
.
98
Cerrato
F
,
Wallins
JS
,
Webb
ML
,
McCarty
ER
,
Schmidt
BA
,
Labow
BI
.
Outcomes in pediatric atypical Spitz tumors treated without sentinel lymph node biopsy
.
Pediatr Dermatol
.
2011
;
29
(
4
):
448
453
.
99
Kamino
H
,
Flotte
TJ
,
Misheloff
E
,
Alba Greco M, Ackerman AB. Eosinophilic globules in Spitz's nevi: new findings and a diagnostic sign
.
Am J Dermatopathol
.
1979
;
1
:
319
324
.
100
Spatz
A
,
Calonje
E
,
Handfield-Jones
S
,
Barnhill
RL
.
Spitz tumors in children: a grading system for risk stratification
.
Arch Dermatol
.
1999
;
135
(
3
):
282
285
.
101
Hantschke
M
,
Bastian
BC
,
LeBoit
PE
.
Consumption of the epidermis: a diagnostic criterion for the differential diagnosis of melanoma and Spitz nevus
.
Am J Surg Pathol
.
2004
;
28
(
12
):
1621
1625
.
102
Requena
C
,
Botella
R
,
Nagore
E
, et al.
Characteristics of spitzoid melanoma and clues for differential diagnosis with spitz nevus
.
Am J Dermatopathol
.
2012
;
34
(
5
):
478
486
.
103
Crotty
KA
,
Scolyer
RA
,
Li
L
,
Palmer
AA
,
Wang
L
,
McCarthy
SW
.
Spitz naevus versus Spitzoid melanoma: when and how can they be distinguished?
Pathology
.
2002
;
34
(
1
):
6
12
.
104
Cerroni
L
,
Barnhill
R
,
Elder
D
, et al.
Melanocytic tumors of uncertain malignant potential: results of a tutorial held at the XXIX Symposium of the International Society of Dermatopathology in Graz, October 2008
.
Am J Surg Pathol
.
2010
;
34
(
3
):
314
326
.
105
Gerami
P
,
Busam
K
,
Cochran
A
, et al.
Histomorphologic assessment and interobserver diagnostic reproducibility of atypical spitzoid melanocytic neoplasms with long-term follow-up
.
Am J Surg Pathol
.
2014
;
38
(
7
):
934
940
.
106
Li
LX
,
Crotty
KA
,
McCarthy
SW
,
Palmer
AA
,
Kril
JJ
.
A zonal comparison of MIB1-Ki67 immunoreactivity in benign and malignant melanocytic lesions
.
Am J Dermatopathol
.
2000
;
22
(
6
):
489
495
.
107
Ritter
A
,
Tronnier
M
,
Vaske
B
,
Mitteldorf
C.
Reevaluation of established and new criteria in differential diagnosis of Spitz nevus and melanoma
.
Arch Dermatol Res
.
2018
;
310
(
4
):
329
342
.
108
Taylor
LA
,
O'Day
C
,
Dentchev
T
, et al.
p15 expression differentiates nevus from melanoma
.
Am J Pathol
.
2016
;
186
(
12
):
3094
3099
.
109
Lee
S
,
Barnhill
RL
,
Dummer
R
, et al.
TERT promoter mutations are predictive of aggressive clinical behavior in patients with spitzoid melanocytic neoplasms
.
Sci Rep
.
2015
;
5
:
11200
.
110
Gerami
P
,
Li
G
,
Pouryazdanparast
P
, et al.
A highly specific and discriminatory FISH assay for distinguishing between benign and malignant melanocytic neoplasms
.
Am J Surg Pathol
.
2012
;
36
(
6
):
808
817
.
111
Lazova
R
,
Seeley
EH
,
Keenan
M
,
Gueorguieva
R
,
Caprioli
RM
.
Imaging mass spectrometry—a new and promising method to differentiate Spitz nevi from Spitzoid malignant melanomas
.
Am J Dermatopathol
.
2012
;
34
(
1
):
82
90
.
112
Wiesner
T
,
Kutzner
H
,
Cerroni
L
,
Mihm
MC
Jr
,
Busam
KJ
,
Murali
R.
Genomic aberrations in spitzoid melanocytic tumours and their implications for diagnosis, prognosis and therapy
.
Pathology
.
2016
;
48
(
2
):
113
131
.
113
Star
P
,
Goodwin
A
,
Kapoor
R
, et al.
Germline BAP1-positive patients: the dilemmas of cancer surveillance and a proposed interdisciplinary consensus monitoring strategy
.
Eur J Cancer
.
2018
;
92
:
48
53
.
114
Wiesner
T
,
He
J
,
Yelensky
R
, et al.
Kinase fusions are frequent in Spitz tumours and spitzoid melanomas
.
Nat Commun
.
2014
;
5
:
3116
.
115
Bastian
BC
,
LeBoit
PE
,
Pinkel
D.
Mutations and copy number increase of HRAS in Spitz nevi with distinctive histopathological features
.
Am J Pathol
.
2000
;
157
(
3
):
967
972
.
116
Gerami
P
,
Scolyer
RA
,
Xu
X
, et al.
Risk assessment for atypical spitzoid melanocytic neoplasms using FISH to identify chromosomal copy number aberrations
.
Am J Surg Pathol
.
2013
;
37
(
5
):
676
684
.
117
Gerami
P
,
Cooper
C
,
Bajaj
S
, et al.
Outcomes of atypical Spitz tumors with chromosomal copy number aberrations and conventional melanomas in children
.
Am J Surg Pathol
.
2013
;
37
(
9
):
1387
1394
.
118
Requena
C
,
Heidenreich
B
,
Kumar
R
,
Nagore
E.
TERT promoter mutations are not always associated with poor prognosis in atypical spitzoid tumors
.
Pigment Cell Melanoma Res
.
2017
;
30
(
2
):
265
268
.
119
Harms
PW
,
Hocker
TL
,
Zhao
L
, et al.
Loss of p16 expression and copy number changes of CDKN2A in a spectrum of spitzoid melanocytic lesions
.
Hum Pathol
.
2016
;
58
:
152
160
.
120
Wang
Y
,
Zhao
Y
,
Ma
S.
Racial differences in six major subtypes of melanoma: descriptive epidemiology
.
BMC Cancer
.
2016
;
16
:
691
.
121
Jung
HJ
,
Kweon
SS
,
Lee
JB
,
Lee
SC
,
Yun
SJ
.
A clinicopathologic analysis of 177 acral melanomas in Koreans: relevance of spreading pattern and physical stress
.
JAMA Dermatol
.
2013
;
149
(
11
):
1281
1288
.
122
Costello
CM
,
Pittelkow
MR
,
Mangold
AR
.
Acral melanoma and mechanical stress on the plantar surface of the foot
.
New Engl J Med
.
2017
;
377
(
4
):
395
396
.
123
Carrera
C
,
Gual
A
,
Diaz
A
, et al.
Prognostic role of the histological subtype of melanoma on the hands and feet in Caucasians
.
Melanoma Res
.
2017
;
27
(
4
):
315
320
.
124
Arrington
JH
III
,
Reed
RJ
,
Ichinose
H
,
Krementz
ET
.
Plantar lentiginous melanoma: a distinctive variant of human cutaneous malignant melanoma
.
Am J Surg Pathol
.
1977
;
1
:
131
143
.
125
Coleman
WP
III
,
Loria
PR
,
Reed
RJ
,
Krementz
ET
.
Acral lentiginous melanoma
.
Arch Dermatol
.
1980
;
116
(
7
):
773
776
.
126
Moon
KR
,
Choi
YD
,
Kim
JM
, et al.
Genetic alterations in primary acral melanoma and acral melanocytic nevus in Korea: common mutated genes show distinct cytomorphological features
.
J Invest Dermatol
.
2018
;
138
(
4
):
933
945
.
127
Yeh
I
,
Jorgenson
E
,
Shen
L
, et al.
Targeted genomic profiling of acral melanoma
.
J Natl Cancer Inst
.
2019
;
111
(
10
):
1068
1077
.
128
Bastian
BC
,
Kashani-Sabet
M
,
Hamm
H
, et al.
Gene amplifications characterize acral melanoma and permit the detection of occult tumor cells in the surrounding skin
.
Cancer Res
.
2000
;
60
:
1968
1973
.
129
Scanlon
P
,
Tian
J
,
Zhong
J
, et al.
Enhanced immunohistochemical detection of neural infiltration in primary melanoma: is there a clinical value?
Hum Pathol
.
2014
;
45
(
8
):
1656
1663
.
130
Nakamura
Y
,
Fujisawa
Y
,
Teramoto
Y
, et al.
Tumor-to-bone distance of invasive subungual melanoma: an analysis of 30 cases
.
J Dermatol
.
2014
;
41
(
10
):
872
877
.
131
Palicka
GA
,
Rhodes
AR
.
Acral melanocytic nevi: prevalence and distribution of gross morphologic features in white and black adults
.
Arch Dermatol
.
2010
;
146
(
10
):
1085
1094
.
132
Clemente
C
,
Zurrida
S
,
Bartoli
C
,
Bono
A
,
Collini
P
,
Rilke
F.
Acral-lentiginous naevus of plantar skin
.
Histopathology
.
1995
;
27
:
549
555
.
133
Saida
T
,
Koga
H
,
Uhara
H.
Key points in dermoscopic differentiation between early acral melanoma and acral nevus
.
J Dermatol
.
2011
;
38
(
1
):
25
34
.
134
Saida
T
,
Koga
H
,
Goto
Y
,
Uhara
H.
Characteristic distribution of melanin columns in the cornified layer of acquired acral nevus: an important clue for histopathologic differentiation from early acral melanoma
.
Am J Dermatopathol
.
2011
;
33
(
5
):
468
473
.
135
Su
J
,
Yu
W
,
Liu
J
, et al.
Fluorescence in situ hybridisation as an ancillary tool in the diagnosis of acral melanoma: a review of 44 cases
.
Pathology
.
2017
;
49
(
7
):
740
749
.
136
Curtin
JA
,
Busam
K
,
Pinkel
D
,
Bastian
BC
.
Somatic activation of KIT in distinct subtypes of melanoma
.
J Clin Oncol
.
2006
;
24
(
26
):
4340
4346
.
137
Furney
SJ
,
Turajlic
S
,
Stamp
G
, et al.
The mutational burden of acral melanoma revealed by whole-genome sequencing and comparative analysis
.
Pigment Cell Melanoma Res
.
2014
;
27
(
5
):
835
838
.
138
Liang
WS
,
Hendricks
W
,
Kiefer
J
, et al.
Integrated genomic analyses reveal frequent TERT aberrations in acral melanoma
.
Genome Res
.
2017
;
27
(
4
):
524
532
.
139
Niu
HT
,
Zhou
QM
,
Wang
F
, et al.
Identification of anaplastic lymphoma kinase break points and oncogenic mutation profiles in acral/mucosal melanomas
.
Pigment Cell Melanoma Res
.
2013
;
26
(
5
):
646
653
.
140
Merkel
EA
,
Gerami
P.
Malignant melanoma of sun-protected sites: a review of clinical, histological, and molecular features
.
Lab Invest
.
2017
;
97
(
6
):
630
635
.
141
Spencer
KR
,
Mehnert
JM
.
Mucosal melanoma: epidemiology, biology and treatment
.
Cancer Treat Res
.
2016
;
167
:
295
320
.
142
Saida
T
,
Kawachi
S
,
Takata
M
, et al.
Histopathological characteristics of malignant melanoma affecting mucous membranes: a unifying concept of histogenesis
.
Pathology
.
2004
;
36
(
5
):
404
413
.
143
Hou
JY
,
Baptiste
C
,
Hombalegowda
RB
, et al.
Vulvar and vaginal melanoma: a unique subclass of mucosal melanoma based on a comprehensive molecular analysis of 51 cases compared with 2253 cases of nongynecologic melanoma
.
Cancer
.
2017
;
123
(
8
):
1333
1344
.
144
McGovern
VJ
,
Cochran
AJ
,
Van der
EEP
,
Little
JH
,
MacLennan
R
.
The classification of malignant melanoma, its histological reporting and registration: a revision of the 1972 Sydney classification
.
Pathology
.
1986
;
18
:
12
21
.
145
Scholz
SL
,
Cosgarea
I
,
Susskind
D
, et al.
NF1 mutations in conjunctival melanoma
.
Br J Cancer
.
2018
;
118
(
9
):
1243
1247
.
146
Sexton
FM
,
Maize
JC
.
Melanotic macules and melanoacanthomas of the lip: a comparative study with census of the basal melanocyte population
.
Am J Dermatopathol
.
1987
;
9
:
438
444
.
147
Massi
D
,
Nardini
P
,
De
G
,
V, Carli P. Simultaneous occurrence of multiple melanoma in situ on sun-damaged skin (lentigo maligna), solar lentigo and labial melanosis: the value of dermoscopy in diagnosis
.
J Eur Acad Dermatol Venereol
.
1999
;
13
(
3
):
193
197
.
148
Prieto
VG
,
Dehner
LP
,
Pfeiffer
JD
,
Wick
MR
.
Genital and mucosal melanocytic tumors
.
In
:
Elder
DE
,
Massi
D
,
Scolyer
RA
,
Willemze
R
,
eds
.
WHO Classification of Skin Tumours
.
Lyon, France
:
IARC;
2018
:
121
122
.
World Health Organization Classification of Tumours; vol 11
.
149
Furney
SJ
,
Turajlic
S
,
Stamp
G
, et al.
Genome sequencing of mucosal melanomas reveals that they are driven by distinct mechanisms from cutaneous melanoma
.
J Pathol
.
2013
;
230
(
3
):
261
269
.
150
Gong
HZ
,
Zheng
HY
,
Li
J.
The clinical significance of KIT mutations in melanoma: a meta-analysis
.
Melanoma Res
.
2018
;
28
(
4
):
259
270
.
151
Kim
HS
,
Jung
M
,
Kang
HN
, et al.
Oncogenic BRAF fusions in mucosal melanomas activate the MAPK pathway and are sensitive to MEK/PI3K inhibition or MEK/CDK4/6 inhibition
.
Oncogene
.
2017
;
36
(
23
):
3334
3345
.
152
Swaminathan
SS
,
Field
MG
,
Sant
D
, et al.
Molecular characteristics of conjunctival melanoma using whole-exome sequencing
.
JAMA Ophthalmol
.
2017
;
135
(
12
):
1434
1437
.
153
Rivolta
C
,
Royer-Bertrand
B
,
Rimoldi
D
, et al.
UV light signature in conjunctival melanoma; not only skin should be protected from solar radiation
.
J Hum Genet
.
2016
;
61
(
4
):
361
362
.
154
Alper
J
,
Holmes
LB
,
Mihm
MCJ
.
Birthmarks with serious medical significance: nevocellular nevi, sebaceous nevi, and multiple cafe au lait spots
.
J Pediatr
.
1979
;
95
:
696
700
.
155
Krengel
S
,
Hauschild
A
,
Schafer
T.
Melanoma risk in congenital melanocytic naevi: a systematic review
.
Br J Dermatol
.
2006
;
155
(
1
):
1
8
.
156
Zaal
LH
,
Mooi
WJ
,
Sillevis Smitt
JH
,
van der Horst
CM
.
Classification of congenital melanocytic naevi and malignant transformation: a review of the literature
.
Br J Plast Surg
.
2004
;
57
(
8
):
707
719
.
157
Zaal
LH
,
Mooi
WJ
,
Klip
H
,
van der Horst
CM
.
Risk of malignant transformation of congenital melanocytic nevi: a retrospective nationwide study from The Netherlands
.
Plast Reconstr Surg
.
2005
;
116
(
7
):
1902
1909
.
158
Xu
X
,
Bellucci
KS
,
Elenitsas
R
,
Elder
DE
.
Cellular nodules in congenital pattern nevi
.
J Cutan Pathol
.
2004
;
31
(
2
):
153
159
.
159
Hendrickson
MR
,
Ross
JC
.
Neoplasms arising in congenital giant nevi: morphologic study of seven cases and a review of the literature
.
Am J Surg Pathol
.
1981
;
5
(
2
):
109
135
.
160
Yelamos
O
,
Arva
NC
,
Obregon
R
, et al.
A comparative study of proliferative nodules and lethal melanomas in congenital nevi from children
.
Am J Surg Pathol
.
2015
;
39
(
3
):
405
415
.
161
Bauer
J
,
Curtin
JA
,
Pinkel
D
,
Bastian
BC
.
Congenital melanocytic nevi frequently harbor NRAS mutations but no BRAF mutations
.
J Invest Dermatol
.
2007
;
127
(
1
):
179
182
.
162
Charbel
C
,
Fontaine
RH
,
Malouf
GG
, et al.
NRAS mutation is the sole recurrent somatic mutation in large congenital melanocytic nevi
.
J Invest Dermatol
.
2014
;
134
(
4
):
1067
1074
.
163
Lu
C
,
Zhang
J
,
Nagahawatte
P
, et al.
The genomic landscape of childhood and adolescent melanoma
.
J Invest Dermatol
.
2015
;
135
(
3
):
816
823
.
164
Fan
Y
,
Lee
S
,
Wu
G
, et al.
Telomerase expression by aberrant methylation of the TERT promoter in melanoma arising in giant congenital nevi
.
J Invest Dermatol
.
2016
;
136
(
1
):
339
342
.
165
Bastian
BC
,
Xiong
J
,
Frieden
IJ
, et al.
Genetic changes in neoplasms arising in congenital melanocytic nevi: differences between nodular proliferations and melanomas
.
Am J Pathol
.
2002
;
161
(
4
):
1163
1169
.
166
Zembowicz
A
,
Phadke
PA
.
Blue nevi and variants: an update
.
Arch Pathol Lab Med
.
2011
;
135
(
3
):
327
336
.
167
Harris
GR
,
Shea
CR
,
Horenstein
MG
,
Reed
JA
,
Burchette
JL
Jr
,
Prieto
VG
.
Desmoplastic (sclerotic) nevus: an underrecognized entity that resembles dermatofibroma and desmoplastic melanoma
.
Am J Surg Pathol
.
1999
;
23
(
7
):
786
794
.
168
Zembowicz
A.
Blue nevi and related tumors
.
Clin Lab Med
.
2017
;
37
(
3
):
401
415
.
169
Borgenvik
TL
,
Karlsvik
TM
,
Ray
S
,
Fawzy
M
,
James
N.
Blue nevus-like and blue nevus-associated melanoma: a comprehensive review of the literature
.
ANZ J Surg
.
2017
;
87
(
5
):
345
349
.
170
Barnhill
RL
,
Argenyi
Z
,
Berwick
M
, et al.
Atypical cellular blue nevi (cellular blue nevi with atypical features): lack of consensus for diagnosis and distinction from cellular blue nevi and malignant melanoma (“malignant blue nevus”)
.
Am J Surg Pathol
.
2008
;
32
(
1
):
36
44
.
171
Griewank
KG
,
Muller
H
,
Jackett
LA
, et al.
SF3B1 and BAP1 mutations in blue nevus-like melanoma
.
Mod Pathol
.
2017
;
30
(
7
):
928
939
.
172
Busam
KJ
,
Fang
Y
,
Jhanwar
S
,
Lacouture
M.
Diagnosis of blue nevus-like metastatic uveal melanoma confirmed by fluorescence in situ hybridization (FISH) for monosomy 3
.
J Cutan Pathol
.
2012
;
39
(
6
):
621
625
.
173
Maize
JC
Jr
,
McCalmont
TH
,
Carlson
JA
,
Busam
KJ
,
Kutzner
H
,
Bastian
BC
.
Genomic analysis of blue nevi and related dermal melanocytic proliferations
.
Am J Surg Pathol
.
2005
;
29
(
9
):
1214
1220
.
174
van Raamsdonk
CD
,
Bezrookove
V
,
Green
G
, et al.
Frequent somatic mutations of GNAQ in uveal melanoma and blue naevi
.
Nature
.
2009
;
457
(
7229
):
599
602
.
175
van Raamsdonk
CD
,
Griewank
KG
,
Crosby
MB
, et al.
Mutations in GNA11 in uveal melanoma
.
N Engl J Med
.
2010
;
363
(
23
):
2191
2199
.
176
Costa
S
,
Byrne
M
,
Pissaloux
D
, et al.
Melanomas associated with blue nevi or mimicking cellular blue nevi: clinical, pathologic, and molecular study of 11 cases displaying a high frequency of GNA11 mutations, BAP1 expression loss, and a predilection for the scalp
.
Am J Surg Pathol
.
2016
;
40
(
3
):
368
377
.
177
Chan
MP
,
Andea
AA
,
Harms
PW
, et al.
Genomic copy number analysis of a spectrum of blue nevi identifies recurrent aberrations of entire chromosomal arms in melanoma ex blue nevus
.
Mod Pathol
.
2016
;
29
(
3
):
227
239
.
178
Mar
V
,
Roberts
H
,
Wolfe
R
,
English
DR
,
Kelly
JW
.
Nodular melanoma: a distinct clinical entity and the largest contributor to melanoma deaths in Victoria, Australia
.
J Am Acad Dermatol
.
2012
;
68
(
4
):
568
575
.
179
Dessinioti
C
,
Dimou
N
,
Geller
AC
, et al.
Distinct clinicopathological and prognostic features of thin nodular primary melanomas
:
an international study from 17 centers [published online ahead of print March 3
,
2019]
. J Natl Cancer Inst. doi:.
180
Skala
SL
,
Arps
DP
,
Zhao
L
, et al.
Comprehensive histopathological comparison of epidermotropic/dermal metastatic melanoma and primary nodular melanoma
.
Histopathology
.
2018
;
72
(
3
):
472
480
.
181
Gerami
P
,
Shea
C
,
Stone
MS
.
Angiotropism in epidermotropic metastatic melanoma: another clue to the diagnosis
.
Am J Dermatopathol
.
2006
;
28
(
5
):
429
433
.
182
White
WL
,
Hitchcock
MG
.
Dying dogma: the pathological diagnosis of epidermotropic metastatic malignant melanoma
.
Semin Diagn Pathol
.
1998
;
15
:
176
188
.
183
Lestre
S
,
Joao
A
,
Ponte
P
, et al.
Intraepidermal epidermotropic metastatic melanoma: a clinical and histopathological mimicker of melanoma in situ occurring in multiplicity
.
J Cutan Pathol
.
2011
;
38
(
6
):
514
520
.
184
Ruhoy
SM
,
Prieto
VG
,
Eliason
SL
,
Grichnik
JM
,
Burchette
JL
Jr
,
Shea
CR
.
Malignant melanoma with paradoxical maturation
.
Am J Surg Pathol
.
2000
;
24
(
12
):
1600
1614
.
185
Cochran
AJ
,
Bastian
BC
,
Elder
DE
.
Nodular melanoma
.
In
:
Elder
DE
,
Massi
D
,
Scolyer
RA
,
Willemze
R
,
eds
.
WHO Classification of Skin Tumors. 4th ed
.
Lyon, France
:
IARC;
2018
:
145
146
.
World Health Organization Classification of Tumours; vol 11
.

Author notes

The authors have no relevant financial interest in the products or companies described in this article.

DEE, DM, BCB, IAC and RAS are among the authors of the World Health Organisation Classification of Skin Tumours, 4th Edition. The content of this article represents the personal views of the authors and does not represent the views of the authors' employers and associated institutions. Where authors are identified as personnel of the International Agency for Research on Cancer/World Health Organization, the authors alone are responsible for the views expressed in this article and they do not necessarily represent the decisions, policy or views of the International Agency for Research on Cancer/World Health Organization. Though not relevant to this submission, RAS has received fees for professional services from Merck Sharp & Dohme, GlaxoSmithKline Australia, Bristol-Myers Squibb, Dermpedia, Novartis Pharmaceuticals Australia Pty Ltd, Myriad, NeraCare and Amgen. RAS is also supported by an Australian National Health and Medical Research Council Practitioner Fellowship, DEE has received consulting fees from Myriad Genetics and DermPedia, and DM has received honoraria from Novartis, Bayer HealthCare Pharmaceuticals Inc., Pierre-Fabre, Sanofi Genzyme, Merck Sharp & Dohme Italia S.r.l., Roche.