You are on page 1of 72

Chapter 26 

Melanoma
26
CHAPTER

See
www.expertconsult.com
for references and
additional material
Jeffrey P. North, Boris C. Bastian and Alexander J. Lazar

MELANOMA 1310 Prognostic indicators  1320 Angiotropic and angiomatoid (pseudovascular)


Clinical features  1310 Differential diagnosis  1327 melanoma 1341
Lentigo maligna and lentigo maligna Metaplastic melanoma (melanoma with
Immunohistochemistry of melanoma  1328 heterologous differentiation)  1341
melanoma 1311
Superficial spreading melanoma  1312 Histologic variants of melanoma  1330 Pigmented epithelioid melanocytoma (pigment
Nested melanoma  1330 synthesizing melanoma; animal-type
Acral lentiginous melanoma  1312
Minimal deviation melanoma  1330 melanoma) 1342
Nodular melanoma  1313
Nevoid melanoma  1331 Epidermotropic metastatic melanoma  1343
Melanoma arising at noncutaneous (primarily
Small cell melanoma  1334 Blue nevus-like metastatic melanoma  1344
mucosal) sites  1313
Spitzoid melanoma  1334 Desmoplastic and neurotropic
Histologic features  1313 Signet ring cell melanoma  1336
Lentigo maligna and lentigo maligna melanoma  1344
Rhabdoid melanoma  1336
melanoma 1315 Balloon and clear cell melanoma  1337 Melanoma in children  1350
Superficial spreading melanoma (pagetoid Myxoid melanoma  1338 Dermal squamomelanocytic tumor  1351
melanoma) 1317 Melanoma with neuroendocrine
Acral lentiginous melanoma  1318 Basomelanocytic tumor  1351
differentiation 1338
Nodular melanoma  1319 Adenoid and pseudopapillary melanoma  1339 Lentiginous melanoma  1353
Cell types  1319 Blue nevus-like melanoma (malignant blue Primary dermal melanoma  1354
Epithelioid cells  1319 nevus) 1339 Molecular classification of melanoma  1355
Spindled cells  1319

MELANOMA
adults, especially in patients less than 10 years of age; however, metastasis
Clinical features
rates of 36–39% and overall mortality of 10–29% after 5 years or more
Over the last several decades, the conspicuous increasing incidence of this follow-up have been documented.20,27,28 Transplacental spread with resultant
malignant neoplasm has made this disease more prominent, linked to some congenital melanoma has been documented exceptionally rarely.29
rise in morbidity and mortality.1–7 While roughly half of the major cancers Melanoma may:
in the United States showed decreasing incidence trends from 2009 to 2013, • develop within a pre-existent benign (congenital or acquired)
melanoma incidence continued to significantly increase over this period melanocytic nevus,30–36
just as it had done previously in both men and women.8,9 At present it • complicate a dysplastic nevus,
accounts for approximately 4% and 6% of all new malignancies in females • arise de novo,
and males, respectively.5 This has made melanoma the fifth and sixth most • very rarely, evolve within a cellular blue nevus or other dermal
common cancers in the United States in terms of estimated new cases in dendrocytosis (see malignant blue nevus).
2017.10 However, the number of deaths from this disease places it well Precise figures are not available, but considering that the average adult has
outside the top 10 causes of cancer-related deaths for both sexes at 9730 approximately 15–40 acquired melanocytic nevi and that the incidence of
annually.10 Mortality for this disease is still increasing in males, but has melanoma in the United States is roughly 30 per 100,000 of the population
leveled off and appears to be decreasing in females, according to present per annum, then the likelihood of any one benign acquired melanocytic
data.6 Currently, the average rate of increasing incidence is of the order of nevus undergoing malignant transformation is markedly remote.2,37 On
4–6% per year.7,11–13 The estimated number of new cases of melanoma per the other hand, at least 35% of nodular and superficial spreading melano-
year is in excess of 87,000 in the United States.6,10,14 Current projections for mas arise within a background of melanocytic nevi (congenital, acquired
non-Hispanic white individuals in the United States estimate that 1 person or dysplastic).3,30,36,38–42 Only very rarely do the more typical (small) con-
in 28 males and 1 in 44 females will ultimately develop melanoma.10,15 Men genital nevi develop melanomatous features. Giant ‘bathing-trunk’ vari-
are 1.6 times more likely to suffer from melanoma, but 2.4 times more ants, however, are a greater cause for concern, with approximately 3–18%
likely to die of the disease.10 undergoing malignant transformation.22,43–45 Lentigo maligna melanoma is
Although melanoma is seen predominantly in adults, it occasionally pre- not associated with pre-existent benign or dysplastic nevi.
sents in children.10,16–23 The latter often have an associated risk factor, such While the etiology is multifactorial, including genetic and racial factors,
as xeroderma pigmentosum, congenital bathing-trunk nevus, familial dys- by far the most important known predisposing environmental agent in the
plastic nevus syndrome or familial melanoma, and possibly immunosuppres- majority of tumors (with the exception of acral lentiginous and mucosal
sion. Pediatric melanoma accounts for 1–3% of all childhood malignancies, variants) is excessive exposure to ultraviolet (UV) light.46–49 Surprisingly, the
amounting to less than 500 cases per year in the United States.24–26 Its bio- contribution of the UV is modest, with an estimated 1.7-fold increase in
logical behavior is more variable and difficult to predict than melanoma in risk.50 Although traditionally UVB has been regarded as the causative agent,

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Clinical features 1311

more recent evidence implicates UVA, at least in some patients.51–55 This • a size of 6.0 mm diameter or greater,
association is complex and recent compelling work also stresses the impor- • irregularity of the border of the lesion, usually with scalloping,
tance of moderate, non-burning sun exposure for overall human health • irregular and variable pigmentation,
including vitamin D production and other benefits—including prevention of • asymmetry,
skin cancer.56–58 Melanin pigment protects against the effects of solar irra- • recent and rapid change in a pre-existing lesion.
diation and therefore cutaneous melanoma is rare in dark-skinned races, Irritation and bleeding are also a source of concern. The development of
except for albinos and at those sites where pigment is absent (e.g., nail beds, ulceration, which often relates to a rapidly evolving tumor, is important and
palms, soles, and mucous membranes). UV irradiation, possibly by inducing is regarded as an indication of a potentially poor prognosis. Clinical evi-
free-radical formation, results in the development of dimers between adja- dence of nodule formation by definition implies that the tumor has entered
cent pyrimidine molecules. Patients who lack the necessary endonucleases the vertical growth phase. In contrast to benign melanocytic nevi, the skin
for repairing such damage, such as those with xeroderma pigmentosum, lines are usually absent over the surface of a melanoma.
have a greatly increased risk of developing cutaneous neoplasms, including Lesions on the integument have traditionally been classified into four
melanoma.59 major clinicopathological subtypes although in the more recent literature
Melanoma is particularly common in those individuals who are the validity of such subdivision has been questioned, and classification into
most susceptible to the effects of excessive sunlight (e.g., Celts with red more genetically oriented groups may offer advantages.101–103 Gene profiling
hair, blue eyes, and fair complexions who tan poorly and tend towards and the results of other high-throughput technologies will also likely aid
sunburn and excessive freckling, i.e., skin types I and II). It is notewor- in revising our traditional classification scheme so that clinically and ther-
thy that the incidence of superficial spreading melanoma is more closely apeutically relevant subsets can be identified.104,105 An evolving molecular
related to sporadic intensive exposure to sunlight, particularly isolated classification is presented in the final section of this chapter. The currently
episodes of severe sunburn in childhood, rather than to chronic lifelong accepted subdivision is particularly based on perceived differences in prog-
exposure as with lentigo maligna melanoma.47,53,60–62 It is more common nosis and etiology and the genomic features of these groups largely bear this
in those who sporadically sunbathe than in long-term outdoor workers. out. Although any variation in biological behavior relates more to tumor
The relationship between melanoma and sunlight is further highlighted by thickness than histogenetic subtype, there are certainly histologic differences
the increasing incidence towards the equator where UV radiation is most that make the distinction possible in the majority of cases. Contrariwise, in
intense.49,63 some melanomas, it is not possible to effect a precise classification and, in
Patients who already have one melanoma have an increased risk of devel- such instances, an unqualified diagnosis of melanoma is quite sufficient. For
oping a second independent primary tumor. The reported incidence varies the moment, however, subtyping melanoma is firmly entrenched in both the
from 1% to 8% of cases, with the upper end of this range being seen in the literature and pathology dogma.106 Four major subtypes of cutaneous mela-
setting of familial melanoma.64–67 It should be noted, however, that much of noma are currently recognized:29,107–113
the literature referring to multiple melanomas does not take epidermotropic • lentigo maligna melanoma (melanoma arising within a pre-existent
metastatic disease into account and therefore the true incidence is likely to lentigo maligna),
be towards the lower figure. The incidence of other cancers is increased in • superficial spreading melanoma,
melanoma survivors as well.68 • acral lentiginous melanoma,
There is a lack of convincing evidence that pregnancy strongly influ- • nodular melanoma.
ences the outcome and prognosis of melanoma.69–71 Although earlier studies The majority of melanomas have a radial (in situ) growth phase before the
indicated that the mean thickness of pregnancy-associated tumors was sig- development of invasive tumor.114 The radial growth phase is greatest in
nificantly greater than that of nonpregnancy-associated melanomas, later lentigo maligna, of shorter duration in superficial spreading melanoma and
studies have not, and the clinical outcome parallels that of nonpregnant acral lentiginous melanoma, and, by definition, absent in nodular melanoma.
female patients of the same age and tumor characteristics, though dissenting Our understanding of melanoma has been radically transformed through
meta-analyses remain.72–84 Similarly, there is no clearly acceptable proof that research of the genomics and tumor biology of this disease. These topics
the use of oral contraceptives by females increases the risk of developing are discussed in the final section of this chapter, under the molecular clas-
melanoma.69,74,85 sification of melanoma. A number of efficacious targeted treatments are
Familial melanoma accounts for between 8% and 14% of affected available for melanoma such as those inhibiting the constitutively activated
patients.86,87 Genes that have been implicated include CDKN2A (9p21) signaling proteins produced by mutated BRAF and KIT genes. In addition,
(the majority), and very occasionally CDK4 (12q14). The former encodes the current ability to unleash the patient’s own immune system through the
p16, which normally functions to inhibit CDK4 activity.88,89 Approximately application of immune checkpoint inhibitor therapy is providing treatment
20–30% of patients with familial melanoma have germline mutations in for many patients for whom there was little to offer in the past. Such ther-
CDKN2A.86,90,91 Genetic variants in MC1R encoding the melanocortin 1 apies are transforming the treatment landscape for patients with metastatic
receptor and associated with red hair appears to increase the penetrance of disease and much research is ongoing to test such approaches in lower stage
melanoma in CDKN2A and CDK4 germline families.92,93 BAP1 germline disease with adjuvant approaches. Much research is ongoing to identify
deficits are associated with familial ocular melanoma.94–96 Researchers are which subsets of patients will benefit from immuno-oncological approaches
focusing on other predisposition genes as well, such as EBF3, though addi- and why some patients respond while others do not. Many factors includ-
tional work is needed.97 Familial melanoma patients are often younger than ing tumoral mutational load, tumor-infiltrating lymphocytes, serum lactate
those who develop sporadic melanoma and tumors are frequently multiple. dehydrogenase (LDH) levels, oncogenic signaling pathways, gut micro-
Dysplastic nevi can be present.98 biome, and immune checkpoint protein expression, to name a few, have
Melanoma shows a male preponderance (3 : 2) and males have a higher been correlated with responses. However, a full understanding of exactly
mortality.6,10,37 This probably relates at least in part to later presentation. how and why these agents work in some patients and not others has yet
Females generally have thinner tumors than males at first examination. The to emerge and our ability to predict is currently limited. Discoveries are
leg (particularly the calf) is the site of predilection in females, whereas the proceeding at an incredible rate in this area and thus are not addressed in
back is most often affected in males.37 Melanoma arising on the head and greater detail in this chapter.
neck is also more common in males than in females.37
The prognosis of melanoma is to some extent site dependent. Tumors
Lentigo maligna and lentigo maligna melanoma
arising on the BANS sites (upper back, posterior arm, posterior neck, and
posterior scalp) behave less favorably than those on the extremities.37,99 Lentigo maligna (Hutchinson melanotic freckle) is a relatively uncommon
The majority of melanomas arise de novo and as such, at least in their variant of melanoma and typically develops on chronic sun-damaged skin of
radial (in situ) growth phase, they clinically present as flat lesions. The most the elderly.115–117 In the United States, it is estimated to account for 4% of all
important signs for newly acquired potentially early melanoma include100: cases of cutaneous melanoma.118 It is characterized by a positive correlation

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
1312 Melanoma

Fig. 26.1  Fig. 26.3 


Lentigo maligna: this variably pigmented lesion was present for many years. Lentigo maligna melanoma: this is a very advanced lesion with a multinodular
There is no invasive component. From the collection of the late N.P. Smith MD, invasive component. By courtesy of J.C. Pascual, MD, Alicante, Spain.
the Institute of Dermatology, London, UK.

Fig. 26.2  Fig. 26.4 


Lentigo maligna: a dark black nodule of invasive tumor is surrounded by typical Lentigo maligna: the sun-damaged skin of the bald scalp is at risk for developing
lentigo maligna. From the collection of the late N.P. Smith MD, the Institute of this tumor. A concomitant ulcerated basal cell carcinoma is also evident above
Dermatology, London, UK. the melanoma. From the collection of the late N.P. Smith MD, the Institute of
Dermatology, London, UK.
with increasing age.119,120 Sites of predilection are the malar region, nose,
temple, and forehead; much less frequently acral sites, such as the dorsum border of the lesion is characteristic, and hypopigmented areas of regres-
of the hands, may be involved (Figs 26.1–26.5). The tumor presents most sion are also a frequent finding. Ulceration is an important diagnostic (and
often in the sixth and seventh decades as a variably pigmented, gradually prognostic) clue.
enlarging, irregular, flat macule. It may be brown or black and usually
shows areas of hypopigmentation representing areas of regression. The in
Acral lentiginous melanoma
situ lesion is often present for 10–15 years before invasive tumor develops.
Lentigo maligna melanoma is a term used to denote to lentigo maligna that Acral lentiginous melanoma accounts for approximately 8–10% of all mel-
has progressed to dermal invasion.121 anomas in Caucasians.123,124 It is, however, the predominant subtype affect-
ing Afro-Caribbeans and Asians.125–127 The tumor is particularly found on
the digits (especially beneath the nails) and on weight-bearing sites; plantar
Superficial spreading melanoma
tumors are the most common, with the heel being the most frequently
Superficial spreading melanoma is the most common variant and shows an affected region (Figs 26.7 and 26.8).128,129 Subungual variants, which most
equal sex incidence.122 The sites most frequently affected are the leg (espe- commonly affect the great toe and the thumb, are rare tumors, accounting
cially in females) and the back (particularly in males). The lesion initially for only 2% of all cutaneous melanomas. They show a female preponder-
presents as a flat scaly macule or plaque, which after a variable period of ance and present most often in the elderly.123 In addition to acral lentiginous
time develops a blue or blue-black nodule of invasive melanoma (Fig. 26.6). lesions, subungual melanoma can also present as superficial spreading and
Hypopigmented or amelanotic variants are erythematous or flesh colored. nodular histologic variants.130–132
The initial plaque is irregular, often 1–2 cm in diameter, and shows much Acral lentiginous tumors usually present as irregular, gradually enlarging,
greater variation in color than a banal or dysplastic nevus. Scalloping of the and variably pigmented macules. With progression to vertical growth phase,

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Histologic features 1313

Fig. 26.5  Fig. 26.7 


Lentigo maligna: close-up view. From the collection of the late N.P. Smith MD, Acral lentiginous melanoma: this variant arises on the non–sun-damaged skin of
the Institute of Dermatology, London, UK. the palms, soles, and under the nails. From the collection of the late N.P. Smith
MD, the Institute of Dermatology, London, UK.

Fig. 26.6  Fig. 26.8 


Superficial spreading melanoma: there is a large nodule of invasive melanoma Acral lentiginous melanoma: this ulcerated tumor had extended deeply into the
with a surrounding macular component. From the collection of the late N.P. Smith underlying tissues. From the collection of the late N.P. Smith MD, the Institute of
MD, the Institute of Dermatology, London, UK. Dermatology, London, UK.

frequently ulcerated, blue or black nodular lesions are encountered. In Cau- for a vascular tumor, such as lobular capillary hemangioma (pyogenic gran-
casians, acral lentiginous melanoma presents most often in the seventh uloma) (Figs 26.9 and 26.10).
decade, has an equal incidence in both sexes, and is generally associated
with a poor prognosis since tumors are generally thick by the time of diag-
Melanoma arising at noncutaneous
nosis. Mucosal melanomas are often classified within the acral lentiginous
(primarily mucosal) sites
spectrum, given certain partial morphological overlap. These two types of
melanoma are distinct clinically, although genomic characterization does Melanoma may arise at a diverse range of sites other than the skin, including
indicate shared features.103,133,134 the orbit, the oral cavity and nasal cavities, the external genitalia, vagina,
urethra, and anus.137–139 In general, mucosal tumors are associated with
aggressive behavior and a poor prognosis.138,140–143 This relates particularly
Nodular melanoma
to delayed presentation. Rare sites for primary melanoma also include the
Nodular melanoma (3–4%) has no radial growth phase and therefore may meninges, esophagus, stomach, uterus, cervix, breast, biliary system, bron-
be distinguished clinically from nodules of invasive tumor that have arisen chus, and adrenal gland.
in lesions associated with a pre-existent radial growth phase.135 It has a poor
prognosis (the majority being thick tumors by the time of excision), affects
Histologic features
more males than females (2 : 1), and generally arises in the fifth or sixth
decade.136 The trunk and limbs are most commonly involved. The tumor, Central to our understanding of the histologic classification of cutaneous
which may be nodular or polypoid (polypoid melanoma), is often ulcerated melanoma is the concept of radial (horizontal) growth phase and verti-
and, when lacking pigment (amelanotic melanoma), is frequently mistaken cal (invasive) growth phase.33,108,114,144,145 By current definition, the radial

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
1314 Melanoma

Fig. 26.9  Fig. 26.11 


Nodular melanoma: this heavily pigmented, dome-shaped nodule has no adjacent Melanoma: radial growth phase. The tumor is wholly intraepidermal (in situ
macular component. From the collection of the late N.P. Smith MD, the Institute melanoma). Intraepidermal tumor cells at all levels are present (pagetoid spread).
of Dermatology, London, UK.

Fig. 26.10  Fig. 26.12 


Nodular melanoma: amelanotic tumors as shown here are often a source of Melanoma: microinvasive
clinical (and histological) diagnostic difficulty. From the collection of the late N.P. radial growth phase. In
Smith MD, the Institute of Dermatology, London, UK. addition to in situ tumor,
small numbers of single
tumor cells are present in
growth phase may include (in addition to a wholly in situ, intraepidermal the papillary dermis.
component) evidence of microinvasion into the papillary dermis (microinva-
sive radial growth phase), often accompanied by features of regression (Figs phase (Fig. 26.14).33,145,151 Mitotic figures are common.33 Features of regres-
26.11 and 26.12). The microinvasive stage of melanoma is believed to lack sion may be seen but are usually absent at the base of the tumor. The tumor
significant metastatic potential and as a consequence is associated with an cells in the vertical growth phase are pleomorphic and apoptosis is often
excellent prognosis.146–149 Indeed, in a large series of patients discussed by present. Vertical growth phase implies an alteration in biological potential
Clark and coworkers, no tumors in the radial growth phase were associated with a capacity for lymphovascular invasion and metastatic spread.33,145,146
with metastatic spread.109 A subset of melanocytic tumors can be challenging to definitively assess
Histologically, the microinvasive radial growth phase tumor is charac- as either benign or malignant and are described under various names to
terized by single cells or small aggregates of melanoma cells, histologically indicate their uncertain malignant potential. Such cases often have some,
similar to their intraepidermal counterparts and invariably forming tumor but not all or fully developed features of the malignant phenotype histo-
nests smaller than those present within the overlying epidermis.33 A lym- logically. Experts often disagree on the designation of malignancy in this
phohistiocytic infiltrate is usually present. Mitotic figures are absent by defi- group of exceedingly challenging lesions.152–155 Multi-probe fluorescence in
nition. The last feature is of particular importance; multiple levels should situ hybridization (FISH) and genomic hybridization can assist diagnosti-
therefore be carefully examined before making a diagnosis of microinvasive cally, but in some cases the biological potential of these equivocal lesions
radial growth phase melanoma (Fig. 26.13).150 remains uncertain.156 Proposals have suggested that in such cases, group-
Vertical growth phase melanoma is composed of cohesive nests, nodules, ing the lesions into classes linked to the type or extent of excision needed
or plaques larger than those present within the epidermis and consisting of increases agreement and aids in communication of the clinical care needed
tumor cells that are cytologically different from those in the radial growth despite the uncertainty in the diagnosis.157

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Histologic features 1315

Fig. 26.13 
(A, B) Melanoma: lentigo maligna melanoma. This tumor was incorrectly
diagnosed as a microinvasive radial growth phase lesion on the grounds
that only single cells were present in the dermis and there were no nests.
A Dermal mitoses were, however, present (B). The patient developed cerebral
metastases.

Fig. 26.14  Fig. 26.15 


Melanoma: vertical Lentigo maligna: the epidermis is atrophic and flattened. Atypical melanocytes are
growth phase. In addition basally located and the superficial dermis shows marked solar elastosis.
to in situ (radial growth
phase) tumor, there are
multiple nests in the tumor thickness, particularly if obliquely cut sections are examined, when
dermis. These are larger the relationship of the tumor cells to the adnexal epithelium may not be
than the epidermal ones.
clear. In more advanced lesions, junctional nests are apparent and multinu-
cleate tumor giant cells are commonly seen (Figs 26.18–26.20). Pigmentation
is variable, but is often abundant, sometimes involving the full thickness of
Lentigo maligna and lentigo maligna melanoma
the epidermis, including the stratum corneum. The very occasional presence
Lentigo maligna is characterized by proliferation of atypical melanocytes of marked intraepidermal (pagetoid) spread may result in histologic overlap
predominantly located along the dermal–epidermal junction (Figs 26.15 with superficial spreading melanoma, though junctional nesting is usually
and 26.16).158 Chronic sun damage evidenced as extensive solar elastosis is less prominent in lentigo maligna. Assessing excision margins for residual
often prominent, particularly in cases from the head and neck region.159 The atypical melanocytes in this variant is often fraught with difficulty. To avoid
tumor cells often show a conspicuous cytoplasmic fixation retraction arti- unnecessary surgery it is important not to confuse actinically damaged nuclei
fact and contain pleomorphic irregular hyperchromatic, sometimes angular, from residual lentigo maligna (Fig. 26.21), though molecular studies suggest
nuclei. The cells are frequently orientated perpendicular to the surface, and that this may not be possible on strictly morphological grounds given the
involvement of hair follicles and sweat duct epithelium is a characteristic field effect of genomic aberration present in even morphologically normal
finding (Fig. 26.17). This may sometimes result in difficulties in assessing melanocytes.160 This likely underlies the elevated risk of local recurrence in

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
1316 Melanoma

A B

Fig. 26.16 
Lentigo maligna: (A) viewed with higher power, the melanocytes demonstrate a prominent fixation artifact. The nuclei are irregular, angular, and hyperchromatic; (B) in
this example, the tumor cells have abundant eosinophilic cytoplasm and there is early pagetoid spread.

Fig. 26.17  Fig. 26.18 


Lentigo maligna: note
Lentigo maligna: in this example there are conspicuous junctional nests. The
the presence of atypical
tumor cells have a spindled morphology.
melanocytes within the
outer aspect of a hair
follicle. Shave biopsy
of such lesions is
accompanied by a high
recurrence rate.

these lesions. Evaluation of the extent of epidermal involvement in difficult


cases or small samples can be done by immunohistochemistry ideally with
markers that highlight nuclei including MITF and SOX10.
Lentigo maligna arises at sites showing actinic damage; the epidermis is
therefore typically atrophic and the dermis shows solar elastosis. Usually the
papillary dermis contains melanophages and scattered chronic inflammatory
cells. The finding of the latter, particularly when present in large numbers,
is often associated with invasion and should therefore prompt careful exam-
ination of multiple levels of the specimen. Invasive tumor (lentigo maligna
melanoma) can be multifocal and is usually of the spindled cell type (Figs
26.22–26.24). Due to the extensive involvement of neighboring hair follicles
and cross-sectioning, determining the presence or absence of early invasion
can be very challenging and occasionally, impossible. Desmoplasia, often
with neurotropism, is present in a significant percentage of cases. Very occa- Fig. 26.19 
sionally, a storiform growth pattern is evident and if the tumor is amelanotic Lentigo maligna: the tumor cells are pleomorphic and show very marked nuclear
there may be confusion with dermatofibrosarcoma protuberans, particularly hyperchromatism.

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Histologic features 1317

Fig. 26.20 
Lentigo maligna: multinucleate tumor cells as shown in this example are a
common feature and a useful diagnostic clue. Fig. 26.22 
Lentigo maligna
melanoma: in this variant
of melanoma, the invasive
component is often of the
spindle cell type.

Fig. 26.21 
Actinic nuclear atypia: sometimes distinguishing between the former and residual
tumor cells as shown in this field can be a real problem. The former, however,
show little nuclear enlargement and nucleoli are absent. In addition, such cells do
not form nuclear palisades as is typical of lentigo maligna.

in small biopsies (Fig. 26.25). More often, the associated melanoma is des-
moplastic with subtle spindle cells and can be mistaken for fibrosis or scar
rather than invasive tumor. Fig. 26.23 
Exceptionally, lentigo maligna and lentigo maligna melanoma present Lentigo maligna
as amelanotic lesions (amelanotic lentigo maligna and amelanotic lentigo melanoma: higher-power
maligna melanoma), both clinically and histologically.161–164 Clinically, these view.
appear as erythematous scaly lesions resembling actinic keratoses, squamous
cell carcinoma in situ or eczema.162 Immunocytochemistry may therefore be
necessary to arrive at the correct diagnosis (Fig. 26.26).165 of the epithelium while the cells of Paget disease tend to be superficial
to the basal cell layer. The individual cells are epithelioid with abundant
cytoplasm, often showing fine or dusty melanin pigmentation, and contain
Superficial spreading melanoma (pagetoid melanoma)
pleomorphic vesicular nuclei with prominent eosinophilic nucleoli; scattered
The radial growth phase of superficial spreading melanoma is now encoun- mitotic figures including atypical forms may be evident. Characteristic of
tered more commonly, largely as a consequence of public awareness of mel- this variant is tumor growth in continuity from one rete ridge to another
anoma with the consequent removal of an increasing percentage of thin (a pattern not usually seen in acquired junctional or compound nevi). In
early lesions. It is typified by an asymmetrical proliferation of atypical non- contrast to lentigo maligna, there is often minimal visible evidence of actinic
dendritic melanocytes scattered singly and in clusters throughout all levels damage.159 The epidermis often shows acanthosis with partial or complete
of the epithelium (buckshot scatter), giving an appearance reminiscent of effacement of the ridge pattern. Invasive tumor is mostly of the epithelioid
Paget disease (Figs 26.27 and 26.28). The melanoma cells involve all layers type (Figs 26.29–26.32). Desmoplasia and/or neurotropism are uncommon.

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
1318 Melanoma

Fig. 26.24  Fig. 26.27 


Lentigo maligna melanoma: in this example, mitotic figures are conspicuous. Note Superficial spreading melanoma: there is prominent junctional activity and atypical
the nuclear hyperchromatism. melanocytes are widely scattered throughout the epidermis.

Fig. 26.25 
Lentigo maligna melanoma: occasionally the tumor adopts a storiform pattern.
Fig. 26.28 
When amelanotic, this may be confused with dermatofibrosarcoma protuberans
Superficial spreading melanoma: the melanocytes have abundant eosinophilic
or spindle cell squamous carcinoma.
cytoplasm and pleomorphic vesicular nuclei. Nucleoli are conspicuous.

Acral lentiginous melanoma


In the early stages of the radial growth phase of acral lentiginous mela-
noma the changes may be quite subtle, consisting of irregular epidermal
hyperplasia and scattered, basally located, atypical melanocytes.166–170 The
established lesion shows acanthosis with marked elongation of the epider-
mal ridges and obvious melanocytic atypia (Figs 26.33 and 26.34). The
lower reaches of the epidermis are infiltrated by large numbers of atypical
melanocytes characterized by nuclear pleomorphism and hyperchromatism,
and showing a cytoplasmic fixation retraction artifact. Nucleoli are con-
spicuous and mitotic figures may be identified. Although spindled forms are
most often encountered, epithelioid and giant cells are sometimes evident.
Scattered foci of junctional nests may also be detected, usually at the tips of
the epidermal ridges (Fig. 26.35). A heavy bandlike chronic inflammatory
cell infiltrate is frequently present. The invasive tumor is often a spindled
cell in type and may elicit a desmoplastic reaction (Fig. 26.36). Deep exten-
sion along the sweat gland epithelium is common and neurotropism may be
Fig. 26.26  evident in a subset of cases. Cross-sectioning of involved sweat glands may
Lentigo maligna: occasionally it is difficult to distinguish between actinic keratosis lead to the erroneous interpretation of invasion. Occasionally, acral tumors
and lentigo maligna. In such cases, immunohistochemistry using a red chromogen may show a superficial spreading in situ component or represent de novo
(in this case alkaline phosphatase) can make the distinction easy. nodular melanoma with absent radial growth phase.

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Cell types 1319

Fig. 26.29 
Superficial spreading Fig. 26.31 
melanoma: invasive Superficial spreading melanoma: diagnosis of amelanotic tumors, particularly when
tumor is usually of the very pleomorphic as in this example, often depends upon immunohistochemistry
epithelioid type as shown if a junctional component is not evident.
in this field. Note the
abundant cytoplasm,
nuclear pleomorphism,
and prominent nucleoli.

Fig. 26.32 
Superficial spreading melanoma: this example shows a mitotic figure in the center
of the field.

Fig. 26.30 
Superficial spreading melanoma: in this example there is heavy melanin Epithelioid cells
pigmentation.
Epithelioid melanoma cells are large and rounded with abundant, often
eosinophilic, cytoplasm and contain prominent pleomorphic vesicular nuclei
Nodular melanoma
with conspicuous eosinophilic nucleoli. Mitotic figures may be either scant
By definition, nodular melanoma has no evident preceding radial growth or numerous and sometimes abnormal. Pigmentation is variable and can
phase, its growth pattern appearing vertical from inception. Such a tumor, be abundant or minimal. When minimal, Masson-Fontana silver staining is
therefore, does not show an intraepidermal melanocytic proliferation useful to reveal small quantities of pigment not detectable with conventional
beyond three epidermal ridges on either side of the tumor mass (Figs 26.37 hematoxylin and eosin stained sections. Alternatively, immunohistochemis-
and 26.38). This histologic pattern is usually, but not always, seen in the try may be necessary to establish the diagnosis (Fig. 26.39).
same anatomic distribution as superficial spreading melanoma; these have
overlapping genomic features, but some regard them as distinct entities.135
Spindled cells
The spindled cell variant is characterized by cells with elongated, narrow,
Cell types
tapering, cytoplasmic processes, which in nonpigmented variants may be
Although many invasive tumors show a mixed pattern, melanoma cells are confused with cells of mesenchymal derivation and therefore misdiagnosed
typically divided into two major types: epithelioid and spindled cell. Epi- as a variety of soft tissue neoplasms.
thelioid cells most often arise within superficial spreading melanoma and Occasional melanomas are exceedingly pleomorphic, containing tumor
nodular melanoma, while spindled cells are more commonly seen in lentigo giant cells with enlarged nucleoli and conspicuous, frequently abnor-
maligna melanoma and acral lentiginous melanoma. mal, mitotic figures; these variants must sometimes be distinguished from

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
1320 Melanoma

metastatic tumors, including pleomorphic secondary carcinoma, sarco- resultant death, in the hope that adjuvant chemotherapy, immunotherapy,
mas, and even anaplastic CD30+ anaplastic large cell lymphoma (Figs or other forms of treatment might eventually benefit significant numbers
26.40–26.42). of patients. Prognostic indicators include clinical parameters, morpholog-
ical observations, and measurements in addition to immunohistochemical
markers of cell proliferation, cell regulation, and so on. Many of the mor-
Prognostic indicators
phological observations are tried and tested, but the search for the elusive
While the vast majority of thin melanomas do not metastasize, between immunohistochemical marker of metastatic potential has not yet been fruit-
1% and 2% are known to do so. Conversely, occasional thick melanomas ful. Genomic biomarkers are also being explored, in particular limited gene
fail to disseminate. With this in mind, there has been intensive research expression panels.171–173
in an effort to define those tumors that have the capacity for spread with Clinical prognostic indicators include age, sex, and site of the primary
tumor.174–180 Older patients fare worse than younger ones, and males have a
poorer outlook than females.7,176,177,181–184 The latter is independent of tumor
thickness and site.176 Particularly high-risk sites include the back, upper arm,
neck, and scalp (BANS).185 The acral sites are also thought to be associated
with a poorer prognosis.186
When reporting melanoma, it is generally accepted as essential world-
wide to record and comment on the following variables:176,187–193
• tumor thickness (Breslow method),
• level of invasion (Clark method),
• growth phase (vertical or radial),
• mitotic rate,
• ulceration,
• lymphovascular invasion,
• perineural infiltration,
• regression,
• microsatellitosis,
• tumor infiltrating lymphocytes.
Growth phase is also noted although it should be emphasized that great care
must be taken before allocating a melanoma to the microinvasive category
of radial (horizontal) growth phase. Dermal mitoses automatically place a
Fig. 26.33  tumor in the vertical growth phase (see Fig. 26.14). These elements have
Acral lentiginous melanoma: in this in situ lesion, there is irregular acanthosis, become critical for proper staging and prognostic assignment of patients,
hypergranulosis, and hyperkeratosis. Tumor cells are hyperchromatic and but unfortunately even in countries with a high incidence and acute aware-
distributed in a lentiginous and nested pattern. The dermis is scarred and there ness of cutaneous melanoma, many of the above elements are absent from
are conspicuous melanophages and chronic inflammatory cells. routine reports.194

A B

Fig. 26.34  Fig. 26.35 


Acral lentiginous melanoma: there is conspicuous (A, B) Acral lentiginous melanoma: large junctional nests are present at the tips of the rete.
cytoplasmic retraction, hyperchromatism, and
nuclear atypia.

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Prognostic indicators 1321

Fig. 26.36  Fig. 26.38 


Acral lentiginous melanoma: in this example, the invasive component is mixed Nodular melanoma: by definition, there is no melanocytic proliferative activity in
epithelioid, spindled cell, and desmoplastic. the adjacent epidermis.

Fig. 26.37 
Nodular melanoma: this
variant is typically sharply
circumscribed.

Breslow tumor thickness is the single most important single prognos-


tic indicator for primary cutaneous melanoma.195,196 It is measured from
the most superficial aspect of the granular cell layer to the deepest point
of invasion of the tumor. In ulcerated tumors, the measurement should
be from the base of the ulcer. The significance of periadnexal extension is
less clear. If the latter is greater than the conventional Breslow thickness,
the information should also be documented in addition to the traditional
Breslow measurement (Fig. 26.43). Polypoid tumors should be treated no
differently, and should be measured through the thickest region.197 Tumor
B
volume, therefore, is a most important factor influencing outcome in ver-
tical growth phase melanoma. Careful evaluation of the greatest tumor
Fig. 26.39 
thickness provides very useful prognostic guidance.198–201 In the latest (8th) Amelanotic melanoma: (A) the tumor cells are very pleomorphic with large
edition of the American Joint Committee on Cancer (AJCC) staging system vesicular nuclei and prominent nucleoli; (B) there is diffuse S100 expression.
(2017, effective in 2018), the thickness thresholds have been maintained as MART-1 was also positive. It is always of value to use at least two markers
in the 7th Edition at 1.0, 2.0, and 4.0 mm, though the T1 group is subdi- for melanoma as very occasionally S100 protein negative variants may be
vided 0.8 mm as well (Table 26.1).196,202–206 The 7th edition staging is still encountered.

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
1322 Melanoma

Fig. 26.40  Fig. 26.43 


Giant cell melanoma: within the dermis is a deposit of metastatic melanoma. Melanoma: periadnexal extension is of uncertain biological significance except
that it is a potential source for recurrence of inadequately excised tumors. In this
example, there is deep involvement of the eccrine sweat glands.

Major changes and clarifications of the 2017 AJCC melanoma


staging system

• Database completely derived from patients of known sentinel lymph


node status thus creating more homogeneous stage groups
• T0 designates no evidence of primary tumor; Tis designates in situ
disease; TX designates tumor thickness that cannot be determined
• T categories maintained, but T1 with new 0.8 mm threshold
• Tumor thickness now reported only to nearest 0.1 mm
• Mitoses in primary tumor are recorded for prognosis, but no longer
used for staging
• Clark level no longer used for staging primary
• For regional lymph node involvement, “microscopic” is now termed
“clinically occult” and “macroscopic” termed “clinically detected” for
clarity
• Sentinel lymph node is disease burden is prognostic and thus to be
recorded, but not currently used for staging
• N category now has 4 subgroups rather than three
• Status of the primary (Breslow thickness and ulceration) extensively
Fig. 26.41  modifies stage III groupings
Giant cell melanoma: the tumor cells are multinucleate and very pleomorphic with • In transit, satellite and microsatellite metastases are now categorized
abundant cytoplasm and large vesicular nuclei containing prominent eosinophilic as N1c, N2c, N3c based on how many other regional nodes are
nucleoli. involved (if any)
• Given the rapidly evolving systemic therapy landscape, Stage IV is no
longer subdivided; metastatic site designation and LDH elevation are
used solely to define clinically relevant patient groupings

provided here for purposes of comparison (Table 26.2). A pleasant feature


of the new staging system is that Breslow thickness is now only reported to
one significant digit after the decimal point, simplifying measurement with
the micrometer. While certainly not perfect, clinical and particularly micro-
pathological staging in melanoma is exceedingly powerful and serves as a
model for other cancers.
The level of tumor invasion when classified according to Clark levels is
as follows:
• Level I: in situ melanoma,
• Level II: invasion of the papillary dermis by single cells or small nests,
• Level III: invasive tumor usually as an expansile nodule abutting on the
reticular dermal interface,
• Level IV: invasion of the reticular dermis,
• Level V: invasion of the subcutaneous fat.
The dermal interface may be identified by the site of the superficial capillary
Fig. 26.42  plexus. It also corresponds to the zone of transformation of the horizontally
Giant cell melanoma: the tumor cells are HMB-45 positive. The patient had a orientated reticular dermis elastic fibers to the vertically aligned ones of the
melanoma excised several years earlier. papillary dermis. The distinction between Clark level II and III and perhaps

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Prognostic indicators 1323

Table 26.1 
AJCC 2017 (8th Edition) revised melanoma pathological staging system (effective, January 2018)

Disease specific survival1


Stage group Histological features/TNM designation 5 years (%) 10 years (%)
0 Intraepithelial/in situ melanoma2 (TisN0M0) 100 100
IA <0.8 mm primary without ulceration (T1aN0M0)
3
99 98
<0.8 mm primary with ulceration (T1bN0M0) 99 98
0.8-1.0 mm primary without ulceration (T1bN0M0) 99 96
IB > 1.0-2.0 mm primary without ulceration (T2aN0M0) 97 94
IIA > 1.0–2 mm primary without ulceration (T2bN0M0) 94 88
2.0–4.0 mm primary without ulceration (T3aN0M0) 93 88
94 88
IIB 2.0–4.0 mm with ulceration (T3bN0M0) 87 82
> 4 mm without ulceration (T4aN0M0) 86 81
90 83
IIC > 4 mm with ulceration (T4bN0M0) 82 75
IIIA Single regional node, clinically occult4 (T1–2aN1aM0) 93 88
2–3 regional nodes, clinically occult (T1–2aN2aM0)
IIIB Single regional node, clinically detected5 (T1–T3aN1bM0) 83 77
No regional node, in-transit / satellite disease (T1-2aN1cM0)
2–3 regional nodes, at least 1clinically detected (T1–2aN2bM0)
Single regional node, clinically occult (T2b–3aN1aM0)
Single regional node, clinically detected (T2b–T3aN1bM0)
No regional node, in-transit / satellite disease (T2b-3aN1cM0)
2–3 regional nodes, clinically occult (T2b–3aN2aM0)
2–3 regional nodes, at least 1clinically detected (T2b–3aN2bM0)
No evidence of primary tumor (T0N1b-1cM0)
IIIC Single regional node, occult or clinically detected; +/- in-transit / satellite disease (T1a-3aN2cM0) 69 60
4 or more regional nodes, clinically occult (T1a-3aN3aM0)
4 or more regional nodes, at least 1 clinically detected (T1a-3aN3bM0)
2 or more regional nodes clinically occult or detected and/or matted nodes; 2+ regional nodes
+/- in-transit / satellite disease (T1a-3aN3cM0)
Any regional node status greater than N1 (T3b-4aN2a-3cM0)
Any regional node status less than N3 (T4bN1a-2cM0)
No evidence of primary tumor (T0N2b-2c,3b-3cM0)
IIID Any N3 (T4bN3a-3cM0) 32 24
IV Distant metastasis (T1a-4bN1a-3cM1)
 M1a = distant metastasis to skin, soft tissue or nonregional node
 M1b = distant metastasis to lung +/- M1a site(s)
 M1c = distant metastasis to viscera (non-CNS) +/- M1a or M1b site(s)
 M1d = distant metastasis to CNS +/- M1a, M1b or M1c site(s)
 (0) = LDH normal / not elevated
 (1) = LDH elevated
1
Five and 10 year disease specific survival provided for overall stage groups as well as individually definied cohorts that comprise the stage group where available.
2
The underlined portion of the TNM designation is defined by the histologic features for each group.
3
Breslow thickness is defined as the thickness of the lesion using an ocular micrometer to measure the total vertical height of the melanoma from the top of the granular layer
to the area of deepest penetration (and reported to the nearest 0.1 mm). If the entire overlying epithelial surface is ulcerated, measure from the base of the ulcer to the depthest
point of invasion.
4
Clinically occult (previously termed microscopic) metastasis is defined as following pathologic assessment of sentinel lymph node and/or lymphadenectomy specimen. There
is no minimum size lesion for metastasis. Immunohistochemistry can be used to screen for micrometastases, but should include at least one ‘melanoma-specific’ marker (i.e.,
HMB-45, MART1, MelanA).
5
Clinically detected (previously termed macroscopic) metastasis is clinically evident nodal disease known prior to pathologic examination.
Adapted from AJCC Cancer Staging Manual, 8th Edition (2017) Eds: Amin MB, et al. Melanoma of the Skin, pp.563–585. Springer-Verlag, New York. © and Gershenwald, J. E.,
Scolyer, R. A. Hess, K. R., et al. (2017) Melanoma staging: Evidence-based changes in the American Joint Committee on Cancer eighth edition cancer staging manual. CA Cancer
J Clin, 67;472–492.

more so between III and IV is somewhat difficult to apply in practice and is defined as ‘the absence of an intact epidermis overlying a major portion of
is observer dependent.207 Clark level was believed to provide independent the primary melanoma based on microscopic examination of the histologic
prognostic information for thin tumors (1.0 mm or less in thickness) but sections’.203 Trauma and artifactual loss of the epidermis must be excluded.
not for thicker melanomas.208,209 The latest data indicate that if mitotic Ulceration is associated with a very significant increased risk of metastasis
rate is assessable in thin melanomas, then Clark level no longer provides and was first included as a second determinant in the T classification in the
additional prognostic information. The 2017 (8th) AJCC staging system no AJCC 6th Edition and retained in the current 8th Edition.196,203,206,208,209,212
longer recommends the use of the Clark level, even for thin melanomas, as Tumor-infiltrating lymphocytes are an important independent prog-
in the previous edition.196,203,204,206 Mitotic rate in the primary melanoma nostic variable and should be recorded as brisk, nonbrisk, or absent (Figs
correlates with sentinel lymph node positivity for metastasis.210 26.44–26.46).145,206,213–215 The brisk category implies lymphocytes present
The presence or absence of ulceration and the width of the ulcer are throughout the whole vertical growth phase or extending across its entire
independent prognostic indicators and should be recorded.175,211 Ulceration base. Nonbrisk tumor infiltrating lymphocytes implies focal infiltration

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
1324 Melanoma

Table 26.2 
AJCC 7th Edition Staging – For historical comparison only, no longer in clinical use

Overall survival
Stage Histological features/TNM classification 1 year (%) 5 years (%) 10 years (%)
0 Intraepithelial/in situ melanoma (TisN0M0) 100 100
IA ≤ 1 mm without ulceration and mitoses <1/mm * (T1aN0M0) 2
97 93
IB ≤ 1 mm with ulceration or level IV/V (T1bN0M0) 94 87
1.01–2 mm without ulceration (T2aN0M0) 91 83
IIA 1.01–2 mm with ulceration (T2bN0M0) 82 67
2.01–4 mm without ulceration (T3aN0M0) 79 66
IIB 2.01–4 mm with ulceration (T3bN0M0) 68 55
> 4 mm without ulceration (T4aN0M0) 71 57
IIC > 4 mm with ulceration (T4bN0M0) 53 39
IIIA Single regional nodal micrometastasis**, nonulcerated primary (T1–4aN1aM0) 78 68
2–3 microscopic regional nodes, nonulcerated primary (T1–4aN2aM0)
IIIB Single regional nodal micrometastasis, ulcerated primary (T1–4bN1aM0) 59 43
2–3 microscopic regional nodes, ulcerated primary (T1–4bN2aM0)
Single regional nodal macrometastasis***, nonulcerated primary (T1–4aN1bM0)
2–3 macroscopic regional nodes, nonulcerated primary (T1–4aN2bM0)
In-transit metastasis(es)/satellite lesion(s) without metastatic lymph nodes (T1–4a/bN2cM0)
IIIC Single microscopic regional node, ulcerated primary (T1–4bN1bM0) 40 24
2–3 macroscopic regional nodes, ulcerated primary (T1–4bN2bM0)
In-transit metastasis(es)/satellite lesion(s) without metastatic lymph nodes, ulcerated
primary(T1–4bN2cM0)
4 or more metastatic nodes, matted nodes/gross extracapsular extension, or in-transit metastasis(es)/
satellite(s) and metastatic nodes (anyTN3M0)
IV Distant skin, subcutaneous, or nodal metastasis with normal LDH (any T any NM1a) 62
Lung metastasis with normal LDH (any T any NM1b) 53
All other visceral metastasis with normal LDH or any distant metastasis with increased LDH(any T any 33
NM1c)
Breslow thickness is defined as the thickness of the lesion using an ocular micrometer to measure the total vertical height of the melanoma from the top of the granular layer to
the area of deepest penetration. The Clark level refers to levels of invasion according to depth of penetration of the dermis.
*Clark level is now only used when mitotic rate is not available in a T1 tumor. Level IV or V invasion would meet criterion for T1b.
**Micrometastases are defined as following pathological assessment of sentinel lymph node and/or lymphadenectomy specimen. There is no longer a minimum size lesion for
metastasis. Immunohistochemistry may be used to screen for micrometastases, but must include at least one ‘melanoma-specific’ marker (i.e., HMB-45, MART1, MelanA).
***Macrometastasis is clinically detectable with pathological confirmation or gross extracapsular extension on pathological examination.Adapted with permission from AJCC
Cancer Staging Manual, 7th Edition (2009) Eds: Edge SB, et al. Melanoma of the Skin, pp.325–344. Springer-Verlag, New York. © and Balch, C. M., Gershenwald, J. E., Soong, S.
J. et al. (2009) Final Version of 2009 AJCC Melanoma Staging and Classification. J Clin Oncol, 27; 6199–6206.

A B

Fig. 26.44 
(A, B) Melanoma: tumor infiltrating lymphocytes. Category A – Brisk: the lymphocytes infiltrate the tumor and extend along the whole of the base of the lesion.

only. Absent includes two categories: either no lymphocytes at all or lym- 27%, respectively.214 Recently, it has been suggested that tumor-infiltrating
phocytes are present but do not infiltrate the melanoma. Brisk lymphocytic lymphocytes are influenced by sex and sentinel lymph node status only
responses tend to be a feature of thin melanomas whereas absence of a correlated in men and not in women.216 Not surprisingly, the absence of
lymphocytic response is generally seen in thick melanomas.200,214 In a study tumor-infiltrating lymphocytes also correlated with metastasis to senti-
of 285 vertical growth phase tumors, the 10-year survival rates for brisk, nel lymph nodes as a surrogate marker of melanoma risk in a cohort of
nonbrisk, and absent tumor-infiltrating lymphocytes were 55%, 45%, and 887 patients with full multivariate analysis.215 Image analysis and feature

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Prognostic indicators 1325

A A

B B

Fig. 26.45  Fig. 26.47 


(A, B) Melanoma: tumor infiltrating lymphocytes. Category B – Nonbrisk: the (A, B) Regression: no residual epidermal component is present. Note the
lymphocytes infiltrate only part of the tumor. lymphocytic infiltrate, plasma cells, abundant melanin-containing macrophages,
scarring, and conspicuous vasculature.

extraction have been used to assess tumor-infiltrating lymphocytes with


salutary results.217 Assessing tumor-infiltrating lymphocytes in lymph node
metastases also has predictive value.218,219
In thin melanoma it is important to recognize the features of regression,
which may be particularly evident in the dermal component (Figs 26.47 and
26.48).145,220 These include absence or reduced numbers of malignant mela-
nocytes, degenerate (apoptotic) forms, and a chronic inflammatory cell infil-
trate (Fig. 26.49).221–223 Melanophages, horizontal scarring, isolated tumor
islands, and telangiectatic vessels are also commonly present in the later
stage. Clinically, regression presents as macular gray, white, or pink areas.
Although the importance of regression as a determinant of biological
behavior has been the subject of considerable argument in the literature,
a number of authors believe that, in thin tumors, it correlates with an
impaired prognosis, though this contribution may be minor relative to other
factors in many cases.223–232 Complete regression of an undiagnosed primary
melanoma may be the explanation for patients presenting with metastatic
tumor of unknown primary (Fig. 26.50).233 Some authors recommend senti-
nel node biopsy for thin melanomas if there is evidence of extensive (> 50%)
Fig. 26.46  regression, while other series do not show an association between regression
Melanoma: tumor and nodal status.176,234
infiltrating lymphocytes.
Mitotic rate is determined by the number of mitotic figures/1 mm2 of
Category C – Absent:
lymphocytes are present
tumor in the most mitotically active area. It has become clear that tumors
but they do not invade displaying a high mitotic rate are associated with a poorer prognosis and
the tumor. The category that mitoses are a more robust predictor of outcome than ulceration and thus
also includes complete mitotic rate was incorporated into the 7th AJCC staging system.145,204,235–240
absence of lymphocytes. While mitoses are still recognized as a prognostic factor, the current 8th

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
1326 Melanoma

AJCC staging system relies on ulceration rather than mitoses due to stage number of studies.215,248–253 The use of immunohistochemistry for D2-40 sig-
group stability.196,206 Despite earlier contrary evidence and the complex nificantly increases the sensitivity for detection of lymphatic invasion and
interrelations of tumor thickness, ulceration, and mitotic rate, mitotic rate also correlates with lymph node metastasis and survival.254–257 More study
appears to carry prognostic information independent of both of the other and a prospective trial is needed to validate the application and interpreta-
factors.176,241,242 tion of the technique. Contrariwise, absence of vascular involvement in thick
The presence of a microscopic satellite is defined by a distinct tumor melanomas correlates with increased survival in a number of studies.258,259
nodule of any size at any distance discontinuous from the main primary Perineural and intraneural infiltration are most often encountered in desmo-
tumor mass separated by normal tissue (no scarring, fibrosis or extensive plastic variants, thereby accounting, in part, for the increased risk of local
inflammation).206 It is found in thicker tumors and is associated with an recurrence (Fig. 26.52). In this context, CD57 immunohistochemistry can
increased risk of local recurrence, regional lymph node metastases, and be helpful in identifying residual nerve (Fig. 26.53).
diminished survival.243–246 The presence of microsatellites results in upstag- Angiogenesis, which is defined as the increasing development of new
ing to N1c if there is no regional lymph node involvement, and upstaging to blood vessels at the base of the melanoma, parallels increases in tumor
N2c or N3c with one or more than one lymph nodes involved, respectively,
in the 2017 AJCC melanoma staging system.206
The presence of lymphatic invasion correlates with the development
of in-transit metastases (Fig. 26.51).247 Lymphovascular invasion has been
shown to represent a predictor of diminished survival in melanoma in a

Fig. 26.49 
Regression: in this
example, there is
Fig. 26.48  conspicuous apoptosis.
Partial regression: in this variant, the junctional component is still present. Its Note the eosinophilic
significance is less certain. bodies.

Fig. 26.50 
(A, B) Regression: in this example, there is no residual tumor. The dermis is
scarred and there is abundant melanin pigment. The patient presented with an
A unknown primary. By courtesy of M. Forder, MD, St Anne’s Medical Center,
Pietermaritzburg, South Africa.

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Differential diagnosis 1327

Fig. 26.53 
Melanoma: CD57 immunohistochemistry may be particularly valuable when tumor
growth has largely destroyed the nerve.

thickness.260–265 Increasing angiogenesis therefore correlates with thick


tumors, ulceration, relapse, and tumor-associated death.263,266 Perhaps related
to the likelihood of lymphovascular invasion, increased intratumoral and
peritumoral lymphangiogenesis as measured by D2-40 or VEGF-C also cor-
relates with metastasis to sentinel lymph nodes and reduced disease-specific
survival.267,268
Sentinel node biopsy also adds extremely valuable prognostic informa-
tion.269–273 It has been shown to represent the most important factor in
determining likelihood of tumor recurrence and patient survival in stage I
and stage II disease.270 Completion lymph node dissection for positive sen-
tinel lymph nodes is expected to become less common with the MSLT-II
trial failing to show a survival benefit of the completion procedure.274,275
B The latest AJCC staging system requires incorporation of sentinel lymph
node data in cases where it would inform management, as this procedure
Fig. 26.51  significantly increases the accuracy and discriminatory power of the stage
Melanoma: vascular invasion: (A) pleomorphic tumor cells are adherent to the groups.204,206 Lymph nodes are bivalved or serially sectioned along their long
endothelium; (B) tumor cells are growing into the lumen. axis and in addition to the examination of hematoxylin and eosin stained
sections, immunohistochemistry – such as S100 protein, HMB-45, and/or
MART-1 (melanoma antigen recognized by T cells 1) – should invariably be
performed in those cases where the initial sections are negative. Detection of
single or tiny groups of cells using an antibody against a ‘melanoma-specific’
antigen such as HMB-45 or MART1 is now incorporated into the AJCC
staging system. More recently, molecular techniques including reverse tran-
scriptase polymerase chain reaction for tyrosinase messenger RNA have
been proposed.276,277 A major disadvantage of such procedures is the likeli-
hood of false-positive results due to the common presence of banal capsular
nevi, though the use of markers more specific to melanoma over nevus cells
could potentially alleviate this problem.278
Sentinel node biopsy is currently recommended for all tumors measur-
ing 1.00 mm or more in thickness. Other possible indications include ulcer-
ated tumors, tumors with 50% or more regression, tumors having achieved
the vertical growth phase, and those lesions which have been biopsied and
involve the deep margin.176,272,279,280 The procedure should also be consid-
ered for tumors that are less than 1.00 mm thick but display conspicuous
mitotic activity.281

Differential diagnosis
Amelanotic spindled cell melanoma may be histologically indistinguishable
from other cutaneous spindled cell tumors including leiomyosarcoma, spin-
dled cell squamous carcinoma, atypical fibroxanthoma, and even dermato-
Fig. 26.52  fibrosarcoma protuberans. In such cases, the use of an appropriate panel
Melanoma: intraneural invasion. Note the pleomorphic tumor nuclei within this of immunohistochemical reagents, including antibodies to S100 protein,
nerve trunk. SOX10, HMB-45 or MART-1, pan-cytokeratin, AE1/AE3, smooth muscle

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
1328 Melanoma

actin (SMA), and CD34, is often essential. Similarly, highly pleomorphic,


amelanotic epithelioid melanoma sometimes has to be distinguished from
anaplastic carcinoma and occasionally anaplastic lymphoma. The use of
antibodies to keratins, epithelial membrane antigen (EMA), carcinoembry-
onic antigen (CEA), leukocyte common antigen (LCA), T- and B-cell anti-
gens, and CD30 may be necessary to achieve a definitive diagnosis.

Immunohistochemistry of melanoma
Immunohistochemistry is a valuable adjunct to histology in the diagnosis
of melanoma, particularly in amelanotic, epithelioid, and spindled cell vari-
ants and their distinction from undifferentiated carcinomas and mesenchy-
mal tumors.1–3 Owing to problems of specificity and sensitivity, it is prudent
to use two or even three ‘melanoma markers’ in such problematical cases.
Using these markers as part of a panel looking at multiple lines of differ-
entiation is also of practical use, such as inclusion of keratins to exclude
epithelial tumors. In morphologically challenging cases, a panel of stains
that supports the ultimate diagnosis by their pattern of reactivity or nonre- A
activity is very helpful. The role of immunohistochemistry is to provide sup-
portive data. It should rarely if ever be used as the sole criterion by which a
diagnosis of melanoma is achieved.
S100 protein remains the yardstick in the immunohistochemical diag-
nosis of melanoma.4–6 Although there are now substantial numbers of new
markers available, none as yet, in isolation, measures up to S100 protein.
However, S100 protein lacks specificity and there are very exceptional
S100 protein-negative melanomas, virtually all metastatic.7,8 In cases where
the diagnosis remains in doubt, use of a battery of immunohistochemical
markers may be of great value.1
S100 protein is a calcium binding F-band protein, isolated from brain.
It is variably positive in 94–100% of primary and metastatic melanomas.1,2
In addition to melanocytes, Schwann cells, myoepithelial cells, adipocytes,
chondrocytes, macrophages, Langerhans cells, and tumors derived thereof,
express S100 protein. Staining of Langerhans cells can sometimes be a
problem, particularly when assessing the extent of intraepidermal melano-
cyte spread. In such instances, the addition of MITF, SOX10, HMB-45, or
MART-1 may be helpful. S100 protein may also be expressed in a number of B
breast carcinomas and undifferentiated carcinomas.5,6 The most commonly
employed antibody against S100 is a purified rabbit polyclonal antibody Fig. 26.54 
against S100 protein purified from bovine brain. More than 20 members of Melanoma: this tumor has arisen in a background of a banal nevus. (A) Compare
this family exist and monoclonal antibodies are available for many of them. the eosinophilic, pleomorphic tumor cells with the small basophilic nevus cells;
While not well established in large series, there may be some selectivity of (B) the melanoma cells are HMB-45 positive; the nevus cells are negative.
the isoforms between melanoma and other traditionally S100 protein reac-
tive neoplasms in the differential diagnosis.9 Sertoli cell, and granulosa cell tumors, and tumors in the PEComa group of
HMB-45 reacts with the cytoplasmic premelanosome glycoprotein gp100 lesions including angiomyolipoma, lymphangiomyomatosis, and clear cell
and is less sensitive than S100 protein.9 It is expressed by 80–86% of met- sugar tumors of the lung in addition to melanoma.18,19 It has a similar sensi-
astatic melanomas and between 90% and 100% of primary tumors.1,4,9 tivity to S100 protein in epithelioid melanomas but is less sensitive in spin-
However, expression is often more focal than with S100. Sensitivity dimin- dled cell tumors and is not usually expressed in the desmoplastic variant.
ishes in spindled cell variants and it is usually negative in desmoplastic mel- A study has shown evidence that diminished MART-1 expression correlates
anoma (see below).10,11 The junctional and superficial dermal components with increasing tumor thickness, reduced disease-free interval, and increased
of banal nevi also react with HMB-45 but the deeper dermal nevus cells are patient mortality.18 MART-1 expression has also been demonstrated in
generally negative. This differential staining pattern may be of value in con- compound, dermal nevi and Spitz nevi with the exception of neurotized
firmation of pre-existent banal nevus cells associated with a melanoma and variants.15–17
in particular their distinction from small cell and nevoid melanoma, which Antityrosinase antibody (e.g., T311) appears to be less sensitive than
are typically positive in a patchy fashion in the deepest nests of cells (Fig. either S100 protein, HMB-45, or Melan-A (A103).21–26 It does not appear
26.54). It is very important to remember that this pattern is lost in cases to label desmoplastic melanoma. Cocktails of HMB-45, MART-1, and
where melanocytes are pigmented as the latter are positive for HMB-45. antityrosinase antibodies are used by some to increase sensitivity for detec-
Dysplastic nevi are similarly labeled. Blue nevi and deep penetrating nevi tion of melanocytic differentiation.27
are also HMB-45 positive.1 Spitz nevi are often HMB-45 positive in the Microphthalmia transcription factor (MITF) is a transcriptional regula-
superficial aspect of the lesion and this is usually, but by no means always, tor important for tyrosinase expression.28 It is strongly positive in nevi and
lost with depth. This finding may be of value in its distinction from spit- epithelioid melanoma.29–31 Sensitivity is reduced in spindled cell and des-
zoid melanoma in which staining is typically present throughout the tumor. moplastic variants.32 Spitz nevi and neurotized banal nevi show diminished
Although it is more specific than S100 protein, HMB-45 also reacts with the expression. The specificity of MITF is low and this limits its use in the
group of perivascular epithelioid cell tumors (PEComas) including angiomy- differential diagnosis of mimickers of melanocytic lesions. Its main use is
olipoma, lymphangiomyomatosis, and clear cell sugar tumor of the lung.12,13 in the evaluation of intraepidermal melanocytes as the staining is nuclear
MART-1 (Melan-A) is a melanosomal differentiation antigen recognized and other cells that reside in the epidermis are not positive for this marker.
by autologous cytotoxic T cells.14–20 Some antibodies raised to this protein The monoclonal antibody, SM5-1, was created by a subtractive immu-
(e.g., A103) label a variety of lesions including adrenocortical, Leydig cell, nization protocol using human melanoma samples and binds a variant of

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Immunohistochemistry of melanoma 1329

fibronectin.33,34 Initial reports indicated that its sensitivity is similar to S100


protein with improved specificity for other traditionally S100 protein reac-
tive, but the antibody also reacts with hepatocellular carcinoma and breast
cancer cells.26,35 This antibody is not widely used for clinical diagnosis.
SOX10 is a transcription factor that is a critical regulator of
melanocytes.36–38 Antibodies raised against it represent another marker of
melanocytic and Schwann cells and tumors derived from the neural crest.39
While it lacks specificity and stains other cells it is equally sensitive and
more specific than S100 protein in relevant differential diagnoses with soft
tissue neoplasms.40,41 It is also useful in desmoplastic melanoma, though
expression has also been noted in scars.42–44 It has gained widespread use
as a sensitive marker for melanoma, often used in combination with other
melanocytic markers.45 It has been suggested to be more specific than other
melanocytic markers in the setting of determining melanocytic density in
chronically sun-damaged skin where misleading false-positive rates are
noted with other markers.46 A role has also been suggested in evaluation of
sentinel lymph nodes.47
The bar is high for inclusion of new markers as standards for melanoma
in clinical practice. All antibodies have a natural history of decreasing speci-
ficity with study of additional tumor types and new antibodies are not as far Fig. 26.55 
down this curve as older antibodies, creating potential for misinterpretation Melanoma: this tumor (right side of field) has arisen in a background of a
congenital nevus (left side of field).
if the new reagents are used alone.
Melanoma cells can express epithelial markers including keratins, EMA,
and CEA.2,48–50 Keratin, particularly those of low molecular weight, may
be identified in as many as 10% of melanomas, both on frozen and on
paraffin-embedded sections.2 CEA is commonly encountered if polyclonal
antibodies are utilized.49,50 Metastatic disease more often shows such aber-
rant staining patterns than primary tumors. Diagnostic difficulties are
unlikely to be encountered provided S100 protein and/or other melanoma
markers have been included in the antibody panel. SMA and desmin are
very rarely expressed in melanoma with the exception of the desmoplas-
tic variant where, as in most spindle cell tumors, SMA can be detected to
varying degrees.51 One study with dual labeling for S100 protein and SMA
in desmoplastic melanoma indicates that the SMA reactivity may be in
accompanying stromal myofibroblasts.52
Melanoma cells can express histiocytic markers such as CD68 (KP1) in
80% or more of tumors.53,54 Mac 387 and α1-antitrypsin may also be pos-
itive. This is of particular significance since the distinction between tumor
cells and histiocytes, particularly in sentinel lymph node specimens, can
sometimes be problematical. In addition, distinguishing between balloon
cell melanocytic lesions and xanthomatous infiltrates may require immu-
nohistochemical confirmation, particularly if no residual recognizable mela-
nocytic component is visible. In such circumstances, positive melanocytic
markers are obviously of major diagnostic importance.
There is an ever-expanding range of reputed immunohistochemical A
prognostic markers.55–57 Most of these lack appropriately powered full
multivariate analysis linking the marker with specific outcome such as
melanoma-specific mortality supplemented by hazard ratios and do not fully
describe the methods utilized as recommended by the REMARK (Report-
ing recommendation for tumor MARKer prognostic studies) guidelines of
the NCI-EORTC or AJCC precision medicine reporting recommendations
for tumor marker prognostic studies.58–62 The more useful of these are very
briefly discussed below. Few of these are validated to the level necessary
for routine application to clinical samples; virtually all are reported in ret-
rospective cohorts with referral and other biases. Use of multiple markers
and application of rigorous methods of quantification have efficacy, distin-
guishing different prognostic groups.63 The markers discussed below are
primarily used to help support a diagnosis of melanoma or nevus in the
relatively small subset of cases where this determination is challenging on
purely histologic grounds.
Ki-67 (MIB-1) is a particularly valuable adjunct in the distinction
between benign melanocytic nevi (including Spitz nevi) and melanoma.64,65
In nevi, less than 5% of nuclei are positive (and these are usually located B
in the most superficial aspect of the dermal component) whereas in mel-
anoma 25% or more of cells are labeled (Figs 26.55 and 26.56). Its role Fig. 26.56 
in predicting biological behavior is controversial; thus although in earlier (A, B) Melanoma: the melanoma shows very brisk MIB-1 expression, and the
studies increased expression in thick tumors was thought to correlate with nevus is completely negative.

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
1330 Melanoma

poor survival, more recently it has been claimed that increased expression including some that are U.S. FDA-approved companion diagnostics, but
in thin tumors (< 1.5 mm) is of greater significance.66–69 A further study indi- such testing is not currently required to use immune checkpoint inhibitor
cates that increased MIB-1 reactivity is a poor prognostic factor in terms of therapy in melanoma.123 Indeed, melanomas can respond to PD-1 (CD247/
disease-specific survival independent of tumor thickness.70 Obviously, there programmed cell death protein 1) inhibitors even if they completely lack
is a proportional, though not exact, relationship between degree of nuclear membranous PD-L1 expression.124 Most PD-L1 antibodies have very similar
MIB-1 reactivity and mitotic rate. Mitotic rate is an important determinate staining profiles in melanoma and other tumors as well.125,126 Specific scoring
of melanoma outcome, though it no longer drives groupings in the 8th approaches have been suggested and used for clinical trials and in routine
Edition of the AJCC staging system.71 How to correlate Ki-67 reactivity practice. Staining for CD8-positive T-cells in melanoma can also be per-
with mitotic rates is evolving. Increased Ki-67 expression also correlates formed and higher levels are associated with increased likelihood of response
with overexpression of p53 protein and loss of p16 (see below).68,70 to immuno-oncological therapy. Approaches for scoring these infiltrates have
Phosphohistone H3 (PHH3) recognizes mitotic figures directly rather been suggested, but are not widely employed in routine clinical practice.127–130
than cell cycle like MIB-1 and can assist in more objectively determining These biomarker approaches are used primarily in metastatic disease and
mitotic rates which do correlate with outcome.72–79 Care must be taken their correlation with response is not perfect.131,132 This is an active area
when increasing the sensitivity of mitotic rate detection since the majority of research and a number of genomic and other patient factors have also
of the data is derived using the less sensitive, but currently gold standard been associated with patient response. Companion immunohistochemical
microscopic detection on H&E stains, as inappropriate inflation of progno- approaches are not currently used in clinical practice for CTLA-4 (CD152)
sis can occur. inhibitors in melanoma. Anti-CTLA-4 therapy is sometimes used in combi-
Bcl-2 is strongly expressed in normal melanocytes and in a variety of nation with PD-1 inhibitors. PD-L1 expression can inform whether PD-1
nevi including banal, congenital, Spitz, blue, and dysplastic variants.80–83 monotherapy or combination approaches are employed in melanoma.124
Although there is some variability of results in the literature, diminished Additional work in this area is likely to produce more associated biomarkers
expression has been said to correlate with melanoma, particularly meta- as immuno-oncology expands to a number of emerging novel agents such
static lesions. Similarly, in problematical spitzoid lesions, strong expression as inhibitors of IDO1, LAG-3, and others where expression of the protein
may favor a diagnosis of benignancy.84 Bcl-2 downregulation has been cor- targets might be associated with treatment efficacy.133,134
related with melanoma progression.85–87 High bcl-2 was associated with
improved disease-specific outcome when assessed in either primary or met-
Histologic variants of melanoma
astatic lesions using automated assessment of immunofluorescent immu-
nohistochemistry.88 From our own experience, however, bcl-2 is as often Quite a range of histologic variants have been described.1–3 With the excep-
positive in melanoma as it is in banal nevi and it is therefore of little value tion of desmoplastic melanoma, these are of limited prognostic significance
in difficult cases. but are of importance because they may easily be mistaken for other tumors,
Cyclin D1 may be of help in differentiating between a banal nevus and a with potentially serious consequences. In addition, some of the types are
nevoid melanoma or a nevic component within a melanoma. Its expression more or less likely to have mutations such as BRAF V600E that represent a
throughout the full thickness of a melanoma has been reported in up to therapeutic target, though the genotype histology correlation is not enough
60% of cases, whereas staining may be absent or limited to only occasional for treatment in most cases and direct molecular testing is required.4,5
cells in the superficial aspect of a nevus.89,90 Increased cyclin D1 reactiv-
ity may correlate with amplification of its encoding genetic locus.91–93 High
Nested melanoma
levels of cyclin D3 in superficial melanoma have been found to correlate
with early relapse and decreased survival.94 This finding regarding cyclin This is a rare, recently described distinctive type of melanoma described as
D3 needs further confirmation.90 Interestingly, cyclin D1 may interact more a variant of superficial spreading melanoma.1,2 It presents in middle-aged to
with CDK4 while cyclin D3 interacts more readily with CDK6, suggesting elderly adults with the same sex incidence. Although the clinical features are
different functions for these two cyclin D isoforms.95 usually suspicious of melanoma, the histologic appearances are misleading.
Loss of p16 protein expression correlates with invasive and metastatic Tumors are in-situ or invasive and the junctional component is distinctively
melanoma.96–99 It has also been shown to correlate with increased Ki-67 nested with little or no proliferation of melanocytes between the nests. The
immunolabeling and independently to predict a poor prognosis.70,98 P16 latter may be confluent and are located at the tips or less commonly, the
may also be helpful in differentiating capsular nevus from metastatic mela- slopes of the rete-ridges. Mitotic activity tends to be low within the nests
noma in sentinel lymph nodes.100,101 and cytologic atypia varies from moderate to severe. Multiple chromosomal
Although mutation of the TP53 gene is relatively uncommon in mela- abnormalities are demonstrated by cytogenetic analysis.
noma at around 20%, overexpression of p53 protein may be identified in
up to 40% of cases of invasive and metastatic melanoma, perhaps indicat-
Minimal deviation melanoma
ing disruption of this pathway.102–107 It may therefore be of value in distin-
guishing between nevi (banal and Spitz) and melanoma. The former show The concept of minimal deviation melanoma was introduced to define a
virtually no positive cells whereas in the nodular and to a lesser extent subset of invasive melanoma that showed minimal histologic deviation from
superficial spreading melanoma they may be conspicuous. Expression of banal nevi and therefore lacked the pleomorphism seen in classic melanoma.
p53 also appears to be an independent predictor of poor prognosis.68,106,108 It was further postulated that such tumors appeared to have a better prog-
Immunohistochemistry for the BRAF V600E mutation using the VE1 nosis than classic melanoma.1–8 Over the last decade, use of this nomen-
clone is a highly sensitive and specific means of determining the mutational clature has precipitously declined and it is now presented primarily for
status of melanoma, usually applied in the metastatic setting.109–111 It does historical interest.
not reliably detect common alternative BRAF V600 variants such as V600K As defined, two subtypes were recognized6:
that constitute 15–20% of BRAF mutations in some series.112,113 This can • borderline minimal deviation melanoma where the dermal component is
be helpful in the rapid assessment for eligibility for BRAF inhibitor therapy, confined to the papillary dermis (Clark level 3),
but DNA sequencing is still the gold standard for determining BRAF muta- • minimal deviation melanoma where there is infiltration into the
tional status. A similar approach can detect a subset of the common NRAS reticular dermis or beyond (Clark level 4 or 5).
mutant proteins Q61R and Q61L in melanoma with two separate antibody Minimal deviation melanoma is characterized by the presence of an expan-
clones, but is not widely adopted.110,114–118 sile nodule showing only mild to moderate cytological atypia. It lacks the
In the setting of immunotherapy, membranous expression of PD-L1 (pro- disorderly, more marked pleomorphism of conventional epithelioid or spin-
grammed death-ligand 1) can influence the choice of immuno-oncological dled cell melanoma. The spectrum includes epithelioid variants composed
agents, but is not required to use such approaches.119–122 A number of antibodies of cells resembling those of a compound melanocytic nevus, spitzoid forms,
are available against PD-L1 for use in routine clinical immunohistochemistry, spindled cell variants, and halo nevus-like lesions.1

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Histologic variants of melanoma 1331

Fig. 26.57  Fig. 26.58 


Verrucous nevoid melanoma: verrucous nevoid lesions in the elderly should be Nodular nevoid melanoma: the diagnosis of nodular nevoid melanoma depends
viewed with suspicion. upon careful scrutiny of all ‘nevi.’ Inspection at scanning magnification will ensure
misdiagnosis as a banal nevus!

Although widely publicized, the concept has not gained general accep-
tance.1 This relates particularly to a lack of precise definitions combined
with the well-known unpredictable behavior of many melanomas. Patients
with apparently high-grade and thick tumors sometimes survive for many
years, or even decades, while the converse may also be true. The postu-
lated good behavior of these lesions has been challenged on the basis that
those that neither recurred nor metastasized may have been benign nevi and
those that metastasized and/or proved fatal were obviously malignant at the
outset.
In our view, although there are histologic variants of melanoma that
show only limited morphological deviation from banal nevi, the evidence
relating to biological behavior suggests that these tumors fare no better than
classical melanoma and should be treated in exactly the same way. Sub-
types that could be included within this category of ‘minimal deviation’ are
nevoid melanoma, small cell melanoma, and spitzoid melanoma. We prefer
these categories for histologic classification and these three are dealt with
below.

Nevoid melanoma Fig. 26.59 


Nevoid melanoma:
Nevoid melanoma is a rare histologic subtype of vertical growth phase expansion of the papillary
melanoma, which on low-power inspection may be (and often is) confused dermis by a diffuse
with a banal melanocytic nevus.1–5 It can be equally challenging to recognize nevoid population with
stretching and thinning of
clinically on patient skin examinations as well.6 Diagnosis depends on a
the associated epidermis
high index of suspicion and careful attention to cytological detail includ-
may be a clue to the
ing a thorough search for dermal mitotic activity. A practical or functional diagnosis.
definition of nevoid melanoma is the ‘diagnosis of a nevus which one later
regrets’.
Because nevoid melanoma is commonly misdiagnosed as a banal nevus, with hyperkeratosis and marked papillomatosis. The epidermis frequently
subsequent delay in appropriate treatment is common, with potentially dev- appears attenuated with loss of the rete-ridge pattern. In nodular tumors, the
astating consequences.1,2,7–12 Confusion with a banal nevus is most probably epidermis is similarly thin and stretched directly over the surface of a dense
a reflection of too cursory an inspection of a ‘nevus,’ frequently at scan- dome-shaped tumor cell population. Junctional activity in either variant is
ning magnification. Grossly, there are no particular distinguishing features, often minimal and limited to atypical cells distributed predominantly along
most lesions being clinically described as verrucous to dome-shaped variably the basal layer of the epidermis. Very occasionally, more obvious in situ
pigmented nevi or non-specific papules or nodules. Follow-up information melanoma may be seen.
indicates a recurrence rate of 50%, a metastasis rate of 25–50%, and a At medium-power examination, diffuse and, less often, nested growth
mortality rate of at least 25%.12 patterns may be encountered (Figs 26.59 and 26.60). In the latter there is
Histologically, on low-power examination nevoid melanoma may present often considerable variation in nest size accompanied by little tendency for
as a warty/verrucous or dome-shaped lesion (the former pattern in an elderly diminution in nest size with depth.
patient is itself a clue to the diagnosis, as nevi in the elderly are more often The infiltrating tumor cells are small, epithelioid melanocytes (reminis-
dome-shaped papulonodules) (Figs 26.57 and 26.58). cent of type-A nevus cells) with pale staining or eosinophilic cytoplasm and
The verrucous variant has a rather characteristic histologic appearance round to oval vesicular nuclei with small eosinophilic nucleoli. Although the
presenting as an often asymmetrical, poorly circumscribed, warty lesion low-power impression may suggest maturation with depth, closer inspection

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
1332 Melanoma

A
Fig. 26.60 
Nevoid melanoma: the
tumor cells are very
uniform. Nuclei are
vesicular and nucleoli
prominent. A small
number of tumor cells are
evident in the overlying
epidermis.

Fig. 26.62 
(A, B) Nevoid melanoma: this lesion at low power is suggestive of a congenital
nevus. At high-power magnification, multiple mitoses were present.

Fig. 26.61 
Nevoid melanoma: there
may sometimes be a
suggestion of maturation
with depth. Note the
mitosis. This field is taken
from the deep aspect
of the lesion shown in
Figure 26.58.

invariably shows this to be false (Fig. 26.61).13 Pleomorphism, while subtle,


is typically present and mitoses (frequently multiple) can invariably be iden-
tified, often within the deeper aspect of the tumor. Pigmentation, although
variable, is sometimes seen in the deeper reaches of the dermal component
and in a minority of cases, tumor-infiltrating lymphocytes are present. Occa-
sionally, perineural infiltration is present (Figs 26.62 and 26.63). A single
report describes a case with Homer-Wright type rosettes.14
Verrucous variants must be distinguished from keratotic melanocytic
nevi, the most important histologic discriminants being the lack of mat- Fig. 26.63 
uration, subtle pleomorphism, and mitotic activity.15 Others have termed Nevoid melanoma: note the perineural infiltration (same tumor as Fig. 26.64).

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Histologic variants of melanoma 1333

Fig. 26.64 
Nevoid melanoma: MIB-1 expression is usually brisk and
positively staining nuclei are commonly seen at all levels
of the tumor compared with banal nevi in which only very
A B rare positive cells are seen. (A) Nevoid melanoma; (B) banal
dermal nevus.

Fig. 26.65 
Nevoid melanoma: nuclear cyclin D1 expression is typically
brisk and present throughout the depth of the tumor
A B compared to a banal nevus where only scattered cells are
positive. (A) Nevoid melanoma; (B) banal nevus.

very similar lesions ‘papillomatous nevoid’ melanoma.16 Very occasionally, with melanoma cells when viewing MIB-1 stained sections. Double or dual
melanoma metastatic to the skin from another cutaneous site can present staining with MIB-1 and a melanocytic marker such as MART-1 can be
with a nevoid appearance.17 particularly valuable. Correlation with the hematoxylin and eosin stained
Immunohistochemistry may be of help in distinguishing between nevoid section is always advised. HMB-45 can also sometimes be useful. Dermal
melanoma and a banal nevus.18 In our experience, demonstration of ele- nevi are only positive in the most superficial part of the nevus as a counter-
vated proliferative activity using MIB-1 and sometimes cyclin D1 can be part to morphological maturation, whereas in melanoma positive cells may
particularly valuable (Figs 26.64 and 26.65). Banal nevi show only very be identified throughout the depth of the tumor.
scattered positive cells in the superficial part of the dermal component, FISH and comparative genomic hybridization (CGH) studies will often
whereas in melanoma they may be numerous and present throughout the show findings similar to conventional melanoma and thus are frequently
thickness of the lesion. It is important not to confuse positive lymphocytes helpful in the diagnosis of nevoid melanoma.5,19

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
1334 Melanoma

Fig. 26.66  Fig. 26.67 


Small cell melanoma: this variant of melanoma simulates type-B nevus cells at Small cell melanoma: high-power view. The tumor is commonly mistaken for a
low-power magnification, the tumor cells being small and deeply basophilic. lymphoma or neuroendocrine carcinoma if melanin pigment or junctional activity
is absent.

While one must always be wary of missing nevoid melanoma, it is also


important not to overdiagnose melanoma in nevi. This diagnostic reversal in the literature. This being said, age is a revealing variable with most
can also have untoward clinical and medicolegal consequences.20 Spitz-like features in young children being benign, but the same features in
adults, particularly older adults, are much more likely to be associated with
malignancy.5,6 There have thus been a number of papers describing melano-
Small cell melanoma
mas with features reminiscent of Spitz nevus (spitzoid melanoma, malignant
The term ‘small cell melanoma’ is often a source of confusion.1–3 In the lit- Spitz nevus) and diagnostic criteria to afford their distinction have been pro-
erature it has been applied to a variety of different lesions. It has been used posed and extensively discussed.7–20 Agreement between expert dermatopa-
as a synonym for nevoid melanoma as defined above.4,5 It has also been thologists in assessing malignancy in atypical spitzoid melanocytic tumors is
utilized in the context of a high-grade variant of childhood melanoma. Such quite poor.21 Agreement on what should be considered spitzoid or Spitz-like
tumors, which present on the scalp or developing in a congenital nevus, is also suboptimal.22 Given this uncertainty, treatment recommendations
are highly aggressive.6,7 Other authors have employed the term to describe from pathologists can show considerable variance.22
a tumor occurring in adults, which mimics lymphoma, cutaneous neuroen- Features that may favor a diagnosis of spitzoid melanoma include large
docrine carcinoma, or metastatic undifferentiated carcinoma.8–11 It has been size, deep dermal penetration, parakeratosis, epidermal atrophy (as opposed
used in the Australian and UK literature to describe a low-grade variant to the marked hyperplasia typical of Spitz nevus), asymmetry and lack of
of melanoma arising on the sun-damaged skin of middle-aged males.12,13 A circumscription, pagetoid spread at the edge of the lesion (pagetoid spread
recent study indicates that a small cell component in cutaneous melanoma in the center of the nevus is quite common in a Spitz nevus), absence of
is a independent poor prognostic indicator.14 Kamino (eosinophilic) bodies, dusty pigmentation, dermal nests larger than
In our view, small cell melanoma should be distinguished from nevoid the junctional ones, a diffuse (non-nested) dermal component, an expan-
melanoma. We reserve the term for a high-grade melanoma, presenting as sile rather than infiltrating lower border, lack of maturation, nuclear pleo-
a small blue cell tumor reminiscent of type-B nevus cells. It is characterized morphism and hyperchromatism, conspicuous mitoses, deep mitoses, and
by a monotonous population of cells with minimal cytoplasm and round atypical mitoses (Figs 26.68–26.70).7,9,15,23 In a meta-analysis, Walsh and
to oval hyperchromatic nuclei, often containing prominent nucleoli (Figs coworkers15 found the following to be the most useful and consistently
26.66 and 26.67). Mitoses are frequently conspicuous and karyorrhexis is applied criteria:
common. The tumor cells are sometimes aggregated into elongated expan- • symmetry,
sile nests but may also present as a diffusely sheeted infiltrate.15 Maturation • uniformity of nests from side to side,
with depth is not apparent. A pseudorosette pattern has been documented.15 • Kamino bodies,
In recurrent tumors where junctional activity is no longer apparent, and • brisk mitotic rate,
in metastatic disease, small cell melanoma can be easily confused with a • mitoses close to the base of the lesion,
poorly differentiated carcinoma, a lymphomatous infiltrate, and neuroen- • abnormal mitoses.
docrine carcinoma.8–10,16 Appropriate immunocytochemistry should readily A recent study by 13 experts of 75 cases confirmed the usefulness of all these
solve the problem. factors excepting Kamino bodies and adding ulceration as a feature favoring
malignancy.21 Thus the presence of the first three and absence of the second
three argue for a diagnosis of Spitz nevus and vice versa. Spatz and cowork-
Spitzoid melanoma
ers16 have devised a grading system for dividing atypical Spitz nevi into low-,
The diagnosis of Spitz nevus, and in particular its distinction from mela- medium-, and high-risk categories (of being frankly malignant). This is based
noma, is one of the most difficult areas in dermatopathology.1–3 Spitz nevus on age, diameter, involvement of subcutaneous fat, presence or absence of
is predominantly a lesion of children and young adults, whereas melanoma ulceration, and mitotic rate. The category of atypical Spitz with uncertain
occurs most often in the middle aged and elderly. There is therefore a ten- biological (or malignant) potential has also been much discussed.24,25 This
dency when encountering a spitzoid lesion in a child to automatically try to concept is often applied to spitzoid lesions as many diagnosticians find these
categorize it in the benign group merely on the basis of the age.4 Overreli- tumors challenging to definitively categorize as wholly benign or malignant
ance on age is a dangerous practice, since although rare, melanoma occur- in a subset of cases. Cases in this category may not represent neoplasms with
ring in childhood and showing spitzoid features has been well documented the full biological potential of standard melanoma; however, this category

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Histologic variants of melanoma 1335

Fig. 26.68 
Spitzoid melanoma: (A) low-power view showing deep extension into the
A subcutaneous fat; (B) the presence of ectatic vessels in the superficial dermis is
reminiscent of Spitz nevus.

Fig. 26.69 
A Spitzoid melanoma: (A) multiple Kamino bodies may also mislead the unwary;
(B) multiple mitoses, however, should raise a high index of suspicion.

may be tainted with Spitz nevi containing unusual features. The descriptions feature pointing to melanoma as opposed to Spitz nevus when present,
in the literature and use of these categories highlight the diagnostic difficul- though some question its usefulness.21,26,27 It is important to emphasize that
ties presented by Spitz nevi and spitzoid melanoma. this feature is useful when the thinning of the epidermis is continuous but
Mones and Ackerman20 have emphasized involvement of the deep dermis usually not when it is focal.
or subcutaneous fat by a vertically orientated tumor showing only mild Immunohistochemistry may be of value. Thus 50% or more of tumor
asymmetry on scanning magnification. Other clues include uneven melanin cells in spitzoid melanoma may express proliferating cell nuclear antigen
distribution, often at the base of the tumor, variability in size and shape (PCNA) whereas in Spitz nevus, usually less than 5% of cells are posi-
of the dermal nests and fascicles, confluence of nests and fascicles to form tive.28 A similar staining pattern is noted with MIB-1 where less than 2%
diffuse sheets of tumor cells, and diminution or loss of adnexae. They also strongly favors nevus and greater than 10% strongly favors melanoma with
make the point that the superficial features can be virtually indistinguish- others factors impinging more strongly between these two thresholds.29,30
able from Spitz nevus. Consumption of the overlying epidermis is a helpful HMB-45 expression in the deep dermal component would favor a diagnosis

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
1336 Melanoma

Fig. 26.70 
Spitzoid melanoma:
deep or abnormal Fig. 26.71 
forms are important Signet ring cell melanoma: very occasional signet ring melanoma cells are not
diagnostic features and uncommonly seen in epithelioid melanomas; exceptionally, however, they can
are absolute indicators of constitute the majority of the tumor.
an unequivocal diagnosis
of melanoma. Note that
there is no evidence of
maturation. change in recurrent and primary disease. The phenomenon does not have
prognostic significance.1–15
Signet ring cell change is characterized by a large pale or eosinophilic
cytoplasmic globular inclusion or vacuole, which compresses the nucleus to
of spitzoid melanoma.29,31 Amplification of the p-arm of chromosome 11 the edge of the cell (Fig. 26.71). This appearance may affect a melanoma
where the HRAS oncogene resides is seen in a subset of Spitz nevi; this in part or constitute the whole of the lesion. The periodic acid-Schiff (PAS)
change is not encountered in melanoma and thus this finding may be helpful stain is variably weakly positive (diastase resistant) but an Alcian blue reac-
in distinguishing Spitz nevi and melanoma.32–35 Comparative genomic tion is consistently negative.1,2,11 The nature of the PAS-positive material is
hybridization and FISH assays can be used to make this determination. The unknown. The signet ring appearance is due to excess intermediate filament
distinction between Spitz tumor and atypical Spitz tumor is not always well presenting as an intracytoplasmic ‘mass’ or vacuole.
informed by such assays, but can be helpful when characteristic melanoma The signet ring cells contain vimentin and may express S100 protein and
deficits are present.36,37 As for spitzoid melanoma, 6p25 and/or 11q13 gains HMB-45 although the latter two can be patchily distributed and occasion-
or 9p21 deletions on FISH favor malignancy while no FISH abnormality or ally absent altogether.2,4,16 Diagnosis in such a case is then dependent upon
6q23 deletions are associated with less aggressive behavior or benignancy.38 finding more typical melanoma cells with appropriate immunocytochemistry
Spitzoid melanomas generally lack mutations in genes such as BRAF and elsewhere in the specimen. Signet ring cell melanoma must be distinguished
NRAS common in some other melanoma subtypes in some studies, but not from other signet ring cell tumors, particularly mucin-containing adenocar-
others.39,40 This may illustrate the difficulty in histologically defining what cinoma and lymphoma. To this end, antibodies to LCA, keratin, EMA, and
amounts to a spitzoid melanoma versus a standard melanoma. Spitz lesions CEA should invariably be included in the immunocytochemistry panel.
can also show inactivation of the tumor suppressor BAP1 (BRAF V600 Cytological diagnosis of signet ring cell melanoma (by fine needle aspira-
mutations can also be seen with BAP1 loss) as well as rearrangements in tion and from a peritoneal effusion) has been documented.8,10
ALK, RET, ROS, BRAF, NTRK1, and RET. These features can be seen in Signet ring cells have also exceptionally been described in banal and
both benign and malignant Spitz tumors.38,41–48 Spitz melanocytic nevi.6,17,18
Although Smith and coworkers10 suggested that malignant Spitz nevus
might represent a tumor that rarely spread beyond the draining lymph
Rhabdoid melanoma
nodes, subsequent papers have suggested that the tumor is often no differ-
ent from any other melanoma and therefore has the same risk of systemic Rhabdoid melanoma shows considerable overlap with signet ring cell
spread and an identical mortality.15,28,49 Spitz-like lesions that metastasize melanoma.1
to lymph nodes in children do appear to be less aggressive than traditional The term ‘rhabdoid tumor’ was originally coined to describe an aggres-
melanoma in children and adults. Metastasis of Spitzoid lesions in adults sive childhood renal tumor, which was thought to represent a rhabdo-
appears to bear the same prognosis as that of other metastasizing mela- myosarcomatous variant of nephroblastoma.2 It was characterized by the
noma. Some authors recommend sentinel lymph node biopsy in problemati- presence of eosinophilic hyaline intracytoplasmic inclusions, later identified
cal spitzoid lesions to help establish a diagnosis of melanoma in addition to ultrastructurally as whorls of intermediate filaments. Extrarenal rhabdoid
adding prognostic information, though this approach is not without contro- tumors have been identified at a wide range of sites including soft tissues,
versy.19,50–56 Patients younger than 11 years of age with spitzoid melanoma skin, brain, orbit, uterus, bladder, prostate, and liver. A rhabdoid phenotype
may have a better prognosis than older adolescents and adults, but this was first described in metastatic melanoma.3–10 More recently, small numbers
finding has not been universal.49,56 of primary tumors showing rhabdoid features have been documented.11–17
At present, there are too few cases documented to determine whether the
rhabdoid phenotype in melanoma has any prognostic significance.
Signet ring cell melanoma
The rhabdoid phenotype is characterized by the presence of large epithe-
Signet ring cell melanoma is a very rare histologic variant, which shows lioid cells with abundant cytoplasm containing a large round eosinophilic
considerable overlap with rhabdoid variants (see below). It has been mostly hyaline inclusion (Fig. 26.72). The nuclei are typically vesicular and nucleoli
described in metastatic tumors. There are also reports of signet ring cell are eosinophilic and conspicuous. Extrarenal rhabdoid tumors are not a

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Histologic variants of melanoma 1337

Fig. 26.72  Fig. 26.74 


Rhabdoid melanoma: this variant often causes diagnostic difficulty since lesions Balloon cell melanoma: the tumor cells have clear or faintly granular cytoplasm.
are commonly amelanotic. Note the vesicular nuclei, prominent nucleoli, and large This example is amelanotic and could easily be mistaken for a xanthomatous
eosinophilic cytoplasmic inclusions. lesion.

Fig. 26.73  Fig. 26.75 


Rhabdoid melanoma: in the absence of a known history of melanoma, the Balloon cell melanoma: high-power view.
diagnosis commonly depends upon immunohistochemistry (S100 protein).

homogeneous entity; most often rhabdoid features are seen focally in an is more commonly seen than in classic melanoma.2 Prognosis is usually poor
otherwise recognizable and classifiable lesion, i.e., rhabdoid change rep- but this relates to the tumor thickness at presentation rather than any feature
resents a morphological endpoint in dedifferentiation. specific to this unusual melanoma variant. Balloon cell change appears to be
The results of immunocytochemistry are variable. Some tumors are more common in nevi than melanoma.6
characterized by S100 protein, SOX10, and HMB-45 expression (Fig. Balloon cells have abundant eosinophilic or clear cytoplasm showing fine
26.73).11,12 Mart-1 is also often expressed by tumor cells. In a number of granularity or vacuolation (Figs 26.74 and 26.75). The tumor cells are typ-
tumors, however, the rhabdoid cells fail to show any of the above and the ically S100 protein, Mart-1, SOX10, and HMB-45 positive. They contain
inclusions contain keratin, smooth muscle actin, or desmin.3,12 Rare exam- PAS-positive, diastase-resistant granules. These are also iron hematoxylin
ples of melanoma with rhabdoid morphology display rhabdomyosarcoma- positive, hyaluronidase resistant. Sensitivity to ribonuclease indicates that
tous differentiation with expression of desmin, myogenin, and MYOD1. the granules are composed of ribonucleoprotein.2 The cause of the vacuo-
Ultrastructurally, the inclusions have usually been found to consist of lation is uncertain but is believed to represent either abnormal melanosome
aggregates of intermediate filaments. In one series of metastatic melanoma, metabolism or else a melanosome degenerative phenomenon.
the rhabdoid inclusions were composed of tubular inclusions within dilated Balloon cell melanoma can be distinguished from balloon cell nevus
rough endoplasmic reticulum and mitochondria.5 by the absence of nuclear pleomorphism and mitotic activity in the latter.7
Balloon cell melanoma may also be confused with sebaceous and xantho-
matous tumors. Sebaceous lesions have a characteristic ‘bubbly’ cytoplasm
Balloon and clear cell melanoma
and often the nucleus is crenated. In addition, sebaceous tumors express
Balloon cell melanoma is a very rare vertical growth phase variant with less epithelial membrane antigen as well as adipophylin and are S100 protein
than 20 cases well documented in the medical literature.1–5 There are no par- and HMB-45 negative. Xanthomatous lesions contain lipid, express CD68,
ticular distinguishing clinical features except that a polypoid configuration and are S100 protein negative.7–10

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
1338 Melanoma

Fig. 26.76  Fig. 26.77 


Clear cell melanoma: this is an exceedingly rare variant of melanoma. In the Myxoid melanoma: this rare variant can be a source of considerable diagnostic
absence of a known history of melanoma or visible melanin pigment, the difficulty. Note the ‘undifferentiated’ irregular cells associated with an abundant
diagnosis depends upon immunohistochemistry. myxoid stroma.

Pure clear cell variants due to intracytoplasmic glycogen deposition are


very rare and may be encountered in primary tumors or metastatic deposits
(Fig. 26.76).5,11–16 They may be confused with clear cell carcinomas, includ-
ing clear cell squamous carcinoma, clear cell hidradenocarcinoma, and clear
cell metastases such as from clear cell carcinoma of the kidney. In cases
where there is real diagnostic difficulty, the use of immunocytochemistry for
melanocytic and keratin expression will resolve the issue.
Exceptionally, multiple small intracytoplasmic vacuoles can give rise to a
pseudolipoblast appearance with conspicuous nuclear scalloping.

Myxoid melanoma
Myxoid stromal change in melanoma is very rare and may be seen in primary,
recurrent, and metastatic disease.1–11 It is of no prognostic significance.4,8
Myxoid change is a feature of vertical growth phase melanoma. It may
be evident as a minor component of the tumor or be present throughout.
Tumors showing the latter can be a source of considerable diagnostic dif-
ficulty with epithelioid cases suggesting carcinoma and spindle cell cases
mimicking sarcoma. It has been described as a reactive change after pho-
Fig. 26.78 
totherapy.12,13 Typically, the tumor cells are smaller than those present in
Adenoid (pseudoglandular) melanoma: this variant also associated with excess
nonmyxoid areas and may be epithelioid, spindled, or stellate in appearance mucin deposition may easily be misdiagnosed as an acantholytic squamous
(Fig. 26.77). Melanin pigmentation is variable. The mucin, which presents carcinoma or adenocarcinoma if a junctional component or pigment is absent.
as basophilic ‘stringy’ material, is usually PAS negative, colloidal iron and Diagnosis often depends upon immunohistochemistry. By courtesy of R.
Alcian blue positive, and sensitive to hyaluronidase (consistent with stromal Margolis, MD, St Elizabeth’s Medical Center, Boston, USA.
mucin).8 Often the melanoma cells are present as discohesive clumps, cords,
and strands such that a pseudoglandular or ‘acantholytic’ appearance may
sometimes result (Figs 26.78 and 26.79). Occasionally, pseudoacinar dif-
Melanoma with neuroendocrine differentiation
ferentiation and intercellular molding reminiscent of adenocarcinoma are
features.8 Neuroendocrine differentiation has been rarely described in melanoma
Immunohistochemistry is typical for melanoma, the tumor cells being cases and sometimes in non-cutaneous cases.1–4 Mixed melanocytic and
invariably positive for S100 protein and usually positive for HMB-45.8 neuroendocrine tumors have also been described in the lung, thymus, and
HMB-45 negativity has, however, been documented.5 thyroid and challenge current classification schemes.5 Immunohistochem-
Myxoid change may sometimes be seen in the stroma of desmoplastic ical expression of chromogranin and synaptophysin, along with neurofil-
melanoma, but other areas more typical of conventional desmoplastic mel- ament protein, has been noted as has expression of CD56.3 Less specific
anoma are usually present. markers of neuroendocrine differentiation such as neuron-specific enolase
Myxoid melanoma may be confused with mucus-secreting adenocar- and perhaps CD56 expressed in isolation should be interpreted with caution
cinoma and myxoid malignant peripheral nerve sheath tumor (MPNST) when seen in melanoma as they may not imply such differentiation. Char-
or other sarcomas. The former may be excluded on the basis of negative acteristic melanocytic markers remain intact. While not clearly associated
keratin immunocytochemistry and absence of PAS-positive mucin. Myxoid with any difference in natural history, neuroendocrine differentiation can
malignant peripheral nerve sheath tumor usually shows much less strong be associated with diagnostic confusion, especially with presentation at sites
and only focal S100 protein expression, and HMB-45 is negative. In addi- where other neuroendocrine tumors can be seen such as in the sinonasal
tion, MPNST limited to the dermis is exceedingly rare. region.6 Further complicating the distinction is that melanoma exhibiting

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Histologic variants of melanoma 1339

Fig. 26.79  Fig. 26.80 


Adenoid (pseudoglandular) melanoma: high-power view showing undifferentiated Malignant blue nevus: note the heavily pigmented primary tumor associated with
tumor cells dispersed within abundant mucin. By courtesy of R. Margolis, MD, St multiple satellite lesions on this elderly patient’s forehead. By courtesy of the
Elizabeth’s Medical Center, Boston, USA. Institute of Dermatology, London, UK.

neuroendocrine differentiation appears to show small cell morphology.


Interestingly, carcinoid-like trabecular and pseudorosette patterns have
been noted in melanoma as well, but these cases lacked neuroendocrine
markers.7,8

Adenoid and pseudopapillary melanoma


This rare variant shows considerable overlap with myxoid melanoma.
Primary or metastatic deposits show extreme discohesion such that
pseudoglandular spaces sometimes with papillary architecture are formed
and sometimes these can be filled with mucin.1,2 Occasionally ‘intralumi-
nal’ papillary processes and ‘mucin’-filled lakes are formed, heightening the
resemblance to adenocarcinoma.3 A possibly related form has been described
as discohesive melanoma.4,5 In the latter, melanomas showed striking disco-
hesion of cells both in the intraepidermal and invasive component closely
mimicking an acantholytic disease.

Blue nevus-like melanoma (malignant blue nevus)


This term covers a variety of lesions including melanoma arising in a back-
ground of cellular blue nevus and to a lesser extent common blue nevus Fig. 26.81 
or showing morphological overlap with blue nevus.1–5 Variants have also Malignant blue nevus:
been documented complicating nevi of Ito and Ota and pilar neurocris- this tumor arose on the
tic hamartoma.6–8 It also includes cases of melanoma that show histologic scalp. There is a dense
overlap with cellular blue nevi but which are devoid of a precursor lesion expansile tumor nodule
(de novo variants).9–13 Pigmented epithelioid melanocytoma (previously which has extended into
termed animal-type or pigment-synthesizing melanoma; see below) can the subcutaneous fat.
have a highly similar morphological spectrum but are distinct from blue
nevus-like melanoma at the genomic level.14
epithelioid or spindled melanocytes showing a diffuse growth pattern and
Clinical features obvious cytological features of malignancy (Figs 26.83 and 26.84).
Malignant blue nevi are often very slowly growing lesions that commonly Tumors that mimic cellular blue nevus, but in which no precursor lesion
present as a consequence of a sudden onset of growth and show a predilec- can be identified, commonly show an expansile growth pattern with usually
tion for the scalp (Fig. 26.80). Males are affected more often than females. pushing borders extending into the subcutaneous fat (Fig. 26.85). Occasion-
No age group is immune and exceptionally children are affected. This is a ally a dumbbell scanning morphology may be evident and rarely an alveolar
high-grade lesion with outcome similar to stage-matched cases of traditional growth pattern is seen.11 Typically, at low-power magnification, a diagnosis
melanoma cases.15,16 The lung, liver, and lymph nodes are most commonly of benign cellular nevus is anticipated. The tumor cells most often have
affected by metastatic disease. spindled cell morphology but epithelioid and mixed variants are sometimes
encountered. High-power detail, however, shows an increased nuclear-to-cy-
Histologic features toplasmic ratio, mild to moderate nuclear pleomorphism, hyperchromatism,
Tumors that arise in a background of pre-existent blue nevus typically show nucleolar prominence, and increased mitotic activity (Figs 26.86 and 26.87).
an abrupt transition from a benign precursor lesion to obvious melanoma Necrosis, sometimes exhibiting a geographic pattern, may also be present
(Figs 26.81 and 26.82). The latter presents as one or more nodules of and perineural infiltration is an occasional feature.

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
1340 Melanoma

Fig. 26.82 
Malignant blue nevus:
a pilar blue nevus was
evident in the adjacent
skin.

Fig. 26.84 
Malignant blue nevus: (A) there are multiple dendritic cells; (B) note the two
mitoses.

Fig. 26.83 
Malignant blue nevus: the tumor cells have vesicular nuclei with prominent
nucleoli.

Scattered dendritic cells are often present in this second variant. Whether
this represents a residual benign precursor lesion or indicates malignant
dendritic cells is problematical. In favor of the latter possibility is the pres-
ence of dendritic cells in metastases (Fig. 26.88).
A subset of melanocytic proliferations is difficult to classify definitively
as benign or malignant. These have been referred to as atypical cellular blue
nevi or cellular blue melanocytic proliferation of uncertain malignant poten-
tial.1,17 There appears to be a lack of consensus regarding diagnostic criteria
for malignancy across the blue nevus spectrum.18
Both blue nevus and blue nevus-like melanoma are associated with muta-
tion in GNAQ or less commonly GNA11, encoding the alpha subunit of
a heterotrimeric G-protein.14,19 Similar to ocular melanoma, mutations in Fig. 26.85 
BAP1, SF3B1, and EIF1AX are commonly encountered in blue nevus-like Malignant blue nevus: a
melanoma and their presence may help to establish malignancy.14,20 The precursor lesion was not
standard multi-probe FISH assay can be helpful in distinguishing blue identified in this case.

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Histologic variants of melanoma 1341

Fig. 26.86 
Malignant blue nevus: note the central mitotic figure.

Fig. 26.88 
(A, B) Malignant blue nevus: the sentinel node contained metastatic melanoma. A
small number of dendritic cells are present. This is the same case as illustrated in
Figures 26.81–26.84.
Fig. 26.87 
Malignant blue nevus: dendritic cells are conspicuous in this field.
cases.14,15 Angiotropism can also be seen in nevi, particularly congenital
melanocytic nevi.16
nevus-like melanoma from atypical forms of cellular blue nevus, but limited Exceptionally, melanoma may show blood-filled dilated spaces lined
data are available.21,22 by endothelium-like tumor cells reminiscent of angiosarcoma (angioma-
toid melanoma).17–22 Intraluminal tufting may result in pseudoglomeruloid
structures.18
Angiotropic and angiomatoid
(pseudovascular) melanoma
Metaplastic melanoma (melanoma with
Angiotropism has been described in only a few cases of vertical growth
heterologous differentiation)
phase melanoma.1–5
Histologically, this rare manifestation of melanoma is characterized by Exceptionally, melanoma is associated with heterologous metaplastic ele-
the growth of melanoma cells within or along the walls of vessels, prin- ments, including bone and cartilage.1–6 Mostly, these have complicated acral
cipally veins, but usually without showing evidence of intravascular inva- lentiginous lesions (particularly arising in a subungual location) although
sion.1,6 While a relatively rare occurrence, interobserver concordance is high examples have also been described in primary desmoplastic melanoma,
when standardized diagnostic criteria are applied.7 More recently, this phe- mucosal melanoma, and melanoma arising in a congenital nevus.6,7 It can
nomenon has been suggested as a type of vascular invasion and shown to also be seen in metastatic melanoma, both with or without being present
be associated with a poor prognosis, perhaps even brain metastases in some in the cognate primary lesion.8,9 From the limited available literature, these
cases.8,9 It is also independently correlated with the presence of microsat- tumors are high grade (due to thickness at presentation) with frequent
ellites in primary cutaneous melanoma.10 Some contend that this pericytic metastases and considerable mortality. Histologically, they usually present
association may represent an extravascular mechanism for local or regional as sarcomatoid lesions with osteoid and to a lesser extent chondroblastic
metastasis.11,12 Angiotropism has also been described in epidermotropic differentiation (Figs 26.89 and 26.90). A number of these cases represent
and other metastatic melanomas.13 A recent mouse model study indicated osteosarcomatous differentiation, i.e., osteoid deposition in association with
that inflammation induced by UV exposure can promote angiotropism and malignant cells, and/or chondrosarcomatous differentiation.10,11 Too few
proposed this as a means of metastasis also relevant to humans in some cases have been reported to be certain whether heterologous differentiation

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
1342 Melanoma

Fig. 26.89  Fig. 26.91 


Metaplastic melanoma: multiple deposits of osteoid are present in association Metaplastic melanoma: this melanoma was associated with an intense osteoclast-
with malignant cells in this amelanotic melanoma. like giant cell infiltrate.

under the name of pigmented epithelioid melanocytoma. There is certainly


morphological overlap with malignant blue nevus, but genomically these
two entities are distinct and we believe that they represent independent his-
tologic entities based on this and differences in their natural history. The
original term of animal-type melanoma was coined to reflect the similarity
of these tumors to melanocytic lesions occurring in horses and a variety of
experimental animal models – so-called equine melanotic disease.1,2 This is
a condition in which aging gray horses lose their pigmentation and develop
melanocytic tumors particularly around the external genitalia, undersurface
of tail, breast, and lip mucosa. Subsequent development of melanoma in
these animals is an important complication.

Clinical features
Around 200 cases of this seemingly rare tumor have been documented,
mostly as single case reports.3 Although the age range is quite wide, the
majority of lesions have arisen in patients in the second to fourth decades.4–7
There is no recognized sex or site predilection. Most present as 1.0 cm or
more diameter, brown, blue-black or black nodules or plaques clinically
thought to represent melanoma.8 These tumors can occur sporadically or
Fig. 26.90  as part of the autosomal dominant Carney complex where these tumors
Metaplastic melanoma: high-power view. have traditionally been termed epithelioid blue nevus.9 Carney complex
consists of spotty skin pigmentation, and a variety of unusual endocrine
and non-endocrine tumors such as myxomas, pigmented nodular adreno-
influences prognosis. Certain properties of mesenchymal stem or precursor cortical disease, calcifying Sertoli cell tumor and psammomatous melanotic
cells suggest a pluripotency that includes neural crest, osteogenic, and chon- schwannomas. This complex is associated with germline inactivating muta-
drogenic potential and may help explain the existence of this melanoma tions in the protein kinase A regulatory subunit, PRKAR1A on chromo-
variant.12 Some cases have been shown to express immunohistochemical some 17q.10,11 The unrelated Carney triad is a distinct condition comprised
markers of master regulatory genes for osseous and cartilaginous differenti- of gastrointestinal stromal tumor, pulmonary chondroma, and extra-adrenal
ation such as SATB2 and SOX9.6,9 paraganglioma driven by succinate dehydrogenase (SDH) definiciency.12,13
Unusually, melanoma may undergo smooth muscle and rhabdomyoblas- Prognosis seems to be better than that of an ordinary melanoma of
tic differentiation.13–16 Case reports of melanoma showing ganglionic and equivalent thickness and it has been proposed that it may be regarded as
ganglioneuroblastic differentiation have been documented.17–19 a low-grade variant of melanoma. A recent meta-analysis of around 190
There are very occasional reports of conventional and desmoplastic mel- cases seems to confirm this notion, but that study was hampered by limited
anoma containing osteoclast-like giant cells (Fig. 26.91).20–25 follow-up for the reported cases and the lack of uniform diagnostic crite-
Rhabdomyosarcoma has been documented in a congenital nevus, includ- ria.3 Nonetheless, this study showed that with a median Breslow thickness
ing a giant form.26–28 of 3.8 mm and 16% ulceration, there were only six patients with distant
metastases and five patient deaths. Sentinel lymph node positivity rates were
notable at 41% of the 78 patients undergoing this procedure, but subse-
Pigmented epithelioid melanocytoma (pigment
quent progression was uncommon. Thus this tumor has a markedly supe-
synthesizing melanoma; animal-type melanoma)
rior prognosis to conventional melanoma.
An enigmatic lesion with many names, including pigment synthesizing mela-
noma, animal-type melanoma, melanoma with prominent pigment synthesis, Histologic features
equine type melanoma, and low-grade hypermelanotic dermal melanoma, Pigmented epithelioid melanocytoma is characterized histologically by a
this exceedingly rare variant of melanoma is now recognized by the WHO very dense infiltrate of heavily pigmented melanocytes, typically filling the

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Histologic variants of melanoma 1343

Fig. 26.92  Fig. 26.94 


Pigment synthesizing melanoma: this low-power view shows the intense Pigment synthesizing melanoma: the tumor cells are uniform with large vesicular
pigmentation typical of this variant. nuclei and prominent eosinophilic nucleoli. Abundant pigment-laden macrophages
are present.

tumors previously termed pigment synthesizing melanoma.14–16 While these


lesions appear to have metastatic potential to involve lymph nodes, their
outcome is favorable compared to stage-matched cases of traditional mela-
noma.3 More and longer-term study of this entity and the above-mentioned
spectrum will be insightful.
Loss-of-function mutations in PRKAR1A are common. Gene fusions
with PRKCA encoding protein kinase Cα can also be seen in pure mela-
noma.17 In cases of combined nevi with conventional features and an area
resembling pigmented epithelioid melanoma, BRAF V600 mutations can
be seen with the PRKAR1A mutations. Cases with characteristic activating
GNAQ or GNA11 mutations would be better regarded as blue nevus or
blue nevus-like melanoma rather than pigmented epithelioid melanocytoma.
Use of the standard multi-color FISH assay to assess for malignancy in
pigmented epithelioid melanocytoma has been reported as helpful in a few
cases, but should be interpreted with caution given the alternative genetics
of this tumor.18,19

Fig. 26.93  Epidermotropic metastatic melanoma


Pigment synthesizing melanoma: scattered tumor cells are present in the lower
Metastatic melanoma is common and, in the majority of cases, causes
epidermis.
little diagnostic difficulty. Rarely, however, cutaneous deposits are accom-
panied by epidermal involvement such that distinction from a second or
papillary and reticular dermis and often spilling over into the subcutaneous third primary tumor can be extremely difficult, if not impossible, particu-
fat (Fig. 26.92). The epidermis may be atrophic or hyperplastic. A small larly if clinical information is not available (Figs 26.95 and 26.96).1–9 Tra-
number of cases are not purely intradermal but display a junctional com- ditionally, the features in favor of epidermotropic metastatic disease have
ponent (Fig. 26.93). In cases with a junctional component, tumor cells are included a well-circumscribed dermal nodule, widespread lymphovascular
plump and dendritic and do not tend to exhibit pagetoid spread. Tumor invasion, and filling of the papillary dermis by melanoma cells with atrophy
cells are spindled, polygonal, or rounded and are distributed in a fascicular of the overlying epidermis.1,10 Typically, the epidermal component is equal
or nodular growth pattern, sometimes displaying a focal storiform appear- in width to, or is less than, the dermal component. A lateral epidermal
ance.6 Dendritic processes may be identified. The tumor cell cytoplasm typ- collarette may sometimes be present. Exceptionally, patients have presented
ically contains abundant fine to coarse melanin granules, which commonly with showers of metastases which, due to extensive lateral spread of the epi-
obscure the nuclear morphology. Examination of bleached sections can be dermal component beyond the dermal mass or even wholly intraepidermal
helpful. The cytoplasm of tumor cells is pale, and nuclei appear enlarged and disease, have histologically been indistinguishable from a primary tumor. In
sometimes hyperchromatic. A single prominent basophilic nucleolus is often such instances, clinicopathological correlation is essential in establishing a
seen. Although there is cytological atypia, tumor cells display little variation diagnosis of metastatic disease.2,3 A recent study with molecular testing sug-
throughout the tumor (Fig. 26.94).6 Mitoses are typically sparse. The latter gests that the genetic relationship of primary melanomas and their potential
and the uniformity of tumor cells allow distinction from malignant blue epidermotropic metastases is complex, with certain cases showing divergent
nevus. Perineural infiltration may be present but lymphovascular invasion loss of heterozygosity and X-chromosome inactivation patterns consistent
has not been described. with either complex divergent clones or possibly new primaries.11 Addi-
This is an unusual tumor, which shows some histologic overlap with tional study is essential as the distinction has important staging implica-
other dermal dendrocytoses and blue nevus-like melanoma. These lesions tions. Epidermotropic melanoma is a satellite lesion that results in stage III
form part of the spectrum of pigmented epithelioid melanocytoma, which groupings under the 8th edition AJCC, whereas a second primary would be
includes the epithelioid blue nevus associated with the Carney complex and individually staged.12

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
1344 Melanoma

Fig. 26.95  Fig. 26.96 


Epidermotropic metastatic melanoma: numerous tiny blue tumor deposits are Epidermotropic metastatic melanoma: this metastatic deposit is wholly
present in association with the scar from a previously excised melanoma. By intraepidermal and is indistinguishable from in situ superficial spreading
courtesy of J. Gershewald, MD, MD Anderson Cancer Center, Houston, Texas, melanoma. Diagnosis is entirely dependent upon the clinical history.
USA.

Fig. 26.97 
Blue nevus-like metastatic melanoma: (A) at low-power examination, the
features could easily be mistaken for a common blue nevus; (B) in addition to
pigment-laden melanophages and dendritic cells, there are scattered melanoma
A cells. Note the multinucleate form with vesicular nuclei and prominent nucleoli.
The patient had a melanoma previously excised at this site.

Cytogenetic studies by FISH may be of help in establishing the distinction.


Blue nevus-like metastatic melanoma
In a study comparing epithelioid blue nevi with blue nevus-like cutaneous
Blue nevus-like metastatic melanoma is rare but represents a very serious metastasis, it was shown that most of the latter had important copy number
source of misdiagnosis.1–3 One or multiple lesions may be seen and they changes in chromosomes 6p25, 11q13, and centromere 6 while the former
usually, but not always, develop at sites close to the primary tumor. Very lacked cytogenetic changes.2 Uveal melanoma can also give rise to blue
rarely, lesions may be distant. They resemble a blue nevus clinically and nevus-like melanoma which can be detected by detection of genetic events
histologically. They are often small and microscopically are characterized such as monosomy for chromosome 3 resulting in loss of BAP1.4 Detection
by dendritic, deeply pigmented melanocytes. Cytological atypia can be of the GNAS or GNA11 mutation of uveal melanoma is not helpful alone
very subtle and this is further compounded by the prominent pigmentation in this context since these mutations can also be seen in blue nevi.
(Fig. 26.97). Bleaching can therefore be useful. Resemblance to ordinary
blue nevus or epithelioid blue nevus may be seen. Clues to the diagnosis
Desmoplastic and neurotropic melanoma
of malignancy reside in the presence of cytological atypia, mitotic figures,
and inflammation mainly at the periphery of the lesion. Histologic diagno- Desmoplastic and neurotropic variants of spindled cell melanoma are
sis is extremely difficult and close clinicopathological correlation is crucial. inter-related high-grade tumors that are commonly associated with histologic

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Desmoplastic and neurotropic melanoma 1345

Fig. 26.98  Fig. 26.99 


Desmoplastic melanoma: these tumors are frequently amelanotic, as seen in this Desmoplastic melanoma: this tumor was initially thought to represent a cyst. By
patient. By courtesy E. Wilson Jones, MD, Institute of Dermatology, London, UK. courtesy of E. Wilson Jones, MD, Institute of Dermatology, London, UK.

diagnostic difficulty.1–7 A history of a mistaken diagnosis of reactive fibro- disease.3,24–30 Others have not observed this phenomenon as starkly, but this
blastic proliferative lesion is common and relates particularly to the usual could be due to differences in diagnostic criteria.31–34 In this regard it is
absence of melanin pigment and the relative infrequency of obvious epider- important to define desmoplastic melanoma carefully with the pure desmo-
mal melanocytic involvement, particularly in recurrent lesions. Desmoplastic plastic pattern being the predominant one, as cases showing a significant
melanoma represents an extreme degree of fibroblastic or myofibroblastic component (greater than 10%) of traditional melanoma show the more
metaplasia accompanied by abundant collagen synthesis. In the neurotropic characteristic metastasis to lymph node.35–37 Pure desmoplastic melanoma
variant, the metaplasia may be towards Schwann cell-type differentiation. appears to behave more like a sarcoma in terms of local recurrence and
Although not all desmoplastic melanomas are neurotropic and not all neu- metastatic pattern with the lung being the most common initial site rather
rotropic melanomas are desmoplastic, many authors regard them as vari- than regional lymph nodes. In regard to disease-specific survival, desmoplas-
ations on a theme.8 These tumors are in general associated with a high tic melanoma may behave similarly to classic melanoma, the poor progno-
incidence of recurrence and metastasis and a poor prognosis. sis reflecting an increased thickness of greater than 1.5 mm at presentation
in the vast majority of patients.3,38 Others have found that disease-specific
Clinical features survival for desmoplastic melanoma is superior to conventional mela-
Although the tumor may present at any age, the majority arise in the elderly noma.28 Pure and mixed variants of desmoplastic melanoma are reported
(mean age 61 years) and show a male preponderance (as high as 2.7 : 1), by some to have similar survival.39 There is some debate on this topic, with
which is particularly evident with the frankly neurotropic lesions.3,9–17 The some data that indicate an improved outcome for desmoplastic melanoma
head and neck are most frequently affected, but examples have been doc- patients.28,40–42 Poor prognostic indicators are high mitotic rate, tumor thick-
umented at a wide range of sites, including the trunk and upper or lower ness, ulceration, and inadequate excision margins (< 1.0  cm).3,17,23,43 The
limbs.9,18 The leg is particularly involved in females.3 Although the majority 5-year survival rate is of the order of 70–80%, and the overall mortality
of these neoplasms arise on sun-damaged skin in a background of lentigo varies from 11% to 66%.3,15,23
maligna (melanoma), superficial spreading and acral lentiginous melano-
mas, including subungual variants, have occasionally been documented.12,13 Histologic features
Desmoplastic melanoma has also been described on the palate, gingiva, lip, Desmoplastic melanoma is characterized by a diffusely infiltrative, some-
vulva, anus, and conjunctiva.19 Although a superimposed pigmented lesion times paucicellular, malignant spindle cell tumor with marked interstitial
of lentigo maligna may draw attention to this tumor, more often they present fibrosis and collagenization (Figs 26.100 and 26.101).1,2,9,19,44,45 It is to be
as amelanotic, flesh-colored or erythematous nodules or indurated plaques distinguished from the more common spindled cell melanoma unassociated
(Figs 26.98 and 26.99). These tumors are notoriously deeply infiltrative and with significant desmoplasia.
commonly have extended widely by the time of diagnosis. In addition to The infiltrate often leaves the immediate subepidermal papillary dermis
contributing to the high incidence of recurrence, neurotropism may result in unaffected, but frequently is found to extend into the subcutaneous fat or
peripheral and cranial neuropathy. Spread along the cranial nerves into the beyond at the time of diagnosis. Involvement of skeletal muscle or underly-
base of the skull with eventual meningeal involvement is a rare but import- ing bone is not uncommon. The tumor cytology is variable, the cells resem-
ant complication with an almost 100% mortality.18,20 Although most des- bling fibroblasts, smooth muscle cells, or Schwann cells. They are typically
moplastic melanomas arise in a background of severe sun damage, previous elongated and have eosinophilic or more commonly basophilic cytoplasm
irradiation (therapeutic or otherwise) is occasionally of etiological impor- (Fig. 26.102). Nuclei may be tapered and hyperchromatic or cigar-shaped
tance.9 Rarely, examples have complicated congenital melanocytic nevi and and vesicular with prominent eosinophilic nucleoli (Figs 26.103 and
even chronic burns scarring.21,22 26.104). Mitoses are scanty but may be conspicuous, and sometimes abnor-
Recurrence is common (range 22–77%, mean 46%) and metastasis mal forms are present (Fig. 26.105). Most commonly, the tumor has a dis-
(particularly to the lungs) frequently supervenes (range 11–56%, mean of tinctly fascicular arrangement, but focal storiform areas are occasionally
approximately 30%).10,15 Local control in the head and neck region, where evident, which can result in a mistaken diagnosis of dermatofibroma or der-
many of these tumors arise, is particularly problematic.23 Careful evalua- matofibrosarcoma protuberans. Foci of myxoid change, giving the tumor a
tion of surgical margins is critical for local control.17 Surprisingly, nodal feathery appearance, are sometimes seen. Recurrent tumors are frequently
spread has been documented very infrequently such that sentinel lymph paucicellular and are easily taken for scar tissue (Figs 26.106 and 26.107).
node biopsy is not regarded by some as an effective staging tool in this Tumor giant cells are an infrequent manifestation. Lymphocytic infiltrates,

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
1346 Melanoma

Fig. 26.100 
Desmoplastic melanoma: Fig. 26.102 
low-power view showing Desmoplastic melanoma: the tumor cells have basophilic cytoplasm and are
a spindle cell tumor dispersed in a densely collagenous stroma.
extending throughout
the full thickness of the
dermis and associated
with superficial lymphoid
infiltrates.

Fig. 26.103 
Desmoplastic melanoma: nuclei are vesicular and nucleoli are prominent.

Fig. 26.101 
Desmoplastic melanoma: the overlying epidermis shows an atypical lentiginous
melanocytic proliferation.

often evident as nodular aggregates, are a characteristic (but not diagnostic)


feature (Fig. 26.108). They are, however, a useful histologic pointer in early
lesions.46,47 Vascular invasion is sometimes present.
Careful scrutiny of the overlying epidermis not uncommonly shows fea-
tures of atypical melanocytic hyperplasia, most often of the lentigo maligna
pattern.9 Very occasionally, the changes of superficial spreading melanoma
are present. In a significant proportion of cases no such in situ change is
detected (the so-called de novo variant) (Fig. 26.109). Whether this rep-
resents regression, inadequate sampling, a de novo dermal tumor, or a previ-
ously treated in situ component is uncertain. In most examples, the invasive
tumor cell population is amelanotic.9
Vascular changes including hemangiopericytomatous features and glo-
meruloid vascular lesions may rarely be encountered.48 Fig. 26.104 
The neurotropic variant of melanoma may show a range of manifesta- Desmoplastic melanoma: the tumor cells are compressed by the adjacent
tions.2,9,19,48 Microscopic perineural or endoneural involvement is common sclerotic fibrous stroma.

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Desmoplastic and neurotropic melanoma 1347

Fig. 26.105 
Desmoplastic melanoma: note the central mitotic figure.

Fig. 26.106  Fig. 26.107 


(A, B) Desmoplastic Desmoplastic melanoma:
melanoma: this example (A) high-power view
represents recurrent of tumor shown in
tumor following a previous Figure 26.106; (B)
excision. Melanoma can immunohistochemistry for
easily be mistaken for S100 protein highlights
scar tissue, particularly if B the considerable tumor
the clinical history is not cell population.
available. The lymphoid
aggregates should
A raise the suspicion of
desmoplastic melanoma.

Fig. 26.108 
B Desmoplastic melanoma: nodular aggregates of lymphocytes are a useful
diagnostic clue.

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
1348 Melanoma

Fig. 26.109 
Desmoplastic melanoma: Fig. 26.111 
in this example, the Desmoplastic melanoma: epithelioid cells are sometimes encountered within the
overlying epidermis tumor. This example shows extensive peri- and intraneural spread.
shows no evidence of
a pre-existent atypical
melanocytic proliferative
lesion (the so-called de
novo variant).

Fig. 26.112 
Desmoplastic melanoma (neural transformation): low-power view of a tumor
which has infiltrated through the full thickness of the dermis.

Fig. 26.110 
Desmoplastic melanoma: perineural infiltration is very commonly present,
accounting in part for the high recurrence rate of this tumor.

in many desmoplastic melanomas (Figs 26.110 and 26.111). The term neu-
rotropic, however, is often reserved for lesions where nerve involvement
is very marked or more particularly when it results in clinical evidence of
nerve irregularity and thickening. In rare examples, the tumor appears to be
limited to a nerve trunk with no discernible spread to the adjacent tissues.9
Occasionally, the spindled cell population displays histologic features rem-
iniscent of neurofibroma, schwannoma, or malignant nerve sheath tumor
(sometimes known as neural transformation) (Figs 26.112–26.114).18,49
These latter features often comprise smaller cells, with pale eosinophilic
cytoplasm and irregular wavy nuclei separated by variable amounts of
collagen and ground substance and sometimes giving the tumor a loose
myxoid appearance. Mitotic activity is variable and therefore these vari- Fig. 26.113 
ants can sometimes be very deceptive, resulting in a mistaken diagnosis Desmoplastic melanoma (neural transformation): the tumor cells have small,
of benign nerve sheath tumor. Recently, neurotropic melanoma associated twisted, and comma-shaped nuclei with ill-defined eosinophilic cytoplasm
with profound vascular infiltration (so-called angiotropic melanoma) has reminiscent of peripheral nerve sheath tumor. Note the nuclear hyperchromatism.

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Desmoplastic and neurotropic melanoma 1349

positivity was documented in the earlier literature, more recent studies have
not confirmed this observation.70,71 Loss of p16 reactivity can be useful in
distinguishing desmoplastic melanoma from desmoplastic nevus when this
differential diagnosis arises in small biopsies, but not all desmoplastic cases
show loss.72,73 P75 nerve growth factor receptor,WT1, and nestin immuno-
histochemical markers have also been suggested to have clinical utility.74–76
Desmoplastic melanoma has been described to MPNST. Loss of H3K27
nuclear staining characteristic of MPNST is not helpful for distinguishing
MPNST from melanoma, desmoplastic, or otherwise, as a subset of melano-
mas also show nuclear loss of this histone trimethylation marker.77
Evaluation of scar versus desmoplastic melanoma can be difficult as
the degree of fibrosis in desmoplastic melanoma can be extensive with
only scattered melanoma cells. This same problem complicates the assess-
ment of margins. Use of polyclonal S100 protein immunohistochemistry in
the setting of re-excision can be complex given the focal S100 reactivity
present in many scars due to scattered macrophages and other cells showing
Schwannian differentiation.78 Careful assessment of the degree of nuclear
hyperchromasia in these spindled cells is paramount in evaluating these
situations.79
Fig. 26.114  Metastases may be desmoplastic, neuroid, or more obviously melano-
Desmoplastic melanoma: the adjacent epidermis shows atypical melanocytic cytic. Although some authors have suggested that the desmoplasia is a
lentiginous hyperplasia.
consequence of reactive stromal fibroblast hyperplasia, it is now generally
accepted that these features are of melanocytic derivation.
Ultrastructural studies have shown that the tumor spindled cells may
rarely contain premelanosomes and melanosomes.1,2 Fibroblastic, myofibro-
blastic, and Schwann cell differentiation, particularly in neurotropic variants
(including elongated and interdigitating cellular processes sometimes encir-
cling collagen fibers reminiscent of mesaxon formation, discontinuous basal
lamina, and intercellular junctions) have also been documented.19,71,80–83
The genome of desmoplastic melanoma harbors approximately an order
of magnitude more mutations than that seen in conventional cutaneous mel-
anoma.84 Most of these are UV-linked. Exome sequencing revealed recur-
rent NFKBIE promoter mutations, and activation of the MAP kinase and
PI-3 kinase pathways through mutations in genes such as NF1, MAP2K1,
MAP3K1, EGFR, MET, RAC1, and PIK3CA.84–88 Mutations in BRAF V600
and NRAS G61 were not seen. TERT promoter mutations are common as
well.89 Standard multi-probe FISH can be useful to demonstrate desmoplas-
tic melanoma over other mimics.90

Differential diagnosis
The diagnosis of desmoplastic melanoma is frequently missed, particularly
if only superficial biopsies are available for study. The lesion is commonly
misdiagnosed as a reactive process, such as scarring and superficial nodular
Fig. 26.115 
fasciitis, and deeper lesions may be mistaken for a fibromatosis or even
Desmoplastic melanoma: the tumor cells are strongly S100 protein positive.
fibrosarcoma. Examples showing storiform morphology have sometimes
been confused with atypical fibrous histiocytoma and dermatofibrosarcoma
protuberans. Scar tissue may be a particular problem, especially in recurrent
been documented.50 Metaplastic bone formation has been described albeit lesions.91,92 Pointers towards the latter include horizontal orientation of the
rarely.51 Sarcomatoid dedifferentiation is also rarely noted.49 fibroblast population, absence of adnexae, loss of the rete-ridge pattern, and
Immunohistochemically, desmoplastic melanoma expresses S100 protein vertically orientated dermal blood vessels. Immature scar tissue, however,
(94–100%), neuron-specific enolase, and vimentin (Fig. 26.115).3,36–38,40,41 can be much more problematical and, in the case of re-excision specimens
HMB-45 (gp100) may be positive in the superficial papillary dermal tumor or recurrences, the features may represent an admixture of both.92 Recur-
cells but more often is totally negative.52–57 The results with MART-1 rent lesions following surgery may sometimes be associated with marked
(melan-A) are variable (24–60% positive), and tyrosinase does not appear nerve proliferation in addition to scar tissue. SOX10 and S100 may display
to be present in desmoplastic melanoma.58–60 Microphthalmia transcription positivity in scars and although this positivity can be strong it is usually
factor (D5) is of limited value, expression having been documented in from restricted to a small number of cells.93
only 35% to 55% of cases.61,62 A relatively new marker, SOX10, is reactive Diagnosis is dependent upon an awareness of the condition and often
in melanoma, desmoplastic melanoma, and peripheral nerve sheath tumors requires the inclusion of a battery of immunohistochemical markers to
and can be a very useful marker.63–66 Members of the S100 protein family establish the histogenesis of the tumor infiltrate. In many instances, however,
such as S100A6 may have efficacy in distinguishing desmoplastic melanoma careful scrutiny of the epidermis to detect atypical melanocytic hyperplasia
from malignant peripheral nerve sheath tumors, but the diffuse pattern of combined with the use of antibodies to S100 protein and pankeratin (to
traditional polyclonal S100 protein expression in melanoma is usually suffi- exclude desmoplastic spindled cell squamous carcinoma) will be sufficient
cient in this regard.67 Melanoma cell adhesion molecule may be more useful, to establish the correct diagnosis. Occasionally, in recurrent disease, pro-
82% of tumors in a series of 17 tumors having shown strong expression.68 liferating neural elements may be admixed with scar and residual tumor
EMA may be present in up to 43% of tumors but keratin, leu 7, CD31, and with resultant increased diagnostic difficulty. Neurofilament protein, which
CD34 are uniformly negative.48,55,56,68 Smooth muscle actin is often positive, is expressed by the neural elements but not the melanomatous component,
reflecting the myofibroblastic population.55,56,69 Although occasional desmin can often be of discriminatory value.

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
1350 Melanoma

Melanoma in children

Clinical features
Melanoma is extremely rare in children; less than 1% of all melanomas
present in childhood.1–20 Historically, many so-called examples have likely
represented misdiagnosed Spitz nevi.13 As a consequence, pathologists and
clinicians are extremely reluctant to make the diagnosis, with resultant delay
in treatment and potentially devastating consequences. Although excep-
tionally the tumor may develop in utero (complicating a large congenital
nevus or arising de novo) or be acquired transplacentally (from a maternal
melanoma), the majority are acquired and may be related to sun exposure
during childhood.4,14,21 Predisposing conditions include xeroderma pigmen-
tosum, dysplastic nevi, familial melanoma, melanoma-pancreatic cancer
syndrome, giant congenital nevi, neurocutaneous melanosis (leptomeningeal
melanoma), prior irradiation, Li-Fraumeni syndrome, and immunodefi-
ciency.3,4,14,22–27 Melanoma arises in up to 12% of giant congenital nevi (i.e.,
those that measure 20 cm or more in diameter) and this can occur during
childhood or later adult life.28–30 It has been suggested that childhood mel-
anoma can be segregated into three distinct categories based on the age of
diagnosis: congenital melanoma which occurs in utero up to birth; infan-
tile melanoma that presents after birth up to 1 year of age; and childhood
melanoma that occurs after 1 year of age to puberty.31 Some include in the Fig. 26.116 
Childhood melanoma:
childhood melanoma category patients up to the age of 18, but such upper
example of melanoma
age ranges are clearly arbitrary.32 which has arisen in a
The sexes are affected equally and tumors are more common in the giant congenital nevus.
second than the first decade.9 The majority of childhood melanomas have
developed in Caucasians.7 Lesions arise on the trunk and extremities, with
only 20% affecting the head and neck.5 Many childhood melanomas are not
easily recognized clinically.13,33 Sometimes, however, clinical features similar
to those described in adults – including increase in size, change in color, and
onset of bleeding – may be encountered.14,17
Some earlier reports suggested that the prognosis of melanoma in chil-
dren might be favorable.34 In our experience, however, childhood tumors
behave no differently from adult ones with the possible exception of spit-
zoid melanomas in very young patients as discussed further below. This is
also generally borne out in the more recent literature.4–6,13,18,35–37 Overall,
childhood melanoma should therefore be treated in much the same way
as adult tumors and, when appropriate, sentinel lymph node mapping and
biopsy are recommended.4,6,13,38–40 The majority of children present with
stage 1 disease with a 5-year disease-free survival rate of 77%.4,7 Crude
survival figures for the largest series reported to date are 71% overall, 64%
males, 81% females.9 Some, but clearly not all or even most in some series,
melanomas of childhood exhibit Spitz-like histology.41 These cases may have
a more favorable outcome than in adults, particularly in patients aged 11 or
less, but this finding is not universal.41–44 This could be due to inappropriate
assignment of Spitz nevi, albeit atypical, to the melanoma group; however,
even such tumors metastatic to lymph nodes appear to have a better prog-
nosis.45,46 Clearly, more study is required. Nonetheless, mortality is encoun-
tered within this age group. Spitzoid melanoma is discussed in more detail Fig. 26.117 
in the section above. Childhood melanoma: in this view, pleomorphic tumor cells are seen on the left;
nevus is present on the right.
Histologic features
Melanoma in children may arise within the context of a precursor lesion
such as a banal, congenital, or dysplastic nevus or it may present de novo.
Melanoma arising within a giant congenital nevus presents most often as a TERT promoter mutations are common, excepting the cases arising in con-
dermal, sharply delineated nodule composed of tumor cells showing obvious genital nevi.49
cytological features of malignancy (Figs 26.116–26.118).30 Although all the As with adult melanoma, the most important prognostic indicators are
common subtypes may be encountered, superficial spreading and nodular Breslow thickness and presence of ulceration. Melanoma in children should
melanoma variants are most frequently seen (Figs 26.119–26.123). Partic- be reported in exactly the same way as adult melanoma.51
ular subtypes that often cause diagnostic difficulty include small cell mel-
anoma, spitzoid melanoma, and malignant blue nevus.7,11,47 These are all Differential diagnosis
described under variants of melanoma (see above). Clinicopathological correlation is of great importance when considering a
Cytogenetic features detected by FISH are similar to those seen in adult diagnosis of melanoma in a child, particularly in the first few years of life.
superficial spreading melanoma.48 BRAF mutations can be seen in free- Significant cytological atypia affecting junctional and dermal components
standing cases while NRAS mutations are seen in the cases associated with accompanied by pagetoid spread and mitotic activity is not uncommon in
congenital nevi.49,50 UV damage signatures are commonly encountered and congenital nevi, especially those affecting neonates.

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Basomelanocytic tumor 1351

Fig. 26.118 
Childhood melanoma: in the center of the field is an atypical mitotic figure. Fig. 26.120 
Childhood melanoma:
there is surface
ulceration. By courtesy of
M. Little, MD, University
College Hospital, Galway,
Ireland.

Fig. 26.119 
Childhood melanoma: this tumor, which presented as a nodular lesion, arose in
a girl aged 13 years. By courtesy of M. Little, MD, University College Hospital,
Galway, Ireland.

Fig. 26.121 
Dermal squamomelanocytic tumor Childhood melanoma:
there is no maturation
Clinical features with depth. Note the
sharply delineated lower
Reports of this tumor are exceedingly rare in the pathology literature.1–6
border. By courtesy of
Patients present with brown or purple-black facial nodules measuring up M. Little, MD, University
to 1.0 cm in diameter. The age range documented is 32–94 years and there College Hospital, Galway,
have been less than 15 cases reported with equal sex distribution.5 A pre- Ireland.
cursor lentigo maligna has been documented in one case.1 The relatively
short follow-up information (mean 2.7 years) has shown no evidence of
recurrence or metastasis.2 A recently reported ‘metastatic’ case purportedly 26.126).2 Matrical differentiation has also been described.5 The two popu-
showing micrometastasis to a sentinel lymph node has an appearance much lations are distinctive morphologically, but the distinction is highlighted by
more characteristic of a capsular nevus.6,7 S100 protein or other melanocytic markers and cytokeratin immunohisto-
chemistry (Fig. 26.127).2
Histologic features
Although a precursor lesion or origin from the epidermis may be identifi-
able, the tumor may present as an upper dermal nodule independent of any Basomelanocytic tumor
epidermal connection (Fig. 26.124).2 A case limited to the epidermis has
also been described.3 It consists of a sharply delineated, lobular tumor com- Clinical features
posed of intimately admixed malignant squamous epithelium and a usually Basomelanocytic tumor is a very rare biphasic neoplasm similar, or
less prominent population of malignant melanocytes (Figs 26.125 and perhaps related, to the dermal squamomelanocytic tumor described above.1

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
1352 Melanoma

Fig. 26.122  Fig. 26.125 


Childhood melanoma: the presence of expansile nodules is an important Dermal squamoproliferative tumor: in this field, the tumor is composed of variably
diagnostic clue. By courtesy of M. Little, MD, University College Hospital, Galway, pigmented cells with irregular hyperchromatic nuclei. By courtesy of S. Poole,
Ireland. MD, Beth Israel Deaconess Medical Center, Boston, USA.

Fig. 26.123 
Childhood melanoma: multiple dermal mitoses are present in this field. By
courtesy of M. Little, MD, University College Hospital, Galway, Ireland. Fig. 26.126 
Dermal squamoproliferative tumor: focal squamous differentiation is evident. By
courtesy of S. Poole, MD, Beth Israel Deaconess Medical Center, Boston, USA.

However, basomelanocytic tumor is a combination of basaloid carcinoma


and melanoma.2–4 As would be expected, the melanocytic component of the
tumor appears to be the more aggressive component and has been reported
to metastasize with fatal outcome.3

Histologic features
In this combined tumor, the basaloid component in the few described cases
can take the appearance of a basaloid squamous cell carcinoma or be more
basal cell carcinoma-like (Fig. 26.128). Unlike so-called collision tumors,
biphasic tumors show an intimate association of the epithelial and mela-
nocytic components. An intraepidermal component is not always present.
Cases of melanoma metastatic to a basal cell carcinoma or colonization
by melanoma in situ have been described as mimics of basomelanocytic
tumor.5,6
The melanoma component stains with traditional melanocytic markers
such as S100 protein, MART-1, and HMB-45 (Fig. 26.129). Cytokeratins,
Fig. 26.124  including AE1/AE3, mark the basaloid component, and Ber-EP4 can also be
Dermal squamomelanocytic tumor: low-power view of a facial tumor. By courtesy positive (Fig. 26.130).3,4 Characteristic copy number alterations can be seen
of S. Poole, MD, Beth Israel Deaconess Medical Center, Boston, USA. in the melanoma component.1

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Lentiginous melanoma 1353

Fig. 26.129 
Basomelanocytic tumor: the inner cells express HMB-45. Mart-1 was also
positive. By courtesy of L. Erickson, MD, Mayo Clinic, Rochester, Minnesota.

Fig. 26.127 
Dermal squamoproliferative tumor: (A) the squamous component is highlighted
with keratin immunohistochemistry; (B) S100 protein. By courtesy of S. Poole,
MD, Beth Israel Deaconess Medical Center, Boston, USA.

Fig. 26.130 
Basomelanocytic tumor: the background population of basaloid cells expresses
keratin. By courtesy of L. Erickson, MD, Mayo Clinic, Rochester, Minnesota.

Lentiginous melanoma

Clinical features
This type of melanoma should be distinguished from acral lentiginous mela-
noma. These tumors have a propensity for the shoulders and upper back in
elderly patients while the lower legs, particularly in younger female patients,
may also be affected.1–6 Similar to lentigo maligna, this form of melanoma
may be associated with an extended in situ phase of many years, and in an
important number of cases there is a precursor lesion. The latter have been
described as atypical lentiginous melanocytic nevus and dysplastic atypical
nevus of the elderly with some questioning whether these lesions are truly
malignant.7–9 Additional study is required to further define and characterize
this seemingly distinct lesion.

Histologic features
Lentiginous melanoma, as the name implies, consists of a primarily single
Fig. 26.128  cell array of melanocytes achieving confluence at least focally with limited
Basomelanocytic tumor: surrounding undifferentiated tumor cells is a striking associated junctional nests (Figs 26.131–26.133). Pagetoid intraepidermal
palisade of basophilic cells typical of basal cell carcinoma. By courtesy of L. ascent can be noted but it tends to be very focal when present. Epidermal
Erickson, MD, Mayo Clinic, Rochester, Minnesota. atrophy is not present and there is preservation of normal rete architecture

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
1354 Melanoma

at the dermal–epidermal junction. Cytological atypia varies and may not be


prominent, and solar elastosis is not a feature. These features help separate
this entity from other forms of lentiginous melanoma. Melanocytic stains
may be helpful to better visualize the increased density of atypical mela-
nocytes. These lesions may be more readily recognized in larger excision
specimens as the features may be difficult to discern in smaller biopsies.2 As
stated before, in many lesions there is evidence of a lentiginous nevus with
variable atypia, suggesting a precursor lesion. This is the reason the diagno-
sis can be missed in small biopsies.

Primary dermal melanoma

Clinical features
The described cases of this apparently rare presentation of melanoma show
a dermal nodule lacking an identifiable in situ component or association
with a pre-existing nevus.1–4 Published series indicate that this presentation
of melanoma represents less than 1% of all melanomas.5–7 A wide ana-
tomic distribution is reported, but the head and neck region and extremities
Fig. 26.131  are the most common sites. In one study, in a cohort of 13 patients, the
Lentiginous melanoma: in this field, there is a lentiginous and focally nested average Breslow depth was 9.6 mm with 92% survival at a mean of about
atypical melanocytic proliferation. Note the upper dermal lymphocytic infiltrate.
4 years.8,9 Subsequent studies have documented a somewhat less impressive
but still relatively favorable 88% 5-year survival in a cohort of nine cases
with a median Breslow thickness of 3.4 mm.6 A series of 49 cases with
mean Breslow depth of 3.0 mm and mean mitotic rate of 4.3 per mm2 is
also reported.7 A caveat of this latter series is that 14 cases showed associa-
tion with a pre-existing intradermal nevus and average follow-up was only
26 months. Nineteen percent of patients developed locoregional recurrence
and 6% showed distant metastases; only one patient died of disease. Tra-
ditional AJCC parameters did not correlate with outcome. These studies
suggest that this tumor behaves much less aggressively than comparable
traditional nodular melanoma. Others examining a larger series of 101
cases defined as a solitary dermal melanoma (as distinct from melanoma of
unknown primary which usually presents as an isolated lymph node metas-
tasis) within larger well-characterized cohort of 12,817 patients found no
difference in outcome compared to stage-matched traditional melanomas.5
Other studies show various results, but most include melanoma of unknown
primary which could skew the results.10–14 Indeed, in all of these papers, it
is unclear whether the authors are describing the same tumor. Much more
study is needed to refine our definitions and understanding of this possible
entity and delineate its natural history. At the present time, this presenta-
tion is probably best assessed and reported as a standard melanoma with
Fig. 26.132 
Lentiginous melanoma: the tumor cells are predominantly basally located. There is
risk proportional to the assessed Breslow depth, though a note suggesting
no evidence of solar elastosis. that such cases have been described to have more indolent behavior would
certainly be appropriate.

Histologic features
The lesion is characterized by a nodular deposit of melanoma composed
of epithelioid to spindled cells in the dermis and/or subcutis. Spitzoid and
blue nevus-like features have been noted in some cases, but many have
the appearance of traditional melanoma.7 Some cases have conspicuous
mitoses, but overall a Ki-67 index may be lower than that seen in com-
parable cases of nodular melanoma or metastatic melanoma.8 It would be
extremely important to distinguish these cases from metastatic melanoma
or melanoma of unknown primary, both of which have vastly different
staging implications.15 This can be challenging and indeed such cases have
now been described to have BRAF and NF1 mutations as well as P16/
CDKN2A loss and other copy number aberrations similar to traditional
cutaneous melanoma.7,16 Explanations for this entity include an origin in
association with follicular melanocytes and complete regression of a prior
intraepidermal component. More recently, de la Fouchadiere and colleagues
have described five cases of non-pigmented dermal melanoma each with
a CRTC1-TRIM11 fusion that are dermal based and nodular in outline.17
Cytology ranges from epithelioid to spindled sometimes with scattered mul-
Fig. 26.133  tinucleate cells. In some ways the appearance is similar to that of clear cell
Lentiginous melanoma: junctional nests showing moderate to severe cytological sarcoma which can also sometimes be confined to the dermis.18 Behavior is
atypia are evident. low grade. This lesion would appear to fall within the spectrum of primary

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Molecular classification of melanoma 1355

dermal melanoma though where it falls in the end in the spectrum between indicate such a signature.5,6 For example, the most common mutation at
melanoma and dermal clear cell sarcoma remains to be seen. BRAF codon 600 is a T to A transition and thus does not match the clas-
sical UV signature.7 The fact that these mutations lack a UV signature does
not exclude a role for UV radiation in their formation, as they can form as
Molecular classification of melanoma
the result of indirect consequences of UV radiation such as the formation
Over the last decade significant molecular data have shed new light on dif- of reactive oxygen species. Furthermore, activating an oncogene requires
ferences between melanoma subtypes. This continues to inform an emerging mutations to change very specific amino acid sequences in the protein,
classification, which integrates epidemiology, clinical and histopathological whereas abrogating a tumor suppressor protein’s function can occur
features with patterns of somatic mutations and mutational mechanisms. through numerous possible mutations (missense, nonsense, and frameshift
This final portion of the melanoma chapter focuses on conceptual infor- mutations). Therefore, mutations that activate oncogenes tend to cluster in
mation regarding the molecular classification of melanoma and its cor- a few critical hotspots, whereas mutations in tumor suppressor genes are
relation to UV damage, as well as the progression from melanoma in situ more widespread and enriched for damaging mutations that truncate or
to invasive and metastatic melanoma. Molecular data continue to inform scramble the DNA sequence. The need for specific growth promoting muta-
our traditional classification schemes, validating them in some instances tions in oncogenes such as BRAF and NRAS therefore significantly skews
and expanding them in others. While traditional classification categories the mutations observed in these genes. The causative role for UV radiation
of melanoma are still employed, the molecular characterization of mela- in mutagenesis of melanomas on sun-exposed skin is clearly demonstrated
nocytic neoplasia continues to progress and will have to be continually by the high burden of UV signature mutations in the tens to hundreds of
integrated with the traditional features to establish a classification system thousand mutations found in the genomes of cutaneous melanomas and
that correctly describes distinctive phenotypes but is firmly based in the by the observation that mutations in critical genes such as the TERT pro-
underlying mechanistic alterations that are in large part caused by genetic moter, CDKN2A, P53, and PTEN are indeed enriched for UV signature
alterations. mutations.8,9
However, not all melanoma subtypes harbor genomic damage attrib-
Melanoma epidemiology utable to UV radiation. Melanomas arising on the palms and soles, nail
Melanomas are most common in Caucasians, where they typically arise on beds, and mucosal sites, as well as uveal melanoma, typically lack the high
sun-exposed skin. In patients under the age of 50, the highest density is mutation burden with a UV signature found in cutaneous melanomas. This
found on the trunk and extremities. In patients older than 50, the highest indicates that while UV radiation plays a critical role in the pathogenesis of
density of melanomas is found on the head and neck. The high frequency of some melanoma subtypes, it does not in others. This could also explain why
melanoma and non-melanoma skin cancers in Caucasians is considered to acral and mucosal melanomas affect all world populations independent of
be the result of light skin pigmentation with the consequence of increased skin complexion.10
UV-induced mutagenesis. Some aspects of skin color variation are likely due
to selective advantage during human evolution that optimizes benefits from Melanoma is composed of distinct subtypes
solar radiation (e.g., lighter skin pigmentation to enhance vitamin D syn- There is considerable variation in the clinical and histopathological presen-
thesis) in less sunny climates. Association studies have revealed common tation of melanomas that partially depends on the anatomic site in which
polymorphisms in pigmentation genes are genetic factors controlling skin the tumor arises and how much ultraviolet radiation exposure has occurred.
color, tanning ability, and freckling, with some variants increasing mela- As outlined previously in this chapter, these differences formed the tradi-
noma risk. These pigmentation genes include the melanocortin receptor tional ‘histogenetic’ classification of melanoma as initially proposed in the
one (MC1R), its antagonist the agouti signaling protein (ASIP), tyrosinase Sydney Classification.11 There has been considerable controversy on the jus-
(TYR), tyrosinase-related protein (TYPR), the P-gene mutated in oculocuta- tification of this classification and opponents have long posited that mel-
neous albinism type II (OCA2), ion exchange proteins of the solute carrier anoma is a single disease, with the variation in clinical and pathological
family SLC24A5, SLC24A4, and SLC45A2 (MATP), and the interferon reg- features representing secondary phenomena. This belief stemmed both from
ulatory factor IRF4 or MUM1.1 the lack of difference with regards to prognosis of advanced disease and the
Decreased pigmentation in Caucasian skin leads to decreased protection inability in the past to predict a different response to therapy between sub-
from UV radiation and results in increased somatic mutations in skin cells, types.12 In addition, there was significant overlap in the defining phenotypic
including melanocytes. However, increased melanoma susceptibility in Cau- features that hindered unequivocal classification of a considerable portion
casians may not be entirely attributed to differences in skin pigmentation, of primary tumors.13
because albinos of African descent do not share the increased incidence of Considerable progress has now been made in the identification of genetic
melanomas, while they do suffer more frequently from cutaneous squamous alterations in melanoma and has provided strong support for the concept of
cell carcinoma.2 Melanin, which is present in Caucasian skin and absent biologically distinct melanoma types. This is consistent with work leading
in albinos who lack functional tyrosinase, appears to have both protec- to the discovery and validation of clinically relevant genetic subtypes that
tive and deleterious effects for DNA. Mutations from UV radiation con- have emerged in other malignancies such as lung and colon cancer. This is
tinue to accumulate in the skin for hours after removal from UV light.3 a decisive trend in oncology that is critical for the implementation of pre-
These ‘dark’ mutations do not occur in albino mice lacking melanin. In cision cancer medicine. Mutations in specific genes, mutational signatures,
mice with intact melanin production, these mutations were twice as fre- and other genetic alterations such as genome-wide chromosomal aberra-
quent in those with increased pheomelanin (due to MC1R mutation) versus tions correlate with specific clinical and histopathological features of mel-
those with high eumelanin, implicating melanin, particularly pheomelanin, anoma, providing genetic support for the original concept that melanomas
as a critical factor in these ‘dark’ mutations. Further evidence is found in fall into different categories of diseases. New genetic data offer an oppor-
transgenic mouse models in which melanoma development after UVA radi- tunity to integrate causal genetic alterations with clinical or histopatholog-
ation only occurs in black mice with intact melanin production and not in ical disease attributes, as well as predisposing and environmental factors.
albino mice.4 The emerging melanoma subtypes that integrate clinicopathological features
overlap considerably with the original ‘histogenetic’ melanoma types, but
Melanoma and UV radiation offer a more nuanced picture of the categories. This continuity is satisfying
The establishment of a causal link between UV radiation and melanoma and underscores the enduring importance of thoughtful clinicopathologi-
formation requires several considerations. UV radiation causes a wide spec- cal characterization as a basis for initial classification. The extent of UV
trum of mutations. Among these UV signature mutations, pyrimidine dimers radiation damage can serve as an initial branch point in the classification
resulting in C > T or CC > TT transitions are the most common and are due of melanoma: melanomas found in skin with high cumulative sun damage
to a direct photochemical reaction with the DNA. However, mutations in (high-CSD), low cumulative sun damage (low-CSD), and melanomas on
common melanoma oncogenes such as BRAF and NRAS typically do not sites with negligible (e.g., acral) or no sun exposure (e.g., mucosal).

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
1356 Melanoma

%\0XWDWLRQ6WDWXV
Melanoma on sun-exposed skin
0XWDWLRQ %5$) 15$6 1(,7+(5
Epidemiological data alone suggest differences between melanomas from
sun-exposed skin and melanomas on acral or mucosal sites, the latter of :+27\SH
which do not show variation in incidence between races. Additionally, the 3LJPHQW
anatomic site of origin and the cumulative dose of UV radiation are two 6FDWWHU
intricately linked features that are strongly associated with distinctive clin-
1HVWLQJ
ical and histopathological presentations of melanoma. Melanomas on the
head and neck tend to occur in older individuals with a peak incidence of &LUFXPVFULSWLRQ
about 72 years of age.14 The skin surrounding these melanomas typically (SLG&RQWRXU
shows signs of chronic sun damage, as evidenced by pronounced solar elas- 6RODU(ODVWRVLV
tosis and other signs of chronic sun-induced damage such as solar lentigi-
&HOO6KDSH
nes and actinic keratoses.15 Total cumulative UV dose appears to represent
a major risk factor for these melanomas. The traditional lentigo maligna &HOO6L]H
variant of melanoma falls into the high-CSD category.
$JH
By contrast, melanomas involving the trunk arise earlier in life, with a
peak incidence at around age 54, and signs of chronic sun damage (severe Fig. 26.134 
solar elastosis) are typically absent.14 The drop in incidence of melanomas Morphological and clinical features of 302 primary melanomas by mutation
on the trunk after age 54 suggests that it is not the cumulative dose of status: the heat map shows the features that are significantly associated with
UV radiation but timing of UV exposure that plays a decisive role in these mutation status for three groups of melanomas: BRAF mutant, NRAS mutant,
or neither mutation. The scores for the features in the following rows range
melanomas, with a particular window of vulnerability present earlier in
from shades of blue (low score) to gray (intermediate scores) to yellow (high
life. Epidemiological studies demonstrate that exposure to UV irradiation
scores): pigmentation of neoplastic melanocytes, upward scatter of intraepidermal
in childhood plays an important role in the pathogenesis of melanoma in melanocytes, nesting of melanocytes, degree of solar elastosis, cell size, and
general.16 Patients with melanomas on the trunk, but not with melanomas patient age. For circumscription, yellow indicates an abrupt transition from
on chronically sun-exposed sites such as the face, have more melanocytic involved to adjacent uninvolved skin, gray scores represent a continuous
nevi, suggesting a common pathogenesis between low-CSD melanomas and transition, and blue scores a discontinuous transition. Epidermal contour ranges
nevi.17 This is supported by the fact that low-CSD melanomas more fre- from yellow (acanthotic) to blue (atrophic) of the epidermis involved by the
quently have adjacent precursor nevi than CSD melanomas and that nevi melanoma. Cell shapes range from blue (round) to yellow (spindled). Samples
and low-CSD melanomas share a high frequency of BRAF V600E muta- are listed in columns and are ordered within each category using agglomerative
tions. A functional scale for assessing sun damage has been proposed to hierarchical clustering. The color codes for WHO types are superficial spreading
melanoma (SSM) green, lentigo maligna melanoma (LMM) yellow, nodular
grade the degree of solar elastosis involving the dermis.18,19 Recent studies
melanoma (NM) red, and acral lentiginous melanoma (ALM) orange. Unclassifiable
have demonstrated how melanomas progress from such nevi through the
samples are in black.
acquisition of additional UV-associated mutations.20 The fact that most nevi
arise during childhood and adolescence furthers highlights the possibility
of a window of vulnerability towards UV radiation early in life for these
types of melanocytic neoplasms.21 Genetic data also support the concept of loss.20 The latter mutations reflect the importance of replicative senescence
distinct melanoma types based on sun-exposure as discussed below.22 and maintaining G1/S checkpoint control in preventing nevi from progress-
ing to melanoma. UV signature mutation profiles implicate UV radiation as
Melanoma with low cumulative sun damage (low-CSD) the driving factor in this transformation of nevi to melanoma.20
Low-CSD melanomas occur most frequently in patients under 50 on the Progression from in situ to invasive melanoma involves additional genetic
trunk or extremities and show a low to moderate degree of solar elastosis alterations required for unrestrained growth outside the confines of the epi-
histopathologically. BRAFV600E mutations are very common (approximately dermis. Ability to grow and survive in the dermis alone is not indicative of
70%) in low-CSD melanomas, representing the main driver mutation, fol- malignant transformation, as the cells of melanocytic nevi already have this
lowed by NRAS mutations (approximately 20%).19,23 BRAF mutant mel- property. Loss of cell cycle checkpoint regulation is critical in unleashing or
anomas frequently exhibit specific histomorphological features such as increasing the proliferative capacity associated with invasive melanoma as
increased upward scatter of intraepidermal melanocytes (pagetoid scatter), evidenced by frequent homozygous loss of the CDKN2A gene encoding both
a predominance of nests over single cells, thickening of the involved epider- p16 and p14ARF in the transition from in situ to invasive melanoma. Addi-
mis, a sharper demarcation toward the adjacent uninvolved epidermis, and tional mutations in genes encoding critical tumor suppressor proteins such
constituent tumor cells that are larger, rounder, and more heavily pigmented as TP53 and PTEN occur later in the progression to more advanced stage
(Fig. 26.134).24 Simple combinations of these independently associated fea- disease.20 Additionally, invasive melanomas must evade antitumor immune
tures predicted BRAF mutation status with 90% accuracy in one study.24 surveillance through mechanisms such as induction of immune tolerance.
These histomorphological criteria also performed better in predicting muta- The vast majority of melanomas exhibit some degree of genomic insta-
tion status than the traditional WHO melanoma classification, indicating bility in the form of chromosomal gains and losses. This instability seems to
the potential for developing improved classification schemes that define coincide with the transition from benign melanocytic nevi to melanoma in
more biologically homogeneous subsets. This is increasingly relevant with situ, as nevi tend to not have chromosomal copy number changes, whereas
the increased use of targeted therapies to specific genetic alterations includ- they can be detected in melanoma in situ.20 The instability may be related to
ing BRAF inhibitors25 and KIT inhibitors.26–28 telomere shortening and end-to-end fusion of sister chromatids with eroded
The shared high incidence of BRAFV600E mutations in melanocytic nevi telomeres that result in dicentric chromosomes that can rupture during
and low-CSD melanomas supports a common etiology with regard to UV mitosis. The selective advantage of TERT promoter mutations early during
radiation and timing of exposure. Approximately one-fourth to one-third of melanoma progression suggests that replicative senescence becomes an early
melanomas arise within histopathologically identifiable precursor nevi, with barrier to transformation. TERT promoter mutations only lead to marginal
low-CSD melanomas representing the main melanoma type in which precur- telomerase expression, which manages to preserve critically short telomeres
sor nevi can be identified.29 Frequently only melanoma in situ is present with but cannot fully suppress telomere fusions resulting in instability that gen-
the associated nevus, the latter of which remains identifiable in the under- erates DNA copy number changes.30 This explains why the telomeres of
lying dermis. The melanoma in situ harbors the same initiating mutation melanomas with TERT promoter mutations remain short.30,31
as the precursor nevus (typically BRAF or NRAS), with additional genetic While there are some similarities in the patterns of the chromosomal
aberrations such as TERT promoter mutation or heterozygous CDKN2A gains and losses across melanoma subtypes, significant differences exist with

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Molecular classification of melanoma 1357

0.6 Gains Fig. 26.137 


0.2 .,7 .,7DQG .,7DQG 15$6 %5$) Frequency distribution of
-0.2
Non-CSD

Losses 15$6 %5$) genetic alterations in BRAF,


-0.6 NRAS, and KIT among four
0.2 Amplifications groups of melanoma: non-
0.0

Homozygous deletions CSD, melanomas on skin
-0.2
without chronic sun-induced
1 3 5 7 9 11 13 15 17 19 21 damage; CSD, melanomas
on skin with chronic sun-
induced as evidenced by the

3HUFHQW$EHUUDQW
0.6 Gains presence of marked solar
0.2
 elastosis; acral, melanomas
-0.2
Losses on the soles, palms, or
CSD

-0.6
0.2
subungual sites; mucosal,
Amplifications
0.0 melanomas on mucosal
-0.2 Homozygous deletions membranes. One CSD
melanoma had a KIT and
1 3 5 7 9 11 13 15 17 19 21
an NRAS mutation, and one
Fig. 26.135   acral melanoma had a KIT
1RQ&6' &6' $FUDO 0XFRVDO
Frequencies of DNA copy number changes in non-CSD and CSD melanomas: and a BRAF mutation.
the upper histogram for each melanoma type shows the frequencies of gains
(green) and losses (red), and the lower histogram shows amplifications (green)
and homozygous deletions (red). The x axis represents the genomic position By contrast, activating mutations in NRAS activate both the MAP kinase
from chromosome 1 to 22. Vertical solid blue lines mark the boundaries between and the PI3 kinase pathway, therefore obviating the need for a loss of PTEN
chromosomes, and the vertical dashed gray lines mark the position of the in tumors with NRAS mutations.32 Point mutations in the catalytic subunit
centromeres. of PI3 kinase have also been found in a small minority of melanomas.33
Gains of chromosome 7 are more common in low-CSD melanomas.19
BRAF resides at 7q34 and the copy number increase reflects duplication of
Receptor Tyrosine Kinase the mutant BRAF allele, which provides a proliferative advantage.34,35 Copy
(e.g., KIT) number increases of CCND1 encoding cyclin D1 at chromosome 11q13
represent an additional distinguishing feature as they occur infrequently
in low-CSD melanomas, while present in up to 50% of high-CSD mela-
noma.19,36 These genetic changes demonstrate how both the major prolif-
PTEN erative and survival pathways are activated during melanoma progression.
NRAS
BRAF PI3K
This can be accomplished through different combinations of mutations and
chromosomal aberrations such as activating mutations in upstream tyrosine
kinase receptors such as KIT or NRAS which activate both the MAP kinase
MEK AKT
pathway and PI3K pathway, or through combinations of activating muta-
tions in BRAF combined with loss of PTEN (Fig. 26.136).

ERK Survival Melanoma with high cumulative sun damage (high-CSD)


High-CSD melanomas typically occur in Caucasians over age 50 mostly on
p16
the head and neck and display marked solar elastosis histopathologically.
CCND1 In contrast to low-CSD melanomas, high-CSD melanomas typically have
CDK4 no associated precursor nevus, arising ‘de novo’ in the epidermis. These
CDK6 melanomas are often characterized by an extended in situ stage, in which
Proliferation the neoplasm can grow to the size of several centimeters over many years
prior to becoming invasive. Genomic data on the in situ stage of high-CSD
melanoma are limited. Approximately 20% of such in situ melanomas have
Fig. 26.136  BRAF mutations, although BRAFnon-V600E mutations (e.g., BRAFV600K) are
Simplified schematic of pathways important in melanoma: downstream from more frequent than BRAFV600E mutations in this group.37 In addition to the
certain receptor tyrosine kinases lies Ras which bifurcates to the BRAF/ERK and striking difference in BRAF mutations between high-CSD and low-CSD
PI3K/AKT pathways leasing to proliferation and survival. It appears that activation melanomas (15% vs 70%),34,38 additional genetic differences have also been
of both of these pathways (and others) is required for melanoma development found. For instance, some studies report up to 30% of high-CSD melano-
and these are grouped in a rational fashion. For instance, activating mutations
mas harbor mutations or DNA copy number increases of KIT, whereas KIT
in KIT, NRAS, and BRAF are mutually exclusive since they activate the same
pathways and loss of PTEN is seen with BRAF, but not NRAS mutation, because
mutations are rare in low-CSD melanomas (Fig. 26.137).39 Differences in
NRAS activates the PI3K pathway while BRAF does not. These combinations will the mutation frequency of NRAS have been reported by some, but do not
likely have therapeutic relevance as more specific inhibitors are developed. appear to be a consistent finding.19,40
Desmoplastic melanoma represents a specific variant of melanoma that
occurs in chronically sun-damaged skin and has a high mutation burden
respect to tumors based on the degree of UV exposure.19 Gain of chromo- that typically exceeds that of all other melanomas (median 62 mutations/
some 6p and loss of 6q are common in both low-CSD and high-CSD mel- Mb compared to approximately 15/Mb in low-CSD melanoma).41 The
anomas, but chromosome 10 loss occurs in 40% of low-CSD melanomas mutational profile of desmoplastic melanoma has a strong UV radiation
and fewer than 10% of CSD melanomas (Fig. 26.135). The region com- profile and is notable for the complete absence of BRAF and NRAS hotspot
monly lost on chromosome 10 contains the tumor suppressor PTEN, which mutations and a high prevalence of inactivating NF1 mutations, which can
encodes a negative regulator of the PI3 kinase pathway and is considered also be detected in the overlying in situ component.41,42 In addition, there are
to represent the major selective force for chromosome 10 loss. BRAF muta- recurrent mutations in the promoter of NFKBIE, which encodes an inhibi-
tions only activate the MAP kinase pathway, which explains the advantage tor of nuclear factor NF-κB signaling, and inactivating mutations in TP53,
of concurrent PTEN loss to activate the PI3 kinase pathway (Fig. 26.136).32 CDKN2A, ARID1A, ARID2, and RB1. TERT promoter mutations are also

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
1358 Melanoma

Table 26.3 
Genes implicated in melanoma development
Gene Function Associated with melanoma susceptibility
ACD/POT1/TERF2IP Nuclear proteins that bind telomeres and prevent inappropriate recombination events Yes
ARID1/ARID2 Chromatin remodeling
BAP1 Deubiquitinase that binds to BRCA1 and is involved with chromatin dynamics Yes
BRAF Protein kinase in the MAP kinase pathway
CDKN2A Encodes p16, an inhibitor of cyclin-dependent kinases, and p14, an inhibitor of MDM2 Yes
CDK4 Cyclin-dependent kinase that phosphorylates Rb Yes
CTNNB1 Encodes beta-catenin, an adherens junction protein that signals in the WNT pathway
ERBB4 EGFR family of tyrosine kinase receptors involved in differentiation and proliferation
GNAQ/GNA11 G-protein coupled receptor that activates phospholipase C-beta
KIT Tyrosine kinase receptor activating multiple pathways in cell survival and proliferation
MC1R Encodes the melanocortin 1 receptor that binds melanocyte stimulatory hormone Yes
MGMT DNA repair protein Yes
MITF Transcription factor in melanocyte development Yes
NF1 Encodes neurofibromin that negatively regulates RAS signaling
NRAS Tyrosine kinase receptor that activates the MAP kinase and PI3K pathways
POLE DNA polymerase subunit involved in DNA repair Yes
PTEN Lipid phosphatase antagonist of PI3K pathway Yes
RAC1 GTPase involved in cell growth
SLC45A2 Transporter protein in melanin synthesis Yes
TERT Encodes telomerase which maintains telomere length; can be amplified or have Yes
promoter mutation
TP53 Encodes tumor suppressor p53
XP genes Xeroderma pigmentosum genes encode nucleotide excision repair proteins Yes

common in desmoplastic melanomas.41 Desmoplastic melanomas have fewer corneum providing additional UV protection. The nail matrix is protected
chromosomal copy number alterations than most other melanomas, though by the proximal nail fold and the nail plate. With the exception of the lips
focal amplifications of oncogenes such as EGFR, CDK4, CCND1, MDM2, and the bulbar conjunctiva, mucosal sites in which melanoma arise are not
TERT, and MAP3K1 and focal deletions in tumor suppressor genes such as exposed to sunlight. Genomic analyses with CGH and DNA sequencing in
CDKN2A occur.41 melanomas from these UV-protected sites show a high degree of genomic
Numerous other recurrent genetic alterations have been discovered in instability reflected by frequent chromosomal aberrations, with focused gene
melanomas on sun-exposed skin including amplifications or mutation of amplifications in particular and a low mutation burden (Fig. 26.138).19,50–53
MITF and activating mutations in CTNNB1, encoding β-catenin. MITF is a The mucosal example in Fig. 26.138 in particular shows copy number
basic helix-loop-helix (hHLH)-leucine zipper protein that plays a role in the changes affecting virtually every chromosome. Although mucosal melano-
development of melanocytes.43 β-catenin is a critical signaling component of mas are typically detected with considerable latency and therefore tend to
the WNT pathway where it acts as an adherens junction protein that can be thicker than the primaries of the other melanoma categories, this high
also function as a transcription factor and is mutated in a small subset of degree of genomic instability can already be demonstrated in thinner lesions.
melanomas.44,45 It is also increasingly clear that telomere-related proteins In other cancers, gene amplifications usually arise late during progression
are critical factors in melanoma development. Germline mutations in com- and often are a sign of adverse prognosis.54 By contrast, in acral melanoma
ponents of the telomere shelterin complex (e.g., POT1, ACD, TERF2IP), these amplifications arise very early during progression and can already be
which is responsible for capping telomere ends and preventing recombina- detected at the in situ stage.50
tion events, have been linked with familial melanoma.46 Mutations in the Gene amplifications typically arise through repeated cycles of
promoter of TERT, a gene encoding the protein component of telomer- double-stranded DNA breaks and subsequent end-to-end fusions of chro-
ase, have also been described in familial melanoma and in 35% to 70% of matids and thus indicate a loss of control over genomic integrity. The cause
sporadic melanomas.47,48 TERT promoter mutations are the most common of this loss of control and subsequent genetic instability is currently unclear
mutations in melanoma and have a UV signature C > T pattern. The predis- since TP53 and other known instability-inducing mutations are uncommon
posing mutations in TERT and other telomere-related factors are thought in these melanomas. While it is evident that UV radiation does not play
to contribute to increasing melanoma risk by increasing telomere length and a significant role in these melanomas, it is currently unresolved whether
extending the replicative lifespan of neoplastic cells, thereby increasing the or not there are specific carcinogens involved in the pathogenesis of acral
likelihood of acquiring the additional mutations required for transforma- and mucosal melanomas or whether they arise as a consequence of sto-
tion.49 Numerous additional genes have been implicated in melanoma, many chastic alterations without external influence. The marked differences in the
of which are listed in Table 26.3. degree of genomic instability and mutation burden between melanomas on
sun-protected sites and those on sun-exposed skin indicates that the types
Melanomas on UV-protected sites of genetic insults are fundamentally different between these melanoma sub-
Acral and mucosal melanoma types. In sun-exposed melanomas UV photoproducts and oxidative damage
The volar surfaces of the hands and feet are minimally exposed to UV radi- are the predominating drivers of tumorigenesis, whereas double-stranded
ation, and glabrous skin has a thicker epidermis with a very thick stratum DNA breaks as part of breakage-fusion-bridge cycles and/or other yet to be

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Molecular classification of melanoma 1359

2 4 6 8 10 12 14 16 18 20 22 Y 10 mm
2.0
A B
1.5
1.0 C
Non-CSD

0.5
Log2Ratio

0.0

-1.0

-2.0
1 3 5 7 9 11 13 15 17 19 21 X
1.0292
D
2 4 6 8 10 12 14 16 18 20 22 Y
3

2
Log2Ratio

1
CSD

-1

-2
1 3 5 7 9 11 13 15 17 19 21 X Cyclin D1
1.0292
hTERT
65 0.24 sample AM108 acral
2 4 6 8 10 12 14 16 18 20 22 Y Fig. 26.139 
2.0 Field cells in acral melanoma: the top panel shows an acral melanoma with
1.5
close-ups of four particular areas underneath. Area A shows superficially invasive
1.0
melanoma that by FISH shows amplification of CCND1 (cyclin D1) and TERT
0.5
Log2Ratio

telomerase reverse transcriptase (clusters of red and green signals in blue stained
Acral

0.0
nuclei, respectively). Area B shows melanoma in situ with amplification of CCND1
-1.0 and normal copy number of hTERT. Areas C and D show histopathologically
uninvolved epidermis which by FISH harbors single basal melanocytes with
-2.0 amplification of CCND1 (clusters of red signals within blue stained nuclei). In the
1.0292 1 3 5 7 9 11 13 15 17 19 21 X
second FISH panel for area D a melanocyte with CCND1 amplification is situated
72 0.37 sample AM124 amucosal in an eccrine duct.
2 4 6 8 10 12 14 16 18 20 22 Y
2.0
1.5
1.0 that they reflect an early progression phase of disease.55 Once their clini-
Mucosal

0.5 cal relevance has been defined, the determination of field effect may offer
Log2Ratio

0.0
a more objective guidance for the size of surgical margins for removal of
-1.0 primary melanomas than the current empirically derived recommendations.
Although acral and mucosal melanomas share a high incidence of chro-
-2.0 mosomal aberrations, including frequent gene amplifications, the genomic
1.0292 1 3 5 7 9 11 13 15 17 19 21 X
regions affected by these copy number alterations differ significantly
Fig. 26.138  between the two.19 The most commonly amplified site in acral melanomas
Differences in the degree of genomic instability among melanoma types: is the CCND1 locus encoding cyclin D1 on chromosome 11q13. Another
representative examples of array CGH profiles of non-CSD, CSD, acral, and commonly amplified site includes the TERT telomerase locus on chro-
mucosal melanoma. The y axis represents the copy number for each array mosome 5p15. However, while approximately 50% of acral melanomas
element (average value of a triplicate measurement) expressed as the log2 of amplify CCND1, mucosal melanomas typically do not. Instead, a subset of
the ratio of the tumor to reference fluorescence intensities. Values between mucosal melanomas amplifies cyclin dependent kinase 4 (CDK4) on chro-
+0.25 and –0.25 would be considered normal; values above that range are mosome 12q14, encoding the binding partner of cyclin D1.19 CDK4 is nor-
gains and below that range are losses. Values above 0.9 would be considered mally inhibited by p16 function, which functions as the gatekeeper of the
amplifications. In practice, a more complex assessment that takes into account
G1/S transition point. In mucosal melanomas, the amplification of CDK4
the signal-to-noise ratio for each individual case is used.33 The x axis represents
the genomic position from chromosome 1p to 22.
is mutually exclusive with deletions at the p16 locus, indicating that these
genomic aberrations may be functionally equivalent. Mucosal melanomas
also have recurrent mutations in SF3B1, a gene commonly mutated in uveal
discovered mechanism(s) appear critical for acral and mucosal melanoma melanoma, which is typically not found in acral melanoma.56
formation. In contrast to the high number of chromosomal copy number changes
This chromosomal instability in acral and mucosal melanoma occurs and amplifications in acral and mucosal melanomas, their mutational
early in disease progression and has even been detected in histopathologi- burden is much lower than sun exposed melanomas. Many of the mutations
cally normal appearing melanocytes around acral melanoma in situ, so-called show a pattern of mutagenesis seen with aging in which there is sponta-
field cells or field effect (Fig. 26.139).55 Field cells harbor some, but not all neous deamination of 5-methylcytosine at CpG dinucleotides.31 UV signa-
of the gene amplifications found in the adjacent melanoma, indicating that ture mutations are absent for the most part. Most genetic studies of acral
they are precursors of the histopathologically manifest form of melanoma and mucosal melanoma focus on invasive melanoma, with very little pub-
in situ (Fig. 26.140). These cells represent clonal expansions of melanocytes lished on the in situ stage. BRAFV600E mutation in acral melanoma in situ
with severe genetic alterations stemming from loss of control over mainte- has been reported,57 but the incidence of BRAF mutation in acral melanoma
nance of genomic integrity. Such genetic changes are qualitatively different (approximately 15–20%) is significantly lower than in low-CSD melanomas
from point mutations in oncogenes such as BRAF, as they indicate loss (approximately 70%) and even lower in mucosal melanoma (approximately
of protective factors that preserve genomic integrity. Field cells are there- 5%), suggesting BRAF mutation initiates a comparatively small percentage
fore best interpreted as an early form or precursor of melanoma in situ. of these melanomas.34,58–63 NRAS mutation and KIT mutation or amplifi-
These fields of neoplastic melanocytes can be extensive, present up to > 1  cm cation are more frequently found in acral and mucosal melanoma.58,64,65
beyond the histopathologically detectable in situ portion, and the size of the These two mutations activate the MAP kinase pathway and drive prolifera-
field is completely independent of tumor thickness, further demonstrating tion, so it is logical to assume they occur in the early proliferative phase of

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
1360 Melanoma

toward the uninvolved epidermis. These growth characteristics may be


related to biological effects of KIT activation. KIT is essential for mela-
noblast migration from the neural crest into the skin and for homing into
epithelial structures during development. If constitutively active KIT is
introduced into human melanocytes in vitro, it induces a migratory phe-
notype resembling a lentiginous pattern.78 Aberrant KIT signaling in mela-
noma may therefore help explain the field effect described for melanomas
on acral skin (Figs 26.139 and 26.140).50

Melanocytic tumors arising without associations to


epithelial structures
Most melanocytic neoplasms, benign melanocytic nevi as well as mela-
nomas, originate from melanocytes situated within epithelial structures
throughout the body and have mutations in genes such as BRAF, NRAS,
NF1, and KIT. However, a subset of melanocytic neoplasms arises without
an apparent connection to epithelial structures and does not show these
mutations.38,79 One category, uveal melanoma, arises from melanocytes
within the choroidal plexus, the ciliary body, and the iris of the eye and is
biologically distinct from cutaneous melanoma by a very strong propensity
to metastasize to the liver.80 It also differs in the presence of certain chro-
mosomal aberrations such as frequent loss of chromosome 3, which serves
as a negative prognostic indicator in uveal melanoma.81 In the skin, intra-
dermal melanocytic proliferations, which can be congenital or acquired,
present in diverse ways ranging from discrete blue papules (blue nevi) to
large blue-gray patches affecting the conjunctiva and periorbital skin (nevus
of Ota), shoulders (nevus of Ito), and the lower back (Mongolian spot).82 A
potential connection between intradermal melanocytic neoplasms and uveal
melanoma is suggested by the fact that nevus of Ota represents a risk factor
for uveal melanoma in Caucasians. In addition, blue nevi can show cytolog-
ical similarities to uveal melanoma.80 Interestingly, while uveal melanomas
strongly express KIT on immunohistochemistry, they lack mutations in the
KIT gene.79
A forward genetic screen in mice identified germline mutations in the
Fig. 26.140  heterotrimeric G-protein subunits GNAQ and GNA11 that cause skin
Field cells in acral melanoma: bottom panel: clinical picture with (1) melanoma in hyperpigmentation by inducing a subtle intradermal proliferation of mela-
situ; (2) clinically, dermoscopically, and microscopically normal skin; (3) invasive nocytes.83 GNAQ and GNA11 are members of the q class of G-protein
melanoma of 3.0-mm thickness. Black line, surgical margin; white line, field cell alpha subunits and are involved in mediating signals between G-protein
margin identified by amplification of 11q13. Middle and upper panels, respectively: coupled receptors (GPCRs) and downstream effectors.84 Somatic mutations
H&E and FISH for areas 1–3. FISH images represent one focal plane, so not all in GNAQ are found in 83% of blue nevi, 50% of blue nevus-like melanoma
signals are visible. A green immunofluorescent labeled Melan-A antibody was (‘malignant blue nevi’), and 46% of uveal melanomas.85 The mutations in
used to aid in identifying basal melanocytes. Cells with amplification of 11q13 are human tumors differed from the genetic variants discovered in the mouse
highlighted by arrowheads for areas 1 and 2 (upper panels). screen. In human melanocytic tumors, all mutations occurred exclusively at
codon 209, which is homologous to codon 61 of NRAS family members
and leads to constitutive activation. GNAQ activates protein kinase C
melanoma in situ on acral and mucosal sites. Future studies are necessary (PKC) family members PKCδ and ε via the release of diacylglycerol (DAG)
to confirm this supposition. TERT promoter mutations are less common in by phospholipase Cβ.86 In vitro studies show that the GNAQQ209L muta-
acral melanoma (approximately 10%),58 but TERT gene amplification is tion transforms melanocytes with efficiencies comparable to NRAS muta-
frequently seen and has been documented in acral melanoma in situ.66 tions and leads to activation of the MAP kinase pathway. This MAP kinase
While mutations in the receptor tyrosine kinase KIT often occur in acral activation occurs specifically through recruitment of RasGRP3 to the cell
and mucosal melanomas, similar to high-CSD melanomas, their incidence membrane by DAG and its phosphorylation and activation by PKC δ and
varies by anatomic site.34,56,58,67–69 Anorectal and vulvovaginal have the ɛ. RasGRP3 then activates RAS to initiate MAP kinase pathway signaling.86
highest number of KIT mutations (approximately 35%).56,68–70 Anorectal When introduced into murine melanocytes, mutant GNAQ induces heavily
melanomas also have frequent NF1 and SF3B1 mutations (20% each), the pigmented tumors that morphologically resemble the spectrum of human
latter of which is often present in uveal melanomas.56 Twenty percent of blue nevi and pigmented epithelioid melanocytomas, confirming its role in
acral and penile melanomas have KIT mutations58,68 while oral melanomas driving these tumor types.85
have approximately 10% KIT mutation prevalance.62,63 While early clini- While GNAQ or GNA11 mutations are sufficient to initiate growth of
cal trials of unselected melanoma patients failed to show any efficacy with these melanocytic neoplasms (Fig. 26.141), additional genomic aberrations
KIT inhibitors,71 subsequent case reports and follow-up studies indicate that are required for transformation into melanoma (e.g., uveal melanoma, blue
when melanoma patients with confirmed KIT mutations are selected for, nevus-like melanoma). Additional mutation or loss of the tumor suppressor
dramatic responses to KIT inhibitors such as imatinib and other agents do BRCA1-associated protein 1 (BAP1) gene in uveal melanoma is associated
occur.26–28,72–74 Although the majority of melanomas with genetic alterations with a poor prognosis and has also been found in the transformation of blue
in KIT overexpressed the protein as determined by CD117 immunohisto- nevi to melanoma.87,88 Recurrent SF3B1 and EIF1AX mutations have also
chemistry, increased immunoreactivity does not reliably predict responding been found. They typically occur in mutually exclusive fashion from BAP1
patients.39,75–77 Patients with KIT amplification without mutation do not loss and are associated with a more indolent disease course.89,90 Array CGH
appear to significantly benefit from the KIT inhibitor therapy.72–74 profiling of cellular blue nevi, atypical cellular blue nevi, and blue nevus-like
The melanoma categories in which genetic alterations of KIT are found melanoma shows increasing genomic instability during progression through
frequently show a lentiginous growth pattern and poor circumscription these stages, with most nevi having no chromosomal aberrations and a

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Molecular classification of melanoma 1361

features, appears to be a heterogeneous category with the majority of cases


representing BRAF and NRAS mutant melanoma98 rather than melanomas
that evolved from bona fide Spitz nevi or atypical Spitz tumors with HRAS
mutations or various kinase fusions. The genetic features of bona fide
Spitzoid melanomas, i.e., melanomas that originate from Spitz nevi or are
closely related to them molecularly, are currently poorly characterized. Both
homozygous loss of CDKN2A99 and TERT promoter mutation100 appear to
occur in some lethal Spitzoid melanomas, though the small number of cases
reported with these aberrations limits the generalizability of these findings.

Emerging concepts in metastatic melanoma


The traditional concept of melanoma progression to metastasis starting
from primary invasive melanoma and spreading through lymphatics to
regional lymph nodes, and then from regional lymph nodes to distant sites
is inadequate in explaining the observed biological behavior of metastatic
melanoma. Phylogenetic analysis of separate metastatic foci in melanoma
patients shows genetically distinct populations within the same patient,
indicating that parallel seeding of metastatic sites occurs from the primary
tumor followed by subsequent separate genetic evolution at the sites of
metastasis.101–103 Metastatic ‘reseeding’, in which cells from a distant met-
astatic site travel to other sites of metastasis and proliferate,104 may also
occur and could explain the emergence of multifocal treatment resistance
Fig. 26.141  to targeted therapies in some patients.105 Some evidence supporting WNT
An animal model pathway activation in the progression to metastatic melanoma has been
of GNAQ mutant reported,101,106 and MITF amplification has been found to be more preva-
melanocytes: mouse lent in metastatic disease.107 However, multiple genetic analyses of primary
melanocytes stably tumors and metastases have not identified a distinct set of genetic changes
transduced with mutant that reproducibly predict progression to metastasis.108–110
GNAQ, but not with
wild-type GNAQ, induce The Cancer Genome Atlas
highly pigmented tumors
The Cancer Genome Atlas (TCGA) is a National Institute of Health
of spindled and epithelioid
melanocytes after 10 (NIH)-sponsored cooperative effort to comprehensively analyze various
weeks cancer types. The TCGA published results on a collaborative, multi-platform
analysis of 333 melanomas with a high percentage of metastatic melano-
mas (80%, mostly regional lymph node metastases) representing the largest
study of melanoma to date.111 This study categorized melanoma by DNA
minority of atypical cellular blue nevi showing one to two chromosomal mutations, copy number alterations, gene expression (RNA), non-coding
aberrations.88,91 Blue nevus-like melanomas exhibit multiple chromosomal RNA, methylation, protein expression, and histologic images linked to a
aberrations (typically ≥ 3) including recurrent deletions of chromosomes 1p, database with clinical annotation and follow-up. In this analysis, melano-
3p, 4q, 6q, 8p, 9, 16, and 17q and recurrent gains of chromosomes 6p, 8q, mas were classified into four types: BRAF, RAS, NF1, and triple-wild type
20, and 21q.88,91 (WT). The first three types had > 90% UV signature mutation profile with
Another signaling pathway implicated in the pathogenesis in tumors high-frequency TERT promoter mutations corresponding to the low-CSD
that share phenotypic features with blue nevi is the protein kinase A and high-CSD melanoma classes. The triple WT melanoma mutation profile
(PKA) pathway. Loss-of-function mutations in a regulatory subunit of PKA was only 30% UV signature with < 10% TERT promoter mutation and
(encoded by PRKAR1A) are found in epithelioid blue nevi associated with had more copy number changes and fusion driver events, corresponding
Carney complex and result in hyperactivation of PKA with subsequent to the category of acral and mucosal melanomas. RNA expression analy-
activation of the MITF transcription factor.92 Such mutations have also sis identified three clusters with significantly different survival rates, with
been detected in a subset of tumors designated as pigmented epithelioid the ‘immune’ signature subgroup having the most favorable survival rates
melanocytomas.93 and the ‘keratin’ signature subgroup showing an adverse prognosis.111 This
and additional studies demonstrating prognostic value from histologic doc-
Spitzoid melanoma umentation of lymphocytic infiltrates as well as expression profiling of the
Spitzoid melanocytic neoplasms represent a class of neoplasia with a range immune response in these tumors have been reported as well.112 A separate
of malignant potential from Spitz nevi at the benign end to spitzoid mel- TCGA study of primary uveal melanoma cases (n = approximately 80%)
anoma at the malignant end, and atypical Spitz tumors as a biologically demonstrated the near universal detection of GNAQ and GNA11 muta-
intermediate category. Spitz nevi are defined by distinct histopathological tions as well as poor prognosis associated with chromosome 3 monosomy
features and an age distribution skewed towards children. They have a dis- (loss of the tumor suppressor BAP1) on chromosome 3p.90 In addition,
tinctive mutational landscape that distinguishes them from common nevi, EIF1AX and SF3B1 mutations were associated with distinct copy number
which include activating mutations in HRAS and kinase fusions of ROS1, alteration and methylation patterns within the chromosome 3 disomy group
NTRK1, NTRK3, ALK, BRAF, RET, or MET.94–97 Spitz nevi tend to have that corresponded to better prognosis.
no chromosomal aberrations with the exception of lesions with isolated
chromosome 11p gain, which is typically associated with an oncogenic Summary
HRAS mutation on the duplicated chromosomal arm.94 Some Spitz nevi can Genetic analyses have revealed that deregulation of a few key signaling
also have isolated copy number increase of distal chromosome 7q, which pathways is critical in melanoma formation. Common key events include
indicates fusions of BRAF or MET mapping in this area.96,97 activating mutations in the MAP kinase (proliferation) and PI3 kinase (sur-
Atypical Spitz tumors have similar oncogenic alterations as Spitz nevi, vival) pathways and inactivating mutations in the RB1 pathway (cell cycle
but have some additional chromosomal aberrations, often with loss of control). Kinase activation can occur at the receptor level (KIT mutations
chromosome 9. Spitzoid melanoma, when defined by solely by histologic and fusions of receptor tyrosine kinases such as MET, RET, ALK, ROS1,

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
1362 Melanoma

NTRK1, NTRK3) often activating both the MAP kinase and PI3 kinase melanoma, blue nevi, and related tumors have highlighted additional signal-
pathways or further downstream (NRAS, PTEN, BRAF, PI3KCA) acti- ing pathways of relevance in subsets of melanocytic neoplasia. The advent
vation of a pathway. The mutation patterns often correlate with the ana- of high-throughput genomic tools has initiated the construction of sets of
tomic site of the primary tumor and clinical and histologic features. These critical genetic alterations in melanoma which will in time complete the
driver mutations initiate proliferation in early stages of melanocytic neo- foundation for comprehensive classification of melanoma that will continue
plasia and are followed by loss of cell cycle checkpoint inhibition (e.g., to integrate genomic alterations with established phenotypes and is expected
p16, Rb), dysregulation of chromatin remodeling proteins (e.g., ARID1/2), to significantly improve prognostication and treatment stratification.
and protection from replicative senescence through TERT gene amplifica-
tion or TERT promoter mutation. Mutations in GNAQ/GNA11 in uveal Access ExpertConsult.com for the complete list of references

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
References 1362.e1

REFERENCES
31. Kopf AW, Bart RS, Hennessy P. Congenital and nevocytic nevi and melanoma. J
Clinical features Am Acad Dermatol. 1979;1:123–130.
1. Boring CC, Squires TS, Tong T. Cancer statistics, 1991. CA Cancer J Clin. 32. Lorentzen M, Peis M, Bretteville-Jensen G. Incidence of malignant transforma-
1991;41:19–36. tion in giant pigmented nevi. Scand J Plast Reconstr Surg. 1977;11:163–167.
2. McHenry PM, Hole DJ, Mackie RM. Melanoma in people aged 65 and over in 33. Barnhill RL, Mihm MC. The histopathology of cutaneous melanoma. Semin
Scotland 1979-1989. BMJ. 1992;304:746–749. Diagn Pathol. 1993;10:47–75.
3. Mackie RM, Watt D, Doherty V, et al. Melanoma occurring in those aged under 34. Benisch B, Peison B, Kannerstein M, et al. Melanoma originating from intrader-
30 in the West of Scotland 1979-1986: a study of incidence, clinical features, mal nevi: a clinicopathologic entity. Arch Dermatol. 1980;116:696–698.
pathological features and survival. Br J Dermatol. 1991;124:560–564. 35. Okun MR, Bauman L. Malignant melanoma arising from an intradermal nevus.
4. Barnhill RL. Pathology and prognostic factors. Curr Opin Oncol. 1993;5: Arch Dermatol. 1965;92:69–72.
364–376. 36. Shain AH, Yeh I, Kovalyshyn I, et al. The genetic evolution of melanoma from
5. De Vries E, Schouten LJ, Visser O, et al. Rising trends in the incidence and mor- precursor lesions. N Engl J Med. 2015;373:1926–1936.
tality from cutaneous melanoma in the Netherlands: a Northwest to Southwest 37. Garbe C, Orfanos CE. Epidemiology of melanoma in central Europe: risk
gradient. Eur J Cancer. 2003;39:1439–1446. factors and prognostic predictors. Results of the Central Melanoma Regis-
6. Jemal A, Siegel R, Xu J, et al. Cancer statistics, 2010. CA Cancer J Clin. try of the German Dermatological Society. Pigment Cell Res. 1992;(suppl 2):
2010;60:277–300. 285–294.
7. Ward EM, Thun MJ, Hannan LM, et al. Interpreting cancer trends. Ann N Y 38. Morales Suárez-Varela MM, Llopis González A, Lacasana Navarro M, et al.
Acad Sci. 2006;1076:29–53. Nevus and malignant cutaneous melanoma. Rev Sanid Hig Publica (Madr).
8. Jemal A, Ward EM, Johnson CJ, et al. Annual report to the nation on the status 1991;65:233–238.
of cancer, 1975-2014, featuring survival. J Natl Cancer Inst. 2017;109(9). 39. Stolz W, Schmoeckel C, Landthaler M, et al. Association of early melanoma
9. Simard EP, Ward EM, Siegel R, et al. Cancers with increasing incidence trends with nevocytic nevi. Cancer. 1989;63:550–555.
in the United States: 1999 through 2008. CA Cancer J Clin. 2012;62:118–128. 40. Marks R, Dorevitch AP, Mason G. Do all melanomas come from ‘moles’. A
10. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2017. CA Cancer J Clin. study of the histological association between melanocytic nevi and melanoma.
2017;67:7–30. Australas J Dermatol. 1990;31:77–80.
11. Chang AE, Karnell LH, Menck HR. The national cancer database report on 41. Shitara D, Tell-Martí G, Badenas C, et al. Mutational status of naevus-associ-
cutaneous and noncutaneous melanoma. Cancer. 1998;83:1664–1678. ated melanomas. Br J Dermatol. 2015;173:671–680.
12. Wingo PA, Ries LAG, Rosenberg HM, et al. Cancer incidence and mortality. 42. Betti R, Santambrogio R, Cerri A, et al. Observational study on the mitotic
Cancer. 1998;82:1197–1207. rate and other prognostic factors in cutaneous primary melanoma arising
13. Clarke CA, McKinley M, Hurley S, et al. Continued increase in melanoma inci- from naevi and from melanoma de novo. J Eur Acad Dermatol Venereol.
dence across all socioeconomic status groups in California, 1998-2012. J Invest 2014;28:1738–1741.
Dermatol. 2017;137:2282–2290. 43. DeDavid M, Orlow SJ, Provost N, et al. A study of large congenital melano-
14. Landis SH, Murray T, Bolden S, et al. Cancer statistics, 1998. CA Cancer J cytic nevi and associated malignant melanomas: review of cases in the New
Clin. 1998;48:6–29. York University Registry and the world literature. J Am Acad Dermatol.
15. Rigel DS. Malignant melanoma: incidence issues and their effect on diagnosis 1997;36:428–433.
and treatment in the 1990s. Mayo Clin Proc. 1997;72:367–371. 44. Tannous ZS, Mihm MC Jr, Sober AJ, et al. Congenital melanocytic nevi: clinical
16. Ceballos P, Ruiz-Maldonado R, Mihm MC Jr. Melanoma in children. N Engl J and histopathologic features, risk of melanoma, and clinical management. J Am
Med. 1995;332:656–662. Acad Dermatol. 2005;52:197–203.
17. Rao BN, Hayes FA, Pratt CB, et al. Melanoma in children: its management and 45. Vergier B, Laharanne E, Prochazkova-Carlotti M, et al. Proliferative nodules vs
prognosis. J Pediatr Surg. 1990;25:198–203. melanoma arising in giant congenital melanocytic nevi during childhood. JAMA
18. Milton GW, Shaw HM, Thompson JF, et al. Cutaneous melanoma in childhood: Dermatol. 2016;152:1147–1151.
incidence and prognosis. Australas J Dermatol. 1997;38:S44–S48. 46. Augustsson A. Melanocytic nevi, melanoma and sun exposure. Acta Derm
19. Scalzo DA, Hida CA, Toth G, et al. Childhood melanoma: a clinicopathological Venereol. 1991;166(suppl):1–34.
study of 22 cases. Melanoma Res. 1997;7:63–68. 47. Sample A, He YY. Mechanisms and prevention of UV-induced melanoma. Pho-
20. Spatz A, Ruiter D, Hardmeier T, et al. Melanoma in childhood: an EORTC- todermatol Photoimmunol Photomed. 2018;34:13–24.
MCG multicenter study on the clinico-pathological aspects. Int J Cancer. 48. Armstrong BK, Cust AE. Sun exposure and skin cancer, and the puzzle of
1996;68:317–324. cutaneous melanoma: A perspective on Fears et al. Mathematical models
21. Saiyed FK, Hamilton EC, Austin MT. Pediatric melanoma: incidence, treatment, of age and ultraviolet effects on the incidence of skin cancer among whites
and prognosis. Pediatric Health Med Ther. 2017;8:39–45. in the United States. Am J Epidemiol. 1977;105:420–427. Cancer Epidemiol.
22. Bahrami A, Barnhill RL. Pathology and genomics of pediatric melanoma: A 2017;48:147–156.
critical reexamination and new insights. Pediatr Blood Cancer. 2018;65. 49. Rünger TM. Mechanisms of melanoma promotion by ultraviolet radiation. J
23. Stefanaki C, Chardalias L, Soura E, et al. Paediatric melanoma. J Eur Acad Invest Dermatol. 2016;136:1751–1752.
Dermatol Venereol. 2017;31:1604–1615. 50. Berwick M, Wiggins C. The current epidemiology of cutaneous malignant mel-
24. Linabery AM, Ross JA. Trends in childhood cancer incidence in the U.S. (1992- anoma. Front Biosci. 2006;11:1244–1254.
2004). Cancer. 2008;112:416–432. 51. Moan J. Solar radiation and melanomas: is there any doubt about the connec-
25. Tracy ET, Aldrink JH. Pediatric melanoma. Semin Pediatr Surg. tion. Tidsskr Nor Laegeforen. 1998;118:2321–2325.
2016;25:290–298. 52. Ley RD. Ultraviolet radiation A-induced precursors of cutaneous melanoma in
26. Ward E, DeSantis C, Robbins A, et al. Childhood and adolescent cancer statis- Monodelphis domestica. Cancer Res. 1997;57:3682–3684.
tics, 2014. CA Cancer J Clin. 2014;64:83–103. 53. Lea CS, Scotto JA, Buffler PA, et al. Ambient UVB and melanoma risk in the
27. Lewis KG. Trends in pediatric melanoma mortality in the United States, 1968 United States: a case-control analysis. Ann Epidemiol. 2007;17:447–453.
through 2004. Dermatol Surg. 2008;34:152–159. 54. Khan AQ, Travers JB, Kemp MG. Roles of UVA radiation and DNA damage
28. Paradela S, Fonseca E, Pita-Fernández S, et al. Prognostic factors for melanoma responses in melanoma pathogenesis. Environ Mol Mutagen. 2018;59:
in children and adolescents: a clinicopathologic, single-center study of 137 438–460.
patients. Cancer. 2010;116:4334–4344. 55. Pudroma X, Duoji G, Grigalavicius M, et al. Molecular mechanisms of UVA-in-
29. Stephenson HE, Terry CW, Lukens JN, et al. Immunologic factors in human duced melanoma. J Environ Pathol Toxicol Oncol. 2017;36:217–228.
melanoma ‘metastatic’ to products of gestation (with exchange transfusion of 56. Hoel DG, Berwick M, de Gruijl FR, et al. The risks and benefits of sun expo-
infant to mother). Surgery. 1971;69:515–522. sure 2016. Dermatoendocrinol. 2016;8.
30. Gruber SB, Barnhill RL, Stenn KS, et al. Nevomelanocytic proliferations in 57. Reichrath J, Rass K. Ultraviolet damage, DNA repair and vitamin D in nonmel-
association with cutaneous melanoma: a multivariate analysis. J Am Acad Der- anoma skin cancer and in malignant melanoma: an update. Adv Exp Med Biol.
matol. 1989;21:773–780. 2014;810:208–233.

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
1362.e2 References

58. Slominski AT, Brożyna AA, Zmijewski MA, et al. Vitamin D signaling and mel- 88. Pollock PM, Spurr N, Bishop T, et al. Haplotype analysis of two recurrent
anoma: role of vitamin D and its receptors in melanoma progression and man- CDKN2A mutations in ten melanoma families: evidence for common founders
agement. Lab Invest. 2017;97:706–724. and independent mutations. Hum Mutat. 1998;11:424–431.
59. Kraemer KH, DiGiovanna JJ. Xeroderma pigmentosum. In: Adam MP, Ardinger 89. Zuo L, Weger J, Yang Q, et al. Germline mutations in the p16INK4a binding
HH, Pagon RA, et al, eds. GeneReviews® [Internet]. Seattle (WA): University of domain of CDK4 in familial melanoma. Nat Genet. 1996;12:97–99.
Washington; 2003. [updated 2016 Sep 29]. 90. Lin J, Hocker TL, Singh M, et al. Genetics of melanoma predisposition. Br J
60. Armstrong BK. Epidemiology of melanoma: intermittent or total accumulated Dermatol. 2008;159:286–291.
exposure to the sunfi. J Dermatol Surg Oncol. 1988;14:835–849. 91. Soura E, Eliades PJ, Shannon K, et al. Hereditary melanoma: Update on syn-
61. Chang YM, Barrett JH, Bishop DT, et al. Sun exposure and melanoma risk at dromes and management: emerging melanoma cancer complexes and genetic
different latitudes: a pooled analysis of 5700 cases and 7216 controls. Int J counseling. J Am Acad Dermatol. 2016;74:411–420, quiz 421–422.
Epidemiol. 2009;38:814–830. 92. Puntervoll HE, Yang XR, Vetti HH, et al. Melanoma prone families with CDK4
62. Kricker A, Armstrong BK, Goumas C, et al. Ambient UV, personal sun germline mutation: phenotypic profile and associations with MC1R variants. J
exposure and risk of multiple primary melanomas. Cancer Causes Control. Med Genet. 2013;50:264–270.
2007;18:295–304. 93. Demenais F, Mohamdi H, Chaudru V, et al. Association of MC1R variants
63. Lancaster HO. Some geographical aspects of the mortality from melanoma in and host phenotypes with melanoma risk in CDKN2A mutation carriers: a
Europeans. Med J Aust. 1956;1:1082–1087. GenoMEL study. J Natl Cancer Inst. 2010;102:1568–1583.
64. Scheibner A, Milton GW, McCarthy WH, et al. Multiple primary melanoma: 94. Star P, Goodwin A, Kapoor R, et al. Germline BAP1-positive patients: the
review of 90 cases. Australas J Dermatol. 1982;23:1–8. dilemmas of cancer surveillance and a proposed interdisciplinary consensus
65. Kang S, Barnhill RL, Mihm MC, et al. Multiple primary cutaneous melanomas. monitoring strategy. Eur J Cancer. 2018;92:48–53.
Cancer. 1992;70:1911–1916. 95. Pilarski R, Rai K, Cebulla C, et al. BAP1 tumor predisposition syndrome.
66. Schuurman MS, de Waal AC, Thijs EJM, et al. Risk factors for second In: Adam MP, Ardinger HH, Pagon RA, et al, eds. GeneReviews® [Internet].
primary melanoma among Dutch patients with melanoma. Br J Dermatol. Seattle (WA): University of Washington; 2016.
2017;176:971–978. 96. Gupta MP, Lane AM, DeAngelis MM, et al. Clinical characteristics of uveal
67. Chen T, Fallah M, Försti A, et al. Risk of next melanoma in patients with famil- melanoma in patients with germline BAP1 mutations. JAMA Ophthalmol.
ial and sporadic melanoma by number of previous melanomas. JAMA Derma- 2015;133:881–887.
tol. 2015;151:607–615. 97. Artomov M, Stratigos AJ, Kim I, et al. Rare variant, gene-based association
68. Caini S, Boniol M, Botteri E, et al. The risk of developing a second primary study of hereditary melanoma using whole-exome sequencing. J Natl Cancer
cancer in melanoma patients: a comprehensive review of the literature and Inst. 2017;109.
meta-analysis. J Dermatol Sci. 2014;75:3–9. 98. Soura E, Eliades PJ, Shannon K, et al. Hereditary melanoma: Update on syn-
69. Driscoll MS, Grant-Kels JM. Melanoma and pregnancy. G Ital Dermatol Vene- dromes and management: Genetics of familial atypical multiple mole melanoma
reol. 2008;143:251–257. syndrome. J Am Acad Dermatol. 2016;74:395–407, quiz 408–410.
70. Todd SP, Driscoll MS. Prognosis for women diagnosed with melanoma during, 99. Weinstock MA, Morris BT, Lederman JS, et al. Effect of BANS located on the
before, or after pregnancy: weighing the evidence. Int J Womens Dermatol. prognosis of clinical stage 1 melanoma: new data and meta-analysis. Br J Der-
2017;3, 26-29. matol. 1988;119:559–565.
71. Still R, Brennecke S. Melanoma in pregnancy. Obstet Med. 2017;10:107–112. 100. American Academy of Dermatology Ad Hoc Task Force for the ABCDEs of
72. Travers RL, Sober AJ, Berwick M, et al. Increased thickness of pregnancy-asso- Melanoma, Tsao H, Olazagasti JM, et al. Early detection of melanoma: review-
ciated melanoma. Br J Dermatol. 1995;132:876–883. ing the ABCDEs. J Am Acad Dermatol. 2015;72:717–723.
73. O’Meara AT, Cress R, Xing G, et al. Malignant melanoma in pregnancy. A 101. Weyers W, Euler M, Diaz-Cascajo C, et al. Classification of cutaneous malig-
population-based evaluation. Cancer. 2005;103:1217–1226. nant melanoma: a reassessment of histopathologic criteria for the distinction of
74. Roh MR, Eliades P, Gupta S, et al. Cutaneous melanoma in women. Int J different types. Cancer. 1999;86:288–299.
Womens Dermatol. 2017;3:S11–S15. 102. Viros A, Fridlyand J, Bauer J, et al. Improving melanoma classification by inte-
75. Kyrgidis A, Lallas A, Moscarella E, et al. Does pregnancy influence melanoma grating genetic and morphologic features. PLoS Med. 2008;5:e120.
prognosis? A meta-analysis. Melanoma Res. 2017;27:289–299. 103. Bastian BC. The molecular pathology of melanoma: an integrated taxonomy of
76. Jones MS, Lee J, Stern SL, et al. Is pregnancy-associated melanoma associated melanocytic neoplasia. Annu Rev Pathol. 2014;9:239–271.
with adverse outcomes? J Am Coll Surg. 2017;225:149–158. 104. Bittner M, Meltzer P, Chen Y, et al. Molecular classification of cutaneous malig-
77. Khosrotehrani K, Olsen CM, Byrom L, et al. Melanoma during preg- nant melanoma by gene expression profiling. Nature. 2000;406:536–540.
nancy: level of evidence and principles of precaution. J Am Acad Dermatol. 105. Koh SS, Opel ML, Wei JP, et al. Molecular classification of melanomas and
2017;76:e29–e30. nevi using gene expression microarray signatures and formalin-fixed and paraf-
78. Martires KJ, Stein JA, Grant-Kels JM, et al. Meta-analysis concerning mor- fin-embedded tissue. Mod Pathol. 2009;22:538–546.
tality for pregnancy-associated melanoma. J Eur Acad Dermatol Venereol. 106. Duncan LM. The classification of cutaneous melanoma. Hematol Oncol Clin
2016;30:e107–e108. North Am. 2009;23:501–513, ix.
79. Byrom L, Olsen CM, Khosrotehrani K, et al. Reply to Meta-analysis concerning 107. Clark WH, From L, Bernardino EA, et al. The histogenesis and biologic behav-
mortality for pregnancy-associated melanoma. J Eur Acad Dermatol Venereol. ior of primary human melanomas of the skin. Cancer Res. 1969;29:705–726.
2016;30:e106–e107. 108. Clark WH Jr, Elder DE, Van Horn M. The biologic forms of melanoma. Hum
80. Byrom L, Olsen C, Knight L, et al. Increased mortality for pregnancy-associated Pathol. 1986;17:443–450.
melanoma: systematic review and meta-analysis. J Eur Acad Dermatol Vene- 109. McGovern VJ, Mihm MC, Bailly C, et al. The classification of melanoma and
reol. 2015;29:1457–1466. its histologic reporting. Cancer. 1973;32:1446–1457.
81. Byrom L, Olsen CM, Knight L, et al. Does pregnancy after a diagnosis of mel- 110. Elder DE, Jucovy P, Tuthill RJ, et al. The classification of melanoma. Am J
anoma affect prognosis? Systematic review and meta-analysis. Dermatol Surg. Dermatopathol. 1980;2:315–320.
2015;41:875–882. 111. Heenan PJ, Armstrong BK, English DE. Pathobiological and epidemiological
82. Driscoll MS, Martires K, Bieber AK, et al. Pregnancy and melanoma. J Am variants of cutaneous melanoma. Basel: Karger; 1987:107–146.
Acad Dermatol. 2016;75:669–678. 112. McGovern VJ. The classification of melanoma and its relationship with progno-
83. Tellez A, Rueda S, Conic RZ, et al. Risk factors and outcomes of cutane- sis. Pathology. 1970;2:85–98.
ous melanoma in women less than 50 years of age. J Am Acad Dermatol. 113. Elder DE. Pathology of melanoma. Surg Oncol Clin N Am. 2015;24:229–237.
2016;74:731–738. 114. Elder DE. Melanoma progression. Pathology. 2016;48:147–154.
84. Martires KJ, Pomeranz MK, Stein JA, et al. Pregnancy-associated melanoma 115. Knudsen EA, Osterlind AL. Malignant lentigo. Ugeskrift Laeger.
(PAMM): Is there truly a worse prognosis? Would not sound alarm bells just 1994;156:4221–4223.
yet. J Am Acad Dermatol. 2016;75:e77. 116. Kallini JR, Jain SK, Khachemoune A. Lentigo maligna: review of salient charac-
85. Bain C, Hennekens CH, Speizer FE, et al. Oral contraceptive use and mela- teristics and management. Am J Clin Dermatol. 2013;14:473–480.
noma. J Natl Cancer Inst. 1982;68:537–539. 117. Reed JA, Shea CR. Lentigo maligna: melanoma in situ on chronically sun-dam-
86. Greene MH. The genetics of hereditary melanoma and nevi. Cancer. aged skin. Arch Pathol Lab Med. 2011;135:838–841.
1999;86:2464–2477. 118. Wessman LL, Andersen LK, Davis MDP. Incidence of diseases primarily affect-
87. Nelson AA, Tsao H. Melanoma and genetics. Clin Dermatol. 2009;27:46–52. ing the skin by age group: population-based epidemiologic study in Olmsted

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
References 1362.e3

County, Minnesota, and comparison with age-specific incidence rates world- 150. Knezevich SR, Barnhill RL, Elder DE, et al. Variability in mitotic figures in
wide. Int J Dermatol. 2018;[Epub ahead of print]. serial sections of thin melanomas. J Am Acad Dermatol. 2014;71:1204–1211.
119. Newell GR, Sider JG, Bergfeldt L, et al. Incidence of cutaneous melanoma 151. Elder DE. Pathological staging of melanoma. Methods Mol Biol.
in the U.S. by histology with special reference to the face. Cancer Res. 2014;1102:325–351.
1988;48:5036–5041. 152. Elder DE, Piepkorn M, Barnhill RL, et al. Pathologist characteristics associated
120. Higgins HW 2nd, Lee KC, Galan A, et al. Melanoma in situ: Part I. Epidemi- with accuracy and reproducibility of melanocytic skin lesion interpretation. J
ology, screening, and clinical features. J Am Acad Dermatol. 2015;73:181–190, Am Acad Dermatol. 2018;[Epub ahead of print].
quiz 191–192. 153. Geller BM, Frederick PD, Knezevich SR, et al. Pathologists’ use of second opin-
121. Tannous ZS, Lerner LH, Duncan LM, et al. Progression to invasive mela- ions in interpretation of melanocytic cutaneous lesions: policies, practices, and
noma from malignant melanoma in situ, lentigo maligna type. Hum Pathol. perceptions. Dermatol Surg. 2018;44:177–185.
2000;31:705–708. 154. Elmore JG, Barnhill RL, Elder DE, et al. Pathologists’ diagnosis of invasive mel-
122. Horn-Ross PL, Holly E, Brown SR, et al. Temporal trends in the incidence of anoma and melanocytic proliferations: observer accuracy and reproducibility
cutaneous melanoma among Caucasians in the San Francisco-Oakland MSA. study. BMJ. 2017;357:j2813.
Cancer Causes Control. 1991;2:299–305. 155. Gerami P, Busam K, Cochran A, et al. Histomorphologic assessment and
123. MacKie RM. Malignant melanocytic tumors. J Cutan Pathol. 1985;12:251–265. interobserver diagnostic reproducibility of atypical spitzoid melanocytic neo-
124. Bradford PT, Goldstein AM, McMaster ML, et al. Acral lentiginous melanoma: plasms with long-term follow-up. Am J Surg Pathol. 2014;38:934–940.
incidence and survival patterns in the United States, 1986-2005. Arch Derma- 156. Muhlbauer A, Momtahen S, Mihm MC, et al. The correlation of the standard
tol. 2009;145:427–434. 5 probe FISH assay with melanocytic tumors of uncertain malignant potential.
125. Goydos JS, Shoen SL. Acral lentiginous melanoma. Cancer Treat Res. Ann Diagn Pathol. 2017;28:30–36.
2016;167:321–329. 157. Lott JP, Elmore JG, Zhao GA, et al. Evaluation of the Melanocytic Pathol-
126. Kim SY, Yun SJ. Cutaneous melanoma in Asians. Chonnam Med J. ogy Assessment Tool and Hierarchy for Diagnosis (MPATH-Dx) classifica-
2016;52:185–193. tion scheme for diagnosis of cutaneous melanocytic neoplasms: Results from
127. Stubblefield J, Kelly B. Melanoma in non-caucasian populations. Surg Clin the International Melanoma Pathology Study Group. J Am Acad Dermatol.
North Am. 2014;94:1115–1126, ix. 2016;75:356–363.
128. Reed R. Acral lentiginous melanoma. New concepts in surgical pathology of the 158. Clark WH, Mihm MC. Lentigo maligna and lentigo maligna melanoma. Am J
skin. New York: Wiley; 1976:89–90. Pathol. 1969;55:39–67.
129. Feibleman CE, Stoll H, Maize JC. Melanomas of the palm, sole and nail bed: a 159. Curtin JA, Fridlyand J, Kageshita T, et al. Distinct sets of genetic alterations in
clinicopathologic study. Cancer. 1980;46:2492–2504. melanoma. N Engl J Med. 2005;353:2135–2147.
130. Blessing K, Kernohan NM, Park KGM. Subungual melanoma: clinicopatholog- 160. North JP, Kageshita T, Pinkel D, et al. Distribution and significance of occult
ical features of 100 cases. Histopathology. 1991;19:425–429. intraepidermal tumor cells surrounding primary melanoma. J Invest Dermatol.
131. Panizzon R, Krebs A. Subungual melanoma. Der Hautarzt. 1980;31:132–140. 2008;128:2024–2030.
132. Sondergaard K, Olsen G. Melanoma of the foot: clinicopathologic study 161. Allan SJR, Dicker AJ, Tidman M, et al. Amelanotic lentigo maligna and amela-
of 125 primary cutaneous melanomas. Acta Pathol Microbiol Scand [A]. notic lentigo maligna melanoma: a report of three cases mimicking intraepider-
1980;88:275–283. mal squamous carcinoma. J Eur Acad Dermatol Venereol. 1998;11:78–81.
133. Curtin JA, Fridlyand J, Kageshita T, et al. Distinct sets of genetic alterations in 162. Rocamora V, Puig L, Romani J, et al. Amelanotic lentigo maligna melanoma:
melanoma. N Engl J Med. 2005;353:2135–2147. report of a case and review of the literature. Cutis. 1999;64:53–56.
134. Whiteman DC, Pavan WJ, Bastian BC. The melanomas: a synthesis of epidemi- 163. Conrad N, Jackson B, Goldberg L. Amelanotic lentigo maligna melanoma: a
ological, clinical, histopathological, genetic, and biological aspects, supporting unique case presentation. Dermatol Surg. 1999;25:408–411.
distinct subtypes, causal pathways, and cells of origin. Pigment Cell Melanoma 164. Abdulla FR, Kerns MJ, Mutasim DF. Amelanotic lentigo maligna: a report
Res. 2011;24:879–897. of three cases and review of the literature. J Am Acad Dermatol. 2010;62:
135. Greenwald HS, Friedman EB, Osman I. Superficial spreading and nodular mela- 857–860.
noma are distinct biological entities: a challenge to the linear progression model. 165. Robinson JK. Margin control for lentigo maligna. J Am Acad Dermatol.
Melanoma Res. 2012;22:1–8. 1994;31:79–85.
136. Baumert J, Schmidt M, Giehl KA, et al. Time trends in tumour thickness vary 166. Arrington JH, Reed RJ, Ichinose H, et al. Plantar lentiginous melanoma: a
in subgroups: analysis of 6475 patients by age, tumour site and melanoma distinctive variant of human cutaneous melanoma. Am J Surg Pathol. 1977;1:
subtype. Melanoma Res. 2009;19:24–30. 131–143.
137. Feller L, Khammissa RAG, Lemmer J. A review of the aetiopathogenesis 167. Jimbow K, Takahashi H, Miura S, et al. Biological behavior and natural cause of
and clinical and histopathological features of oral mucosal melanoma. Scien- acral melanoma. Clinical and histological features and prognosis of palmoplan-
tificWorldJournal. 2017;2017:9189812. tar, subungual and other acral melanomas. Am J Dermatopathol. 1984;6(suppl
138. Ascierto PA, Accorona R, Botti G, et al. Mucosal melanoma of the head and I):43–53.
neck. Crit Rev Oncol Hematol. 2017;112:136–152. 168. Kuchelmeister C, Schaumburg-Lever G, Garbe C. Acral cutaneous melanoma in
139. Mihajlovic M, Vlajkovic S, Jovanovic P, et al. Primary mucosal melanomas: a Caucasians: clinical features, histopathology and prognosis in 112 patients. Br J
comprehensive review. Int J Clin Exp Pathol. 2012;5:739–753. Dermatol. 2000;143:275–280.
140. Iversen K, Robins RE. Mucosal melanomas. Am J Surg. 1980;139:660–664. 169. Phan A, Touzet S, Dalle S, et al. Acral lentiginous melanoma: histopathological
141. Wanebo HJ, Woodruff JM, Farr GH, et al. Anorectal melanoma. Cancer. prognostic features of 121 cases. Br J Dermatol. 2007;157:311–318.
1981;47:1891–1900. 170. Fernandez-Flores A, Cassarino DS. Histopathological diagnosis of acral lentigi-
142. Sun S, Huang X, Gao L, et al. Long-term treatment outcomes and prognosis of nous melanoma in early stages. Ann Diagn Pathol. 2017;26:64–69.
mucosal melanoma of the head and neck: 161 cases from a single institution. 171. Belter B, Haase-Kohn C, Pietzsch J. Biomarkers in malignant melanoma: recent
Oral Oncol. 2017;74:115–122. trends and critical perspective. In: Ward WH, Farma JM, eds. Cutaneous mel-
143. Heppt MV, Roesch A, Weide B, et al. Prognostic factors and treatment out- anoma: etiology and therapy [Internet]. Brisbane (AU): Codon Publications;
comes in 444 patients with mucosal melanoma. Eur J Cancer. 2017;81:36–44. 2017:[Chapter 3].
144. Clark WH Jr. A classification of melanoma in man correlated with histogenesis 172. Sholl LM, Andea A, Bridge JA, et al. Template for reporting results of bio-
and biological behavior. Advances in the biology of the skin. The pigmentary marker testing of specimens from patients with melanoma. Arch Pathol Lab
system VIII. Oxford: Pergamon Press; 1967:621–647. Med. 2016;140:355–357.
145. Clark WH Jr, Elder DE, Guerry DIV, et al. Model predicting survival in stage I 173. Zager JS, Gastman BR, Leachman S, et al. Performance of a prognostic 31-gene
melanoma based on tumor progression. J Natl Cancer Inst. 1989;81:1893–1904. expression profile in an independent cohort of 523 cutaneous melanoma
146. Elder DE. Pathology of melanoma. Clin Cancer Res. 2006;12:2308s–2311s. patients. BMC Cancer. 2018;18:130.
147. Gimotty PA, Van Belle P, Elder DE, et al. Biologic and prognostic significance of 174. Clark WH Jr, Evans HL, Everett MA, et al. Early melanoma: histologic terms.
dermal Ki67 expression, mitoses, and tumorigenicity in thin invasive cutaneous Am J Dermatopathol. 1991;13:579–658.
melanoma. J Clin Oncol. 2005;23:8048–8056. 175. Duncan LM. Prognostic indicators in melanoma. Adv Dermatol. 1999;15:
148. Mihic-Probst D, Shea C, Duncan L, et al. Update on thin melanoma: outcome 489–517.
of an international workshop. Adv Anat Pathol. 2016;23:24–29. 176. Zettersten E, Shaikh L, Ramirez R, et al. Prognostic factors in primary cutane-
149. Elder DE. Thin melanoma. Arch Pathol Lab Med. 2011;135:342–346. ous melanoma. Surg Clin N Am. 2003;83:61–75.

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
1362.e4 References

177. Scoggins CR, Ross MI, Reintgen DS, et al. Gender-related differences in outcome 206. Gershenwald JE, Scolyer RA, Hess KR, et al. Melanoma of the skin. In: Amin
for melanoma patients. Ann Surg. 2006;243:693–698, discussion 698–700. MB, Edge SB, Greene FL, et al, eds. AJCC Cancer Staging Manual. 8th ed. New
178. Erturk K, Tas F. Effect of biology on the outcome of female melanoma patients. York: Springer International Publishing; 2017:563–585.
Mol Clin Oncol. 2017;7:1093–1100. 207. Murali R, Hughes MT, Fitzgerald P, et al. Interobserver variation in the his-
179. Nosrati A, Wei ML. Sex disparities in melanoma outcomes: the role of biology. topathologic reporting of key prognostic parameters, particularly clark
Arch Biochem Biophys. 2014;563:42–50. level, affects pathologic staging of primary cutaneous melanoma. Ann Surg.
180. Joosse A, van der Ploeg AP, Haydu LE, et al. Sex differences in melanoma sur- 2009;249:641–647.
vival are not related to mitotic rate of the primary tumor. Ann Surg Oncol. 208. Balch CM, Soong S-J, Gershenwald JE, et al. Prognostic factors analysis of
2015;22:1598–1603. 17,600 patients: validation of The American Joint Committee on Cancer mela-
181. Austin PF, Cruse CW, Lyman MG, et al. Age as a prognostic factor in the malig- noma staging system. J Clin Oncol. 2001;19:3622–3634.
nant melanoma population. Ann Surg Oncol. 1994;1:487–494. 209. Masback A, Olsson H, Westerdahl J, et al. Prognostic factors in invasive
182. Schuchter L, Schultz DJ, Synnestvedt M, et al. A prognostic model for predict- cutaneous melanoma: a population-based study and review. Melanoma Res.
ing 10-year survival in patients with primary melanoma. The pigmented lesion 2001;11:435–445.
group. Ann Intern Med. 1996;125:369–375. 210. Wheless L, Isom CA, Hooks MA, et al. Mitotic rate is associated with pos-
183. Unger JM, Flaherty LE, Liu PV, et al. Gender and other survival predictors itive lymph nodes in patients with thin melanomas. J Am Acad Dermatol.
in patients with metastatic melanoma on Southwest Oncology Group trials. 2018;78:935–941.
Cancer. 2001;91:1148–1155. 211. Bønnelykke-Behrndtz ML, Steiniche T. Ulcerated melanoma: aspects and prog-
184. Lasithiotakis K, Leiter U, Meier F, et al. Age and gender are significant inde- nostic impact. In: Ward WH, Farma JM, eds. Cutaneous melanoma: etiology
pendent predictors of survival in primary cutaneous melanoma. Cancer. and therapy [Internet]. Brisbane (AU): Codon Publications; 2017:[Chapter 5].
2008;112:1795–1804. 212. Eigentler TK, Buettner PG, Leiter U, et al. Impact of ulceration in stages I to
185. Garbe C, Buttner P, Bertz J, et al. Primary cutaneous melanoma. Prognostic III cutaneous melanoma as staged by the American Joint Committee on Cancer
classification of anatomic location. Cancer. 1995;75:2492–2498. Staging System: an analysis of the German Central Malignant Melanoma Regis-
186. Wells KE, Reintgen DS, Cruse CW. The current management and prognosis of try. J Clin Oncol. 2004;22:4376–4383.
acral melanoma. Ann Plast Surg. 1992;28:100–103. 213. Clemente CG, Mihm MC Jr, Bufalino R, et al. Prognostic value of tumor infil-
187. Homsi J, Kashani-Sabet M, Messina JL, et al. Cutaneous melanoma: prognostic trating lymphocytes in the vertical growth phase of primary cutaneous mela-
factors. Cancer Control. 2005;12:223–229. noma. Cancer. 1996;77:1303–1310.
188. Piris A, Mihm MC Jr. Progress in melanoma histopathology and diagnosis. 214. Zettersten E, Sagebiel RW, Miller JR 3rd., et al. Prognostic factors in patients
Hematol Oncol Clin North Am. 2009;23:467–480, viii. with thick cutaneous melanomas (>4 mm). Cancer. 2002;94:1049–1056.
189. Payette MJ, Katz M 3rd, Grant-Kels JM. Melanoma prognostic factors found in 215. Taylor RC, Patel A, Panageas KS, et al. Tumor-infiltrating lymphocytes predict
the dermatopathology report. Clin Dermatol. 2009;27:53–74. sentinel lymph node positivity in patients with cutaneous melanoma. J Clin
190. Batistatou A, Cook MG, Massi D. Histopathology report of cutaneous mela- Oncol. 2007;25:869–875.
noma and sentinel lymph node in Europe: a web-based survey by the Derma- 216. Sinnamon AJ, Sharon CE, Song Y, et al. The prognostic significance of tumor-in-
topathology Working Group of the European Society of Pathology. Virchows filtrating lymphocytes for primary melanoma varies by sex. J Am Acad Derma-
Arch. 2009;454:505–511. tol. 2018;[Epub ahead of print].
191. Haydu LE, Holt PE, Karim RZ, et al. Quality of histopathological reporting 217. Gartrell RD, Marks DK, Hart TD, et al. Quantitative analysis of immune infil-
on melanoma and influence of use of a synoptic template. Histopathology. trates in primary melanoma. Cancer Immunol Res. 2018;[Epub ahead of print].
2010;56:768–774. 218. Mihm MC Jr, Clemente CG, Cascinelli N. Tumor infiltrating lymphocytes in
192. Frishberg DP, Balch C, Balzer BL, et al. Protocol for the examination of lymph node melanoma metastases: a histopathologic prognostic indicator and
specimens from patients with melanoma of the skin. Arch Pathol Lab Med. an expression of local immune response. Lab Invest. 1996;74:43–47.
2009;133:1560–1567. 219. Cancer Genome Atlas Network. Genomic classification of cutaneous melanoma.
193. Ivan D, Prieto VG. An update on reporting histopathologic prognostic factors Cell. 2015;161:1681–1696.
in melanoma. Arch Pathol Lab Med. 2011;135:825–829. 220. Aung PP, Nagarajan P, Prieto VG. Regression in primary cutaneous melanoma:
194. Thompson B, Austin R, Coory M, et al. Completeness of histopathology etiopathogenesis and clinical significance. Lab Invest. 2017;[Epub ahead of
reporting of melanoma in a high-incidence geographical region. Dermatology. print].
2009;218:7–14. 221. Blessing K, McLaren KM. Histological regression in primary cutane-
195. Breslow A. Prognosis in stage I cutaneous melanoma: tumor thickness as a ous melanoma: recognition, prevalence, and significance. Histopathology.
guide to treatment. In: Ackerman AB, ed. Pathology of melanoma. New York: 1992;20:315–322.
Masson Publishing; 1981:313–319. 222. Cooper PH, Wanebo HJ, Hagar RW. Regression in thin melanoma; microscopic
196. Gershenwald JE, Scolyer RA, Hess KR, et al. Melanoma staging: Evidence-based diagnosis and prognostic importance. Arch Dermatol. 1985;21:1127–1131.
changes in the American Joint Committee on Cancer eighth edition cancer 223. Sagebiel R. Regression and other factors of prognostic interest in melanoma.
staging manual. CA Cancer J Clin. 2017;67:472–492. Arch Dermatol. 1985;121:1125–1126.
197. McGovern VJ, Shaw HM, Milton GW. Prognostic significance of a polypoid 224. Gromet MA, Epstein WL, Blois MS. The regressing thin melanoma: a distinctive
configuration in malignant melanoma. Histopathology. 1983;7:663–672. lesion with metastatic potential. Cancer. 1978;42:2282–2292.
198. Breslow A. Thickness, cross-sectioned areas and depth of invasion in the prog- 225. Paladagu RR, Yonemoto RH. Biologic behavior of thin malignant melanomas
nosis of cutaneous melanoma. Ann Surg. 1981;172:902–908. with regressive changes. Arch Surg. 1983;118:41–44.
199. Dvir E, Gellei B, Hirshowitz B. Measurement of tumor thickness as a prognostic 226. McGovern VJ, Shaw HM, Milton GW. Prognosis in patients with thin mela-
tool in primary melanoma of the skin. Ann Plast Surg. 1980;5:216–221. noma: influence of regression. Histopathology. 1983;7:673–680.
200. Consensus NIH. Development Panel on Early Melanoma. Diagnosis and treat- 227. Naruns PL, Nizze JA, Cochran AJ, et al. Recurrence potential of thin primary
ment of early melanoma. J Am Med Assoc. 1992;268:1314–1319. melanomas. Cancer. 1986;57:545–548.
201. Slominski A, Ross J, Mihm MC. Cutaneous melanoma: pathology, relevant 228. Slingluff CL Jr, Vollmer RT, Reintgen DS, et al. Lethal ‘thin’ malignant mela-
prognostic indicators and progression. Br Med Bull. 1995;51:548–569. noma. Ann Surg. 1988;208:150–161.
202. Buzaid AC, Ross MI, Balch CM, et al. Critical analysis of the current American 229. Slingluff CL, Seigler HF. Thin’ malignant melanomas: risk factors and clinical
Joint Committee on Cancer staging system for cutaneous melanoma and pro- management. Ann Plast Surg. 1992;28:89–94.
posal of a new system. J Clin Oncol. 1997;15:1039–1051. 230. Kaur C, Thomas RJ, Desai N, et al. The correlation of regression in primary
203. Balch CM, Buzaid AC, Soong S-J, et al. Final version of the American Joint melanoma with sentinel lymph node status. J Clin Pathol. 2008;61:297–300.
Committee on Cancer staging system for cutaneous melanoma. J Clin Oncol. 231. Olah J, Gyulai R, Korom I, et al. Tumour regression predicts higher risk of
2001;19:3635–3648. sentinel node involvement in thin cutaneous melanomas. Br J Dermatol.
204. Balch CM, Gershenwald JE, Soong SJ, et al. Final version of 2009 AJCC mela- 2003;149:662–663.
noma staging and classification. J Clin Oncol. 2009;27:6199–6206. 232. Tas F, Erturk K. Presence of histological regression as a prognostic factor in
205. Edge BS, Byrd DR, Compton CC, et al. 7th Edition of the AJCC Cancer Staging cutaneous melanoma patients. Melanoma Res. 2016;26:492–496.
Manual. New York: Springer-Verlag, New York; 2009:325–344, [Chapter 31]. 233. Cormier JN, Xing Y, Feng L, et al. Metastatic melanoma to lymph nodes in
Melanoma of the Skin. 2010 American Joint Committee on Cancer. patients with unknown primary sites. Cancer. 2006;106:2012–2020.

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
References 1362.e5

234. Burton AL, Gilbert J, Farmer RW, et al. Regression does not predict nodal 258. Blessing K, McLaren KM, McLean A, et al. Thick malignant melanomas
metastasis or survival in patients with cutaneous melanoma. Am Surg. (greater than 3 mm Breslow) and with good clinical outcome: a histological
2011;77:1009–1013. study and survival analysis. Histopathology. 1991;18:143–148.
235. King R, Googe PB, Mihm MC Jr. Thin melanomas. Clin Lab Med. 259. Spatz A, Shaw HM, Crotty KA, et al. Analysis of histopathological factors asso-
2000;20:713–729. ciated with survival of 10 years or more for patients with thick melanomas
236. Karjalainen JM, Eskelinen J, Nordling S, et al. Mitotic rate and S-phase faction (>5 mm). Histopathology. 1998;33:406–413.
as prognostic factors in stage I cutaneous malignant melanoma. Br J Cancer. 260. Fallowfield ME, Cook MG. The vascularity of primary cutaneous melanoma. J
1998;77:1917–1925. Pathol. 1991;164:241–244.
237. Barnhill RL, Katzen J, Spatz A, et  al. The importance of mitotic rate as a prognos- 261. Barnhill RL, Fandrey K, Levy MA, et al. Angiogenesis and tumor progression
tic factor for localized cutaneous melanoma. J Cutan Pathol. 2005;32:268–273. of melanoma. Quantification of vascularity in melanocytic nevi and cutaneous
238. Francken AB, Shaw HM, Thompson JF, et al. The prognostic importance of malignant melanoma. Lab Invest. 1992;67:331–337.
tumor mitotic rate confirmed in 1317 patients with primary cutaneous mela- 262. Marcoval J, Moreno A, Graells J, et al. Angiogenesis and malignant melanoma.
noma and long follow-up. Ann Surg Oncol. 2004;11:426–433. Angiogenesis is related to the development of vertical (tumorigenic) growth
239. Azzola MF, Shaw HM, Thompson JF, et al. Tumor mitotic rate is a more phase. J Cutan Pathol. 1997;24:212–218.
powerful prognostic indicator than ulceration in patients with primary cuta- 263. Kashani-Sabet M, Sagebiel RW, Ferreira CM, et al. Tumor vascularity in
neous melanoma: an analysis of 3661 patients from a single center. Cancer. the prognostic assessment of primary cutaneous melanoma. J Clin Oncol.
2003;97:1488–1498. 2002;20:1826–1831.
240. Gimotty PA, Elder DE, Fraker DL, et al. Identification of high-risk patients 264. Jour G, Ivan D, Aung PP. Angiogenesis in melanoma: an update with a focus on
among those diagnosed with thin cutaneous melanomas. J Clin Oncol. current targeted therapies. J Clin Pathol. 2016;69:472–483.
2007;25:1129–1134. 265. Dewing D, Emmett M, Pritchard Jones R. The roles of angiogenesis in malig-
241. Owen SA, Sanders LL, Edwards LJ, et al. Identification of higher risk thin nant melanoma: trends in basic science research over the last 100 years. ISRN
melanomas should be based on Breslow depth not Clark level IV. Cancer. Oncol. 2012;2012:546927.
2001;91:983–991. 266. Depasquale I, Thompson WD. Microvessel density for melanoma prognosis.
242. Attis MG, Vollmer RT. Mitotic rate in melanoma: a reexamination. Am J Clin Histopathology. 2005;47:186–194.
Pathol. 2007;127:380–384. 267. Massi D, Puig S, Franchi A, et al. Tumour lymphangiogenesis is a possible pre-
243. Day CL, Harrist TJ, Gorstein F, et al. Malignant melanoma: prognostic signif- dictor of sentinel lymph node status in cutaneous melanoma: a case-control
icance of ‘microscopic satellites’ in the reticular dermis and subcutaneous fat. study. J Clin Pathol. 2006;59:166–173.
Ann Surg. 1981;194:108–112. 268. Pastushenko I, Vermeulen PB, Vicente-Arregui S, et al. Peritumoral D2-40
244. Harrist TJ, Rigel DS, Day CL, et al. Microscopic satellites’ are more highly Chalkley score independently predicts metastases and survival in patients with
associated with regional lymph node metastases than is primary melanoma cutaneous malignant melanoma. J Cutan Pathol. 2015;42:699–711.
thickness. Cancer. 1984;53:2183–2187. 269. Morton DL, Wen DR, Wong JH, et al. Technical details of intraoperative lym-
245. Leon P, Daly JM, Synnestvedt M, et al. The prognostic implications of phatic mapping for early stage melanoma. Arch Surg. 1992;127:392–399.
microscopic satellites in patients with clinical stage I melanoma. Arch Surg. 270. Gershenwald JE, Thompson W, Mansfield PF, et al. Multi-institutional mel-
1991;126:1461–1468. anoma lymphatic mapping experience: the prognostic value of sentinel node
246. Bartlett EK, Gupta M, Datta J, et al. Prognosis of patients with melanoma status in 612 stage I or II melanoma patients. J Clin Oncol. 1999;17:976–
and microsatellitosis undergoing sentinel lymph node biopsy. Ann Surg Oncol. 983.
2014;21:1016–1023. 271. Dessureault S, Soong SJ, Ross MI, et al. Improved staging of node-negative
247. Borgstein PJ, Meijjer S, van Diest PJ. Are locoregional cutaneous metastases patients with intermediate to thick melanomas (>1 mm) with the use of lym-
predictablefi. Ann Surg Oncol. 1999;6:315–321. phatic mapping and sentinel lymph node biopsy. Ann Surg Oncol. 2001;8:
248. Thorn M, Ponten F, Bergstrom R, et al. Clinical and histopathologic predictors 766–770.
of survival in patients with malignant melanoma: a population-based study in 272. Leong SPL. Selective sentinel lymphadenectomy for malignant melanoma. Surg
Sweden. J Natl Cancer Inst. 1994;86:761–769. Clin North Am. 2003;83:157–183.
249. Straume O, Akslen LA. Independent prognostic importance of vascular invasion 273. Cochran AJ, Roberts AA, Saida T. The place of lymphatic mapping and sentinel
in nodular melanomas. Cancer. 1996;78:1211–1219. node biopsy in oncology. Int J Clin Oncol. 2003;8:139–150.
250. Kashani-Sabet M, Sagebiel RW, Ferreira CMM, et al. Vascular involve- 274. Faries MB, Thompson JF, Cochran AJ, et al. Completion dissection or
ment in the prognosis of primary cutaneous melanoma. Arch Dermatol. observation for sentinel-node metastasis in melanoma. N Engl J Med.
2001;137:1169–1173. 2017;376:2211–2222.
251. Nagore E, Oliver V, Botella-Estrada R, et al. Prognostic factors in localized 275. Postlewait LM, Farley CR, Seamens AM, et al. Morbidity and outcomes follow-
invasive cutaneous melanoma: high value of mitotic rate, vascular invasion and ing axillary lymphadenectomy for melanoma: weighing the risk of surgery in the
microscopic satellitosis. Melanoma Res. 2005;15:169–177. era of MSLT-II. Ann Surg Oncol. 2018;25:465–470.
252. Namikawa K, Aung PP, Gershenwald JE, et al. Clinical impact of ulceration 276. Prichard RS, Dijkstra B, McDermott EW, et al. The role of molecular staging in
width, lymphovascular invasion, microscopic satellitosis, perineural invasion, malignant melanoma. Eur J Surg Oncol. 2003;29:306–314.
and mitotic rate in patients undergoing sentinel lymph node biopsy for cutane- 277. da Silva AM, de Oliveira Filho RS, Ferreira LM, et al. Relevance of microme-
ous melanoma: a retrospective observational study at a comprehensive cancer tastases detected by reverse transcriptase polymerase chain reaction for mel-
center. Cancer Med. 2018;7:583–593. anoma recurrence: systematic review and meta-analysis. Sao Paulo Med J.
253. Tas F, Erturk K. Histological lymphovascular invasion is associated with nodal 2003;121:24–27.
involvement, recurrence, and survival in patients with cutaneous malignant mel- 278. Manlove JS, Terry-Humen E, Ikramullah EN, et al. The role of parent reli-
anoma. Int J Dermatol. 2017;56:166–170. giosity in teens’ transitions to sex and contraception. J Adolesc Health.
254. Niakosari F, Kahn HJ, McCready D, et al. Lymphatic invasion identified by 2006;39:578–587.
monoclonal antibody D2-40, younger age, and ulceration: predictors of senti- 279. Nahabedian MY, Tufaro AP, Manson PN. Sentinel lymph node biopsy for the
nel lymph node involvement in primary cutaneous melanoma. Arch Dermatol. T1 (thin) melanoma: is it necessary? Ann Plast Surg. 2003;50:601–606.
2008;144:462–467. 280. Boland GM, Gershenwald J. E Principles of melanoma staging. Cancer Treat
255. Creytens D. Prognostic implication of lymphovascular invasion detected by Res. 2016;167:131–148.
double immunostaining for D-40 and MITF1 in primary cutaneous melanoma: 281. Lowe JB, Hurst E, Moley FF, et al. Sentinel lymph node biopsy in patients with
beware of MITF1 specificity and sensitivity in unusual melanoma subtypes. Am thin melanoma. Arch Dermatol. 2003;139:617–621.
J Dermatopathol. 2017;39:709.
256. Feldmeyer L, Tetzlaff M, Fox P, et al. Prognostic implication of lymphovascular Immunohistochemistry of melanoma
invasion detected by double immunostaining for D2-40 and MITF1 in primary
cutaneous melanoma. Am J Dermatopathol. 2016;38:484–491. 1. Banerjee SS, Harris M. Morphological and immunophenotypic variations in
257. Rose AE, Christos PJ, Lackaye D, et al. Clinical relevance of detection of lym- malignant melanoma. Histopathology. 2000;36:387–402.
phovascular invasion in primary melanoma using endothelial markers D2-40 2. Bishop PW, Menasce LP, Yates AJ, et al. An immunophenotypic survey of
and CD34. Am J Surg Pathol. 2011;35:1441–1449. malignant melanomas. Histopathology. 1993;23:159–166.

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
1362.e6 References

3. Ordóñez NG. Value of melanocytic-associated immunohistochemical markers 28. Mitra D, Fisher DE. Transcriptional regulation in melanoma. Hematol Oncol
in the diagnosis of malignant melanoma: a review and update. Hum Pathol. Clin North Am. 2009;23:447–465, viii.
2014;45:191–205. 29. Busam KJ, Iversen K, Coplan KC, et al. Analysis of microphthalmia transcrip-
4. Miettinen M, Fernandez M, Franssila K, et al. Microphthalmia transcription tion factor expression in normal tissues and tumors, and comparison of its
factor in the immunohistochemical diagnosis of metastatic melanoma: compar- expression with S100-protein, gp100 and tyrosinase in desmoplastic melanoma.
ison with four other melanoma markers. Am J Surg Pathol. 2001;25:205–211. Am J Surg Pathol. 2001;25:197–204.
5. Drier J, Swanson PE, Cherwitz D, et al. S100 protein immunoreactivity in 30. King R, Googe PB, Weilbaecher KN, et al. Microphthalmia transcription factor
poorly differentiated carcinomas: immunohistochemical comparison with malig- expression in cutaneous benign, malignant melanocytic, and non-melanocytic
nant melanoma. Arch Pathol Lab Med. 1987;111:447–452. tumors. Am J Surg Pathol. 1999;25:51–57.
6. Herrera GA, Turbat-Herrera EA, Lott RL. S100 protein expression by primary 31. King R, Weilbaecher KN, McGill G, et al. Microphthalmia transcription
and metastatic adenocarcinomas. Am J Clin Pathol. 1998;89:168–176. factor. A sensitive and specific marker for melanoma diagnosis. Am J Pathol.
7. Agaimy A, Specht K, Stoehr R, et al. Metastatic malignant melanoma with 2001;155:731–738.
complete loss of differentiation markers (undifferentiated/dedifferentiated 32. Koch MB, Shih IM, Weiss SW, et al. Microphthalmia transcription factor
melanoma): analysis of 14 patients emphasizing phenotypic plasticity and the and melanoma cell adhesion molecule expression distinguish desmoplastic/
value of molecular testing as surrogate diagnostic marker. Am J Surg Pathol. spindle cell melanoma from morphologic mimics. Am J Surg Pathol. 2001;25:
2016;40:181–191. 58–64.
8. Trefzer U, Hofmann M, Reinke S, et al. Concordant loss of melanoma differen- 33. Trefzer U, Rietz N, Chen Y, et al. SM5-1: a new monoclonal antibody which
tiation antigens in synchronous and asynchronous melanoma metastases: impli- is highly sensitive and specific for melanocytic lesions. Arch Dermatol Res.
cations for immunotherapy. Melanoma Res. 2006;16:137–145. 2000;292:583–589.
9. Nonaka D, Chiriboga L, Rubin BP. Differential expression of S100 protein 34. Trefzer U, Chen Y, Herberth G, et al. The monoclonal antibody SM5-1 rec-
subtypes in malignant melanoma, and benign and malignant peripheral nerve ognizes a fibronectin variant which is widely expressed in melanoma. BMC
sheath tumors. J Cutan Pathol. 2008;35:1014–1019. Cancer. 2006;6:8.
10. Bacchi CE, Bonetti F, Pea M, et al. HMB-45: a review. Appl Immunohistochem. 35. Li B, Wang H, Zhang D, et al. Construction and characterization of a high-af-
1996;4:73–85. finity humanized SM5-1 monoclonal antibody. Biochem Biophys Res Commun.
11. Prieto VG, Woodruff JM. Expression of HMB45 antigen in spindle cell mela- 2007;357:951–956.
noma. J Cutan Pathol. 1997;24:580–581. 36. Su Z, Zheng X, Zhang X, et al. Sox10 regulates skin melanocyte prolifera-
12. Pea M, Bonetti F, Zamboni G, et al. Melanocyte-marker HMB-45 is regularly tion by activating the DNA replication licensing factor MCM5. J Dermatol Sci.
expressed in angiomyolipoma of the kidney. Pathology. 1991;23:185–188. 2017;85:216–225.
13. Gal AA, Koss MN, Hochholzer L, et al. An immunohistochemical study 37. Graf SA, Busch C, Bosserhoff AK, et al. SOX10 promotes melanoma cell
of benign clear cell (‘sugar’) tumor of the lung. Arch Pathol Lab Med. invasion by regulating melanoma inhibitory activity. J Invest Dermatol.
1991;115:1034–1038. 2014;134:2212–2220.
14. Busam KJ, Jungbluth AA. Melan-A. A new melanocytic differentiation marker. 38. Shakhova O, Zingg D, Schaefer SM, et al. Sox10 promotes the formation
Adv Anat Pathol. 1999;6:12–18. and maintenance of giant congenital naevi and melanoma. Nat Cell Biol.
15. Busam KJ, Chen Y-T, Old LJ, et al. Expression of Melan-A (MART-1) in 2012;14:882–890.
benign melanocytic nevi and primary cutaneous melanoma. Am J Surg Pathol. 39. Nonaka D, Chiriboga L, Rubin BP. Sox10: a pan-schwannian and melanocytic
1998;22:976–982. marker. Am J Surg Pathol. 2008;32:1291–1298.
16. Orosz Z. Melan-A/MART-1 expression in various melanocytic lesions and in 40. Karamchandani JR, Nielsen TO, van de Rijn M, et al. Sox10 and S100 in
non-melanocytic soft tissue lesions. Histopathology. 1999;34:517–525. the diagnosis of soft-tissue neoplasms. Appl Immunohistochem Mol Morphol.
17. Bergman R, Azzam H, Sprecher E, et al. A comparative immunohistochemi- 2012;20:445–450.
cal study of MART-1 expression in Spitz nevi, ordinary melanocytic nevi and 41. Shin J, Vincent JG, Cuda JD, et al. Sox10 is expressed in primary melanocytic
malignant melanomas. J Am Acad Dermatol. 2000;42:496–500. neoplasms of various histologies but not in fibrohistiocytic proliferations and
18. Busam KJ, Iversen K, Coplan KA, et al. Immunoreactivity for A103, an anti- histiocytoses. J Am Acad Dermatol. 2012;67:717–726.
body to Melan-A (MART-1), in adrenocortical and other steroid tumors. Am J 42. Nonaka D, Chiriboga L, Rubin BP. Sox10: a pan-schwannian and melanocytic
Surg Pathol. 1998;22:57–63. marker. Am J Surg Pathol. 2008;32:1291–1298.
19. Fetsch PA, Fetsch JF, Marincola FM, et al. Comparison of melanoma antigen 43. Plaza JA, Bonneau P, Prieto V, et al. Desmoplastic melanoma: an updated
recognized by T-cells (MART-1) to HMB-45: additional evidence to support a immunohistochemical analysis of 40 cases with a proposal for an additional
common lineage for angiomyolipoma, lymphangiomyomatosis, and clear cell panel of stains for diagnosis. J Cutan Pathol. 2016;43:2016;313–323.
sugar tumor. Mod Pathol. 1998;11:699–703. 44. Jackett LA, McCarthy SW, Scolyer RA. SOX10 expression in cutaneous scars: a
20. Berset M, Cerottini J-P, Guggisberg D, et al. Expression of Melan-A/MART-1 potential diagnostic pitfall in the evaluation of melanoma re-excision specimens.
antigen as a prognostic factor in primary cutaneous melanoma. Int J Cancer. Pathology. 2016;48:626–628.
2001;95:73–77. 45. Mu EW, Quatrano NA, Yagerman SE, et al. Evaluation of MITF, SOX10,
21. Orchard GE. Comparison of immunohistochemical labeling of melanocyte MART-1, and R21 immunostaining for the diagnosis of residual melanoma in
differentiation antibodies melan-A, tyrosinase and HMB45 with NKIC3 and situ on chronically sun-damaged skin. Dermatol Surg. 2018;44:933–938.
S100 protein in the evaluation of benign nevi and melanoma. J Histochem. 46. Muzumdar S, Argraves M, Kristjansson A, et al. A quantitative comparison
2000;32:475–481. between SOX10 and MART-1 immunostaining to detect melanocytic hyperpla-
22. Kaufmann O, Koch S, Burghardt J, et al. Tyrosinase, melan-A, and KBA62 as sia in chronically sun-damaged skin. J Cutan Pathol. 2018;45:263–268.
markers for the immunohistochemical identification of metastatic amelanotic 47. Vrotsos E, Alexis J. Can SOX-10 or KBA.62 replace S100 protein in immu-
melanoma on paraffin sections. Mod Pathol. 1998;11:740–746. nohistochemical evaluation of sentinel lymph nodes for metastatic melanoma?
23. Takimoto H, Suzuki S, Masui S, et al. MAT-1, a monoclonal antibody that Appl Immunohistochem Mol Morphol. 2016;24:26–29.
specifically recognizes human tyrosinase. J Invest Dermatol. 1995;105:764– 48. Miettinen M, Franssila K. Immunohistochemical spectrum of malignant mela-
768. noma: the common presence of keratins. Lab Invest. 1989;61:623–628.
24. Sato N, Suzuki S, Takimoto H, et al. Monoclonal antibody MAT-1 against 49. Ben-Izhak O, Stark P, Levy R, et al. Epithelial markers in malignant mela-
human tyrosinase can detect melanogenic cells on formalin-fixed paraffin-em- noma. A study of primary lesions and their metastases. Am J Dermatopathol.
bedded sections. Pigment Cell Res. 1996;9:72–76. 1994;16:241–246.
25. Clarkson KS, Sturdgess IC, Molyneux AJ. The usefulness of tyrosinase in the 50. Selby WL, Nance KV, Park HK. CEA immunoreactivity in metastatic mela-
immunohistochemical assessment of melanocytic lesions: a comparison of the noma. Mod Pathol. 1992;5:415–419.
novel T311 antibody (anti-tyrosinase) with S100, HMB45 and A103 (anti- 51. Banerjee SS, Bishop PW, Nicholson CM, et al. Malignant melanoma showing
melan-A). J Clin Pathol. 2001;54:196–200. smooth muscle differentiation. J Clin Pathol. 1996;49:950–951.
26. Reinke S, Koniger P, Herberth G, et al. Differential expression of MART-1, 52. Longacre TA, Egbert BM, Rouse RV. Desmoplastic and spindle-cell malig-
tyrosinase, and SM5-1 in primary and metastatic melanoma. Am J Dermatopa- nant melanoma. An immunohistochemical study. Am J Surg Pathol.
thol. 2005;27:401–406. 1996;20:1489–1500.
27. Orchard G. Evaluation of melanocytic neoplasms: application of a pan-mela- 53. Facchetti E, Bertalot G, Grigolato PG. KP1 (CD68) staining of malignant mela-
noma antibody cocktail. Br J Biomed Sci. 2002;59:196–202. nomas. Histopathology. 1991;19:141–145.

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
References 1362.e7

54. Pernick NL, Dasilva M, Gangi MD, et al. Histiocytic markers in melanoma. 79. Ladstein RG, Bachmann IM, Straume O, et al. Ki-67 expression is superior to
Mod Pathol. 1999;12:1072–1077. mitotic count and novel proliferation markers PHH3, MCM4 and mitosin as a
55. Reed JA, Albino AP. Update of diagnostic and prognostic markers in cutaneous prognostic factor in thick cutaneous melanoma. BMC Cancer. 2010;10:140.
malignant melanoma. Clin Lab Med. 2000;20:817–838. 80. Cerroni L, Soyer HP, Kerl H. BCL-2 protein expression in cutaneous malignant
56. Mandalà M, Massi D. Tissue prognostic biomarkers in primary cutaneous mel- melanoma and benign melanocytic nevi. Am J Dermatopathol. 1995;17:7–11.
anoma. Virchows Arch. 2014;464:265–281. 81. van den Oord JJ, Vandeghinste N, De Ley M, et al. Bcl-2 expression in human
57. Tetzlaff MT, Torres-Cabala CA, Pattanaprichakul P, et al. Emerging clinical melanocytes and melanocytic tumors. Am J Pathol. 1994;145:294–300.
applications of selected biomarkers in melanoma. Clin Cosmet Investig Derma- 82. Saenz-Santamarfa MC, Reed JC, McNutt NC, et al. Immunohistochemical
tol. 2015;8:35–46. expression of BCL-2 in melanomas and intradermal nevi. J Cutan Pathol.
58. Gould Rothberg BE, Bracken MB, Rimm DL. Tissue biomarkers for prognosis 1994;21:393–397.
in cutaneous melanoma: a systematic review and meta-analysis. J Natl Cancer 83. Radhi JM. Malignant melanoma arising from nevi, p53, p16, and Bcl-2:
Inst. 2009;101:452–474. expression in benign versus malignant components. J Cutan Med Surg.
59. McShane LM, Altman DG, Sauerbrei W, et al. Reporting recommenda- 1999;3:293–297.
tions for tumor marker prognostic studies (REMARK). J Natl Cancer Inst. 84. Kanter-Lewensohn L, Hedblad MA, Wejde J, et al. Immunohistochemical
2005;97:1180–1184. markers for distinguishing Spitz nevi from malignant melanomas. Mod Pathol.
60. McShane LM, Altman DG, Sauerbrei W, et al. Reporting recommenda- 1997;10:917–920.
tions for tumor marker prognostic studies (REMARK). J Natl Cancer Inst. 85. Ramsay JA, From L, Kahn HJ. Bcl-2 protein expression in melanocytic neo-
2005;97:1180–1184. plasms of the skin. Mod Pathol. 1995;8:150–154.
61. Altman DG, McShane LM, Sauerbrei W, et al. Reporting recommendations for 86. Tron VA, Krajewski S, Klein-Parker H, et al. Immunohistochemical analysis of
tumor marker prognostic studies (REMARK): explanation and elaboration. Bcl-2 regulation in cutaneous melanoma. Am J Pathol. 1995;146:643–650.
PLoS Med. 2012;9:e1001216. 87. Tang L, Tron VA, Reed JC, et al. Expression of apoptosis regulators in cutane-
62. Kattan MW, Hess KR, Amin MB, et al. American Joint Committee on Cancer ous malignant melanoma. Clin Cancer Res. 1998;4:1865–1871.
acceptance criteria for inclusion of risk models for individualized prognosis in 88. Divito KA, Berger AJ, Camp RL, et al. Automated quantitative analysis of tissue
the practice of precision medicine. CA Cancer J Clin. 2016;66:370–374. microarrays reveals an association between high Bcl-2 expression and improved
63. Gould Rothberg BE, Berger AJ, Molinaro AM, et al. Melanoma prognos- outcome in melanoma. Cancer Res. 2004;64:8773–8777.
tic model using tissue microarrays and genetic algorithms. J Clin Oncol. 89. Georgieva J, Sinha P, Schadendorf D. Expression of cyclins and cyclin depen-
2009;27:5772–5780. dent kinases in human benign and malignant melanocytic lesions. J Clin Pathol.
64. Bergman R, Malkin L, Sabo E, et al. MIB-1 monoclonal antibody to determine 2001;54:229–235.
proliferative activity as an adjunct to the histopathologic differential diagnosis 90. Alekseenko A, Wojas-Pelc A, Lis GJ, et al. Cyclin D1 and D3 expression in
of Spitz nevus. J Am Acad Dermatol. 2001;44:500–504. melanocytic skin lesions. Arch Dermatol Res. 2010;302:545–550.
65. Li LX, Crotty KA, McCarthy SW, et al. A zonal comparison of MIB-1 immu- 91. Curtin JA, Fridlyand J, Kageshita T, et al. Distinct sets of genetic alterations in
noreactivity in benign and malignant melanocytic lesions. Am J Dermatopathol. melanoma. N Engl J Med. 2005;353:2135–2147.
2000;22:489–495. 92. Sauter ER, Yeo UC, von Stemm A, et al. Cyclin D1 is a candidate oncogene in
66. Ramsay JA, From L, Iscoe NA, et al. MIB-1 proliferative activity is a significant cutaneous melanoma. Cancer Res. 2002;62:3200–3206.
prognostic factor in primary thick cutaneous melanoma. J Invest Dermatol. 93. Gerami P, Guitart J, Martini M, et al. Cyclin D1 homogeneous staining regions
1995;105:22–26. by fluorescent in situ hybridization: a possible indicator of aggressive behavior
67. Boni R, Doguoglu A, Burg G, et al. MIB-1 immunoreactivity correlates with in melanomas. Arch Dermatol. 2008;144:1235–1236.
metastatic dissemination in primary thick cutaneous melanoma. J Am Acad 94. Florenes VA, Faye RS, Maelandsmo GM, et al. Levels of cyclin D1 and D3
Dermatol. 1996;35:416–418. in malignant melanoma: deregulated cyclin D3 expression is associated with
68. Vogt T, Zipperer KH, Vogt A, et al. p53 protein and Ki-67 antigen expression poor clinical outcome in superficial melanoma. Clin Cancer Res. 2000;6:3614–
are both reliable biomarkers of prognosis in thick stage I nodular melanomas of 3620.
the skin. Histopathology. 1997;30:57–63. 95. Spofford LS, Abel EV, Boisvert-Adamo K, et al. Cyclin D3 expression in
69. Moretti S, Spallanzani A, Chiarugi A, et al. Correlation of Ki-67 in cutaneous melanoma cells is regulated by adhesion-dependent phosphatidylinosi-
primary melanoma with prognosis in a prospective study: different correlation tol 3-kinase signaling and contributes to G1-S progression. J Biol Chem.
according to thickness. J Am Acad Dermatol. 2001;44:188–192. 2006;281:25644–25651.
70. Straume O, Sviland L, Akslen LA. Loss of nuclear p16 protein expression 96. Reed JA, Loganzo F, Shea CR, et al. Loss of expression of the p16/cyclin depen-
correlates with increased tumor cell proliferation (Ki-67) and poor prog- dent kinase inhibitor 2 tumor suppressor gene in melanocytic lesions correlates
nosis in patients with vertical growth phase melanoma. Clin Cancer Res. with invasive stages of tumor progression. Cancer Res. 1995;55:2713–2718.
2000;6:1845–1853. 97. Soler-Carillo J, Puig S, Palou J, et al. p16 protein expression as an important
71. Gershenwald JE, Scolyer RA, Hess KR, et al. Melanoma staging: Evidence-based prognostic factor in primary cutaneous malignant melanoma. Am J Dermatopa-
changes in the American Joint Committee on Cancer eighth edition cancer thol. 1998;20:603A.
staging manual. CA Cancer J Clin. 2017;67:472–492. 98. Straume O, Sviland L, Akslen LA. Loss of nuclear p16 protein expression
72. Tetzlaff MT, Curry JL, Ivan D, et al. Immunodetection of phosphohistone H3 correlates with increased tumor cell proliferation (Ki-67) and poor prog-
as a surrogate of mitotic figure count and clinical outcome in cutaneous mela- nosis in patients with vertical growth phase melanoma. Clin Cancer Res.
noma. Mod Pathol. 2013;26:1153–1160. 2000;6:1845–1853.
73. Nielsen PS, Riber-Hansen R, Schmidt H, et al. Automated quantification of 99. Mihic-Probst D, Saremaslani P, Komminoth P, et al. Immunostaining for the
proliferation with automated hot-spot selection in phosphohistone H3/MART1 tumour suppressor gene p16 product is a useful marker to differentiate mela-
dual-stained stage I/II melanoma. Diagn Pathol. 2016;11:35. noma metastasis from lymph-node nevus. Virchows Arch. 2003;443:745–751.
74. Ottmann K, Tronnier M, Mitteldorf C. Detection of mitotic figures in thin mel- 100. Albino AP, Vidal MJ, McNutt NS, et al. Mutation and expression of the p53
anomas–immunohistochemistry does not replace the careful search for mitotic gene in human malignant melanoma. Melanoma Res. 1994;4:35–45.
figures in hematoxylin-eosin stain. J Am Acad Dermatol. 2015;73:637–644. 101. Piana S, Tagliavini E, Ragazzi M, et al. Lymph node melanocytic nevi: patho-
75. Nielsen PS, Riber-Hansen R, Jensen TO, et al. Proliferation indices of phospho- genesis and differential diagnoses, with special reference to p16 reactivity.
histone H3 and Ki67: strong prognostic markers in a consecutive cohort with Pathol Res Pract. 2015;211:381–388.
stage I/II melanoma. Mod Pathol. 2013;26:404–413. 102. Kaleem Z, Lind AC, Humphrey PA, et al. Concurrent Ki-67 and p53 immuno-
76. Ladstein RG, Bachmann IM, Straume O, et al. Prognostic importance of the labelling in cutaneous melanocytic neoplasms: an adjunct for recognition of the
mitotic marker phosphohistone H3 in cutaneous nodular melanoma. J Invest vertical growth phase in malignant melanomasfi. Mod Pathol. 2000;13:217–222.
Dermatol. 2012;132:1247–1252. 103. Gelsleichter L, Gown AM, Zarbo RJ, et al. p53 and mdm-2 expression in
77. Schimming TT, Grabellus F, Roner M, et al. pHH3 immunostaining improves malignant melanoma: an immunocytochemical study of expression of p53,
interobserver agreement of mitotic index in thin melanomas. Am J Dermatopa- mdm-2, and markers of cell proliferation in primary versus metastatic tumors.
thol. 2012;34:266–269. Mod Pathol. 1995;8:530–535.
78. Casper DJ, Ross KI, Messina JL, et al. Use of anti-phosphohistone H3 immuno- 104. Yamamoto M, Takahashi H, Saitoh K, et al. Expression of the p53 protein
histochemistry to determine mitotic rate in thin melanoma. Am J Dermatopa- in malignant melanoma as a prognostic indicator. Arch Dermatol Res.
thol. 2010;32:650–654. 1995;287:146–151.

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
1362.e8 References

105. Loggini B, Rinaldi I, Pingitore R, et al. Immunohistochemical study of 49 130. Galon J, Mlecnik B, Bindea G, et al. Towards the introduction of the ‘Immuno-
cutaneous melanomas: p53, PCNA, Bcl-2 expression and multidrug resistance. score’ in the classification of malignant tumours. J Pathol. 2014;232:199–209.
Tumori. 2001;87:179–186. 131. Axelrod ML, Johnson DB, Balko JM. Emerging biomarkers for cancer immuno-
106. de Sa BC, Fugimori ML, Ribeiro Kde C, et al. Proteins involved in pRb and therapy in melanoma. Semin Cancer Biol. 2017. S1044-579X(17)30121-9.
p53 pathways are differentially expressed in thin and thick superficial spreading 132. Diggs LP, Hsueh EC. Utility of PD-L1 immunohistochemistry assays for predict-
melanomas. Melanoma Res. 2009;19:135–141. ing PD-1/PD-L1 inhibitor response. Biomark Res. 2017;5:12.
107. Kim DW, Haydu LE, Joon AY, et al. Clinicopathological features and clinical 133. Zhai L, Spranger S, Binder DC, et al. Molecular pathways: targeting ido1 and
outcomes associated with TP53 and BRAFNon-V600 mutations in cutaneous other tryptophan dioxygenases for cancer immunotherapy. Clin Cancer Res.
melanoma patients. Cancer. 2017;123:1372–1381. 2015;21:5427–5433.
108. Väisänen A, Kuvaja P, Kallioinen M, et al. A prognostic index in skin mela- 134. Castelli C, Triebel F, Rivoltini L, et al. Lymphocyte activation gene-3 (LAG-3,
noma through the combination of matrix metalloproteinase-2, Ki67, and p53. CD223) in plasmacytoid dendritic cells (pDCs): a molecular target for the resto-
Hum Pathol. 2011;42:1103–1111. ration of active antitumor immunity. Oncoimmunology. 2014;3:e967146.
109. Tetzlaff MT, Pattanaprichakul P, Wargo J, et al. Utility of BRAF V600E immu-
nohistochemistry expression pattern as a surrogate of BRAF mutation status in Histologic variants of melanoma
154 patients with advanced melanoma. Hum Pathol. 2015;46:1101–1110.
110. Kakavand H, Walker E, Lum T, et al. BRAF(V600E) and NRAS(Q61L/Q61R) 1. Rongioletti F, Smoller BR. Unusual histological variants of cutaneous malig-
mutation analysis in metastatic melanoma using immunohistochemistry: a study nant melanoma with some clinical and possible prognostic correlations. J Cutan
of 754 cases highlighting potential pitfalls and guidelines for interpretation and Pathol. 2005;32:589–603.
reporting. Histopathology. 2016;69:680–686. 2. Magro CM, Crowson AN, Mihm MC. Unusual variants of malignant mela-
111. Long GV, Wilmott JS, Capper D, et al. Immunohistochemistry is highly sensi- noma. Mod Pathol. 2006;19(suppl 2):S41–S70.
tive and specific for the detection of V600E BRAF mutation in melanoma. Am 3. Barnhill RL, Gupta K. Unusual variants of malignant melanoma. Clin Derma-
J Surg Pathol. 2013;37:61–65. tol. 2009;27:564–587.
112. Siroy AE, Boland GM, Milton DR, et al. Beyond BRAF(V600): clinical muta- 4. Bastian BC. The molecular pathology of melanoma: an integrated taxonomy of
tion panel testing by next-generation sequencing in advanced melanoma. J melanocytic neoplasia. Annu Rev Pathol. 2014;9:239–271.
Invest Dermatol. 2015;135:508–515. 5. Mazurenko NN, Tsyganova IV, Lushnikova AA, et al. [Spectrum of oncogene
113. Greaves WO, Verma S, Patel KP, et al. Frequency and spectrum of BRAF muta- mutations is different in melanoma subtypes]. [Article in Russian]. Mol Biol
tions in a retrospective, single-institution study of 1112 cases of melanoma. J (Mosk). 2015;49:1022–1029.
Mol Diagn. 2013;15:220–226.
114. Felisiak-Goląbek A, Inaguma S, Kowalik A, et al. SP174 antibody lacks speci- Nested melanoma
ficity for NRAS Q61R and cross-reacts with HRAS and KRAS Q61R mutant
proteins in malignant melanoma. Appl Immunohistochem Mol Morphol. 1. Kutzner H, Metzler G, Argenyi Z, et al. Histological and genetic evidence for a
2018;26:40–45. variant of superficial spreading melanoma composed of predominantly of large
115. Lasota J, Kowalik A, Felisiak-Golabek A, et al. SP174, NRAS Q61R mutant-spe- nests. Mod Pathol. 2012;25:838–845.
cific antibody, cross-reacts with KRAS Q61R mutant protein in colorectal carci- 2. Kossard S. Nested melanoma. Mod Pathol. 2013;26:615–616.
noma. Arch Pathol Lab Med. 2017;141:564–568.
116. Uguen A, Talagas M, Costa S, et al. NRAS (Q61R), BRAF (V600E) immuno- Minimal deviation melanoma
histochemistry: a concomitant tool for mutation screening in melanomas. Diagn
Pathol. 2015;10:121. 1. Reed RJ, Ichinose H, Clark WH, et al. Common and uncommon melanocytic
117. Ilie M, Long-Mira E, Funck-Brentano E, et al. Immunohistochemistry as a nevi and borderline melanomas. Semin Oncol. 1975;2:119–147.
potential tool for routine detection of the NRAS Q61R mutation in patients 2. Reed RJ. Minimum deviation melanoma. Am J Surg Pathol. 1978;2:215–220.
with metastatic melanoma. J Am Acad Dermatol. 2015;72:786–793. 3. Muhlbauer JE, Margolis RJ, Mihm MC Jr, et al. Minimal deviation melanoma:
118. Massi D, Simi L, Sensi E, et al. Immunohistochemistry is highly sensitive and a histological variant of cutaneous melanoma in its vertical growth phase. J
specific for the detection of NRASQ61R mutation in melanoma. Mod Pathol. Invest Dermatol. 1983;80:63S–65S.
2015;28:487–497. 4. Phillips ME, Margolis RJ, Merot Y, et al. The spectrum of minimal deviation
119. Wolchok JD, Chiarion-Sileni V, Gonzalez R, et al. Overall survival with com- melanoma: a clinicopathologic study of 21 cases. Hum Pathol. 1986;17:796–
bined nivolumab and ipilimumab in advanced melanoma. N Engl J Med. 806.
2017;377:1345–1356. 5. LeBoit PE. Minimal deviation melanoma: concept or quagmire? Adv Dermatol.
120. Larkin J, Chiarion-Sileni V, Gonzalez R, et  al. Combined nivolumab 1998;13:289–303.
and ipilimumab or monotherapy in untreated melanoma. N Engl J Med. 6. Podnos YD, Jimenez JC, Zainabadi K, et al. Minimal deviation melanoma.
2015;373(1):23–34. Cancer Treat Rev. 2002;28:219–221.
121. Schachter J, Ribas A, Long GV, et al. Pembrolizumab versus ipilimumab for 7. Reed RJ. Minimal deviation melanoma. Borderline and intermediate melano-
advanced melanoma: final overall survival results of a multicentre, randomised, cytic neoplasia. Clin Lab Med. 2000;20:745–758.
open-label phase 3 study (KEYNOTE-006). Lancet. 2017;390:1853–1862. 8. Stas M, van den Oord JJ, Garmyn M, et al. Minimal deviation and/or naevoid
122. Robert C, Schachter J, Long GV. Pembrolizumab versus ipilimumab in advanced melanoma: is recognition worthwhile? A clinicopathological study of nine cases.
melanoma. N Engl J Med. 2015;372:2521–2532. Melanoma Res. 2000;10:371–380.
123. Cottrell TR, Taube JM. PD-L1 and Emerging Biomarkers in immune checkpoint
blockade therapy. Cancer J. 2018;24(1):41–46. Nevoid melanoma
124. Errico A. Melanoma: CheckMate 067–frontline nivolumab improves PFS alone
or in combination with ipilimumab. Nat Rev Clin Oncol. 2015;12(8):435. 1. Levene A. On the histological diagnosis and prognosis of malignant melanoma.
125. Sunshine JC, Nguyen PL, Kaunitz GJ, et al. PD-L1 Expression in Melanoma: J Clin Pathol. 1980;33:101–124.
a quantitative immunohistochemical antibody comparison. Clin Cancer Res. 2. Schmoeckel C, Castro CE, Braun-Falco O. Nevoid malignant melanoma. Arch
2017;23(16):4938–4944. Dermatol Res. 1985;277:362–369.
126. Rimm DL, Han G, Taube JM, et al. A prospective, multi-institutional, Patholo- 3. DiCaudo DJ, McCalmont TH, Wick MR. Selected diagnostic problems in neo-
gist-based assessment of 4 immunohistochemistry assays for PD-L1 expression plastic dermatopathology. Arch Pathol Lab Med. 2007;131:434–439.
in non-Small cell lung cancer. JAMA Oncol. 2017;3(8):1051–1058. 4. Xu X, Elder DE. A practical approach to selected problematic melanocytic
127. Taube JM, Galon J, Sholl LM, et al. Implications of the tumor immune micro- lesions. Am J Clin Pathol. 2004;121(suppl):S3–S32.
environment for staging and therapeutics. Mod Pathol. 2018;31(2):214–234. 5. Diwan AH, Lazar AJ. Nevoid melanoma. Clin Lab Med. 2011;31:243–253.
128. Galon J, Lugli A, Bifulco C, et al. World-Wide Immunoscore Task Force: 6. Jain M, Marghoob AA. Integrating clinical, dermoscopy, and reflectance con-
meeting report from the “Melanoma Bridge”, Napoli, November 30th-Decem- focal microscopy findings into correctly identifying a nevoid melanoma. JAAD
ber 3rd, 2016. J Transl Med. 2017;15(1):212. Case Rep. 2017;3:505–508.
129. Ascierto PA, Capone M, Urba WJ, et al. The additional facet of immunoscore: 7. Blessing K, Evans AT, Al-Nafussi A. Verrucous nevoid and keratotic malig-
immunoprofiling as a possible predictive tool for cancer treatment. J Transl nant melanoma: a clinico-pathologic study of 20 cases. Histopathology.
Med. 2013;11:54. 1995;23:453–458.

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
References 1362.e9

8. Wong TY, Suster S, Duncan LM, et al. Nevoid melanoma: a clinicopathological 16. Hanson IM, Banerjee SS, Menasce LP, et al. A study of eleven malignant mela-
study of seven cases of malignant melanoma mimicking spindle and epithelioid nomas in adults with small cell morphology: emphasis on diagnostic difficulties
cell nevus and verrucous dermal nevus. Hum Pathol. 1995;26:171–179. and unusual features. Histopathology. 2002;40:187–195.
9. Suster S, Ronnen M, Bubis JJ. Verrucous pseudonevoid melanoma. J Surg
Oncol. 1987;36:134–137. Spitzoid melanoma
10. Kossard S, Wilkinson B. Nucleolar organizer regions and image analy-
sis nuclear morphometry of small cell (nevoid) melanoma. J Cutan Pathol. 1. Spitz S. Melanomas of childhood. Am J Pathol. 1948;24:591–609.
1995;22:132–136. 2. Allen AC, Spitz S. Malignant melanoma: a clinicopathological analysis of the
11. McNutt NS, Urmacher C, Hakimian J, et al. Nevoid malignant melanoma: criteria for diagnosis and prognosis. Cancer. 1953;6:1–45.
morphologic patterns and immunohistochemical reactivity. J Cutan Pathol. 3. Kamino H. Spitzoid melanoma. Clin Dermatol. 2009;27:545–555.
1995;22:502–517. 4. Titus L, Barnhill RL, Lott JP, et al. The influence of tumor regression, solar elas-
12. Zembowicz A, McCusker M, Chiarelli K, et al. Morphological characterization tosis, and patient age on pathologists’ interpretation of melanocytic skin lesions.
of nevoid melanoma: a study of 16 cases with a review of the literature. Am J Lab Invest. 2017;97:187–193.
Dermatopathol. 2000;23:167–175. 5. Vollmer RT. Patient age in Spitz nevus and malignant melanoma: implication of
13. Idriss MH, Rizwan L, Sferuzza A, et al. Nevoid melanoma: A study of 43 cases Bayes rule for differential diagnosis. Am J Clin Pathol. 2004;121:872–877.
with emphasis on growth pattern. J Am Acad Dermatol. 2015;73:836–842. 6. Dika E, Ravaioli GM, Fanti PA, et al. Spitz nevi and other spitzoid neoplasms in
14. Falconieri G, Luzar B, Angione V, et al. Primary cutaneous nevoid melanoma children: overview of incidence data and diagnostic criteria. Pediatr Dermatol.
with Homer-wright rosettes: a hitherto unrecognized variant with immu- 2017;34:25–32.
nohistochemical and ultrastructural study. Am J Dermatopathol. 2010;32: 7. Weedon D, Little JH. Spindle and epithelioid cell naevi in children and adults.
606–609. Cancer. 1977;40:217–225.
15. Horenstein MG, Prieto VG, Burchette MC Jr, et al. Keratotic melanocytic 8. Okun MR. Melanoma resembling spindle and epithelioid cell nevus. Arch Der-
nevus: a clinicopathologic and immunohistochemical study. J Cutan Pathol. matol. 1979;115:1416–1420.
2000;27:344–350. 9. Peters MS, Goellner JR. Spitz nevi and malignant melanomas of childhood and
16. Cook MG, Massi D, Blokx WAM, et al. New insights into naevoid melanomas: adolescence. Histopathology. 1986;10:1289–1302.
a clinicopathological reassessment. Histopathology. 2017;71:943–950. 10. Smith KJ, Barrett TL, Skelton HG 3rd., et al. Spindle cell and epithelioid cell
17. Plaza JA, Torres-Cabala C, Evans H, et al. Cutaneous metastases of malignant nevi with atypia and metastasis (malignant Spitz nevus). Am J Surg Pathol.
melanoma: a clinicopathologic study of 192 cases with emphasis on the mor- 1989;13:931–939.
phologic spectrum. Am J Dermatopathol. 2010;32:129–136. 11. Mehregan AH, Mehregan DA. Malignant melanoma in childhood. Cancer.
18. Miura K, Harada H, Tsutsui Y. Small cell type malignant melanoma which 1992;71:4096–4103.
developed in a 16-year-old female with a congenital dermal nevus and metasta- 12. Crotty KA, McCarthy SW, Palmer AA, et al. Malignant melanoma in child-
sized 12 years after excision. Pathol Int. 1999;49:247–252. hood: a clinicopathologic study of 13 cases and comparison with Spitz nevi.
19. Yélamos O, Busam KJ, Lee C, et al. Morphologic clues and utility of fluo- World J Surg. 1992;16:179–185.
rescence in situ hybridization for the diagnosis of nevoid melanoma. J Cutan 13. McCarthy SW, Crotty KA, Palmer AA, et al. Cutaneous malignant melanoma in
Pathol. 2015;42:796–806. teenagers. Histopathology. 1994;24:453–461.
20. Massi G. Melanocytic nevi simulant of melanoma with medicolegal relevance. 14. Barnhill RL, Flotte TJ, Fleischli M, et al. Cutaneous melanoma and atypical
Virchows Arch. 2007;451:623–647. Spitz tumors in childhood. Cancer. 1995;76:1833–1845.
15. Walsh N, Crotty K, Palmer AA, et al. Spitz nevus versus spitzoid malignant
melanoma: an evaluation of the current distinguishing histopathologic criteria.
Small cell melanoma
Hum Pathol. 1998;29:1105–1112.
1. Nakhleh RE, Wick MR, Rocamora A, et al. Morphologic diversity in malignant 16. Spatz A, Calonje E, Handfield-Jones S, et al. Spitz tumors in childhood: a
melanomas. Am J Clin Pathol. 1990;93:731–740. grading system for risk stratification. Arch Dermatol. 1999;135:282–285.
2. Reed WB, Becker SW Sr, Becker SW Jr, et al. Giant pigmented nevi, melanoma 17. Barnhill RL, Argenyi ZB, From L, et al. Atypical Spitz nevi/tumors: lack of
and leptomeningeal melanocytosis. Arch Dermatol. 1965;91:100–119. consensus for diagnosis, discrimination from melanoma, and prediction of
3. Reed RJ. Giant congenital nevi: a conceptualization of patterns. J Invest Derma- outcome. Hum Pathol. 1999;30:513–520.
tol. 1993;10(suppl):300–312. 18. Mooi WJ. Histopathology of Spitz naevi and ‘Spitzoid’ melanomas. Curr Top
4. Kossard S, Wilkinson B. Nucleolar organizer regions and image analy- Pathol. 2001;94:65–77.
sis nuclear morphometry of small cell (nevoid) melanoma. J Cutan Pathol. 19. Crotty KA, Scolyer RA, Li LXL, et al. Spitz nevus versus Spitzoid melanoma:
1995;22:132–136. when and how can they be distinguishedfi. Pathology. 2003;34:6–12.
5. Cook MG, Massi D, Blokx WAM, et al. New insights into naevoid melanomas: 20. Mones JM, Ackerman AB. Melanomas in prepubescent children: review com-
a clinicopathological reassessment. Histopathology. 2017;71:943–950. prehensively, critique historically, criteria diagnostically, and course biologically.
6. Barnhill RL, Flotte TJ, Fleischli M, et al. Cutaneous melanoma and atypical Am J Dermatopathol. 2003;25:223–238.
Spitz tumors in childhood. Cancer. 1995;76:1833–1845. 21. Gerami P, Busam K, Cochran A, et al. Histomorphologic assessment and
7. Spatz A, Ruiter D, Hardmeier T, et al. Melanoma in childhood: an EORTC- interobserver diagnostic reproducibility of atypical spitzoid melanocytic neo-
MCG multicenter study on the clinico-pathological aspects. Int J Cancer. plasms with long-term follow-up. Am J Surg Pathol. 2014;38:934–940.
1996;68:317–324. 22. Zhao G, Lee KC, Peacock S, et al. The utilization of spitz-related nomencla-
8. Attanoos R, Griffiths D. FR. Metastatic small cell melanoma to the stomach ture in the histological interpretation of cutaneous melanocytic lesions by prac-
mimicking primary gastric lymphoma. Histopathology. 1992;21:173–175. ticing pathologists: results from the M-Path study. J Cutan Pathol. 2017;44:
9. House NS, Fedok F, Maloney ME, et al. Malignant melanoma with clini- 5–14.
cal and histologic features of Merkel cell carcinoma. J Am Acad Dermatol. 23. Crotty KA, Scolyer RA, Li L, et al. Spitz naevus versus Spitzoid melanoma:
1994;31:839–842. when and how can they be distinguished? Pathology. 2002;34:6–12.
10. Fitzgibbons PL, Martin SE, Simmons TJ. Malignant melanoma metastatic to the 24. Ludgate MW, Fullen DR, Lee J, et al. The atypical Spitz tumor of uncertain
ovary. Am J Surg Pathol. 1987;11:950–964. biologic potential: a series of 67 patients from a single institution. Cancer.
11. Song HS, Kim YC. Small cell melanoma. Ann Dermatol. 2014;26:419–421. 2009;115:631–641.
12. Kossard S, Wilkinson B. Small cell (nevoid) melanoma: a clinicopathologic 25. Barnhill RL. The Spitzoid lesion: rethinking Spitz tumors, atypical variants,
study of 131 cases. Australas J Dermatol. 1997;38:S54–S58. ‘Spitzoid melanoma’ and risk assessment. Mod Pathol. 2006;19(suppl 2):S21–
13. Blessing K, Grant JJH, Sanders DSA, et al. Small cell melanoma: a variant of S33.
nevoid melanoma: clinicopathological features and histopathological differential 26. Hantschke M, Bastian BC, LeBoit PE. Consumption of the epidermis: a diag-
diagnosis. J Clin Pathol. 2000;53:591–595. nostic criterion for the differential diagnosis of melanoma and Spitz nevus. Am
14. Cuellar FA, Vilalta A, Rull R, et al. Small cell melanoma and ulceration as pre- J Surg Pathol. 2004;28:1621–1625.
dictors of positive sentinel lymph node in malignant melanoma patients. Mela- 27. Walters RF, Groben PA, Busam K, et al. Consumption of the epidermis: a
noma Res. 2004;14:277–282. criterion in the differential diagnosis of melanoma and dysplastic nevi that is
15. Banerjee SS, Harris M. Morphological and immunophenotypic variations in associated with increasing breslow depth and ulceration. Am J Dermatopathol.
malignant melanoma. Histopathology. 2000;36:387–402. 2007;29:527–533.

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
1362.e10 References

28. Ghorbani Z, Dowlati Y, Mehregan AH. Amelanotic Spitzoid melanoma in the 55. Gamblin TC, Edington H, Kirkwood JM, et al. Sentinel lymph node biopsy
burn scar of a child. Pediatr Dermatol. 1996;13:285–287. for atypical melanocytic lesions with spitzoid features. Ann Surg Oncol.
29. Paradela S, Fonseca E, Pita S, et al. Spitzoid melanoma in children: clinicopath- 2006;13:1664–1670.
ological study and application of immunohistochemistry as an adjunct diagnos- 56. Murali R, Sharma RN, Thompson JF, et al. Sentinel lymph node biopsy in
tic tool. J Cutan Pathol. 2009;36:740–752. histologically ambiguous melanocytic tumors with spitzoid features (so-called
30. Vollmer RT. Use of Bayes rule and MIB-1 proliferation index to discriminate atypical spitzoid tumors). Ann Surg Oncol. 2008;15:302–309.
Spitz nevus from malignant melanoma. Am J Clin Pathol. 2004;122:499–505.
31. Bergman R, Dromi R, Trau H, et al. The pattern of HMB-45 antibody staining Signet ring cell melanoma
in compound Spitz nevi. Am J Dermatopathol. 1995;17:542–546.
32. Bastian BC, Wesselmann U, Pinkel D, et al. Molecular cytogenetic analy- 1. Sheibani K, Battifora H. Signet-ring cell melanoma. A rare morphologic variant
sis of Spitz nevi shows clear differences to melanoma. J Invest Dermatol. of malignant melanoma. Am J Surg Pathol. 1988;12:28–34.
1999;113:1065–1069. 2. Bonetti F, Colombari R, Zamboni G, et al. Signet-ring cell melanoma - S100
33. Bastian BC, Olshen AB, LeBoit PE, et al. Classifying melanocytic tumors based negative. Am J Surg Pathol. 1989;13:522–523.
on DNA copy number changes. Am J Pathol. 2003;163:1765–1770. 3. Nakhleh RE, Wick MR, Rocamora A, et al. Morphologic diversity in malignant
34. Bastian BC, LeBoit PE, Pinkel D. Mutations and copy number increase of melanomas. Am J Clin Pathol. 1990;93:731–740.
HRAS in Spitz nevi with distinctive histopathological features. Am J Pathol. 4. Al-Talib RK, Theaker JM. Signet-ring cell melanoma: light microscopic,
2000;157:967–972. immunohistochemical and ultrastructural features. Histopathology. 1991;18:
35. Harvell JD, Bastian BC, LeBoit PE. Persistent (recurrent) Spitz nevi: a histo- 572–575.
pathologic, immunohistochemical, and molecular pathologic study of 22 cases. 5. Eckert F, Baricfievic B, Landthaler M, et al. Metastatic signet-ring cell mela-
Am J Surg Pathol. 2002;26:654–661. noma in a patient with an unknown primary tumor. J Am Acad Dermatol.
36. Massi D, Tomasini C, Senetta R, et al. Atypical Spitz tumors in patients younger 1992;26:870–875.
than 18 years. J Am Acad Dermatol. 2015;72:37–46. 6. LiVolsi VA, Brooks JJ, Soslow R, et al. Signet cell melanocytic lesions. Mod
37. Gerami P, Cooper C, Bajaj S, et al. Outcomes of atypical spitz tumors with Pathol. 1992;5:515–520.
chromosomal copy number aberrations and conventional melanomas in chil- 7. Botev IN. Metastatic signet-ring cell melanoma. J Am Acad Dermatol.
dren. Am J Surg Pathol. 2013;37:1387–1394. 1993;29:510–511.
38. Gerami P, Scolyer RA, Xu X, et al. Risk assessment for atypical spitzoid mela- 8. Tsang WY, Chan JK, Chow LT. Signet-ring cell melanoma mimicking adenocar-
nocytic neoplasms using FISH to identify chromosomal copy number aberra- cinoma. Acta Cytol. 1993;37:559–562.
tions. Am J Surg Pathol. 2013;37:676–684. 9. Won JH, Ahn SK, Lee WS, et al. Signet-ring cell melanoma: poor prognostic
39. Lee DA, Cohen JA, Twaddell WS, et al. Are all melanomas the same? Spitzoid indicator. Br J Dermatol. 1994;136:135–137.
melanoma is a distinct subtype of melanoma. Cancer. 2006;106:907–913. 10. Niemann TH, Thomas PA. Melanoma with signet-ring cells in a peritoneal effu-
40. van Dijk MC, Bernsen MR, Ruiter DJ. Analysis of mutations in B-RAF, N-RAS, sion. Diagn Cytopathol. 1995;12:241–243.
and H-RAS genes in the differential diagnosis of Spitz nevus and spitzoid mela- 11. Breier F, Feldman R, Fellenz C, et al. Primary invasive signet-ring cell mela-
noma. Am J Surg Pathol. 2005;29:1145–1151. noma. J Cutan Pathol. 1999;26:533–536.
41. Wiesner T, He J, Yelensky R, et al. Kinase fusions are frequent in Spitz tumours 12. Bastian BC, Kutzner H, Yen TSB, et al. Signet-ring cell formations in cutaneous
and spitzoid melanomas. Nat Commun. 2014;5:3116. neoplasms. J Am Acad Dermatol. 1999;41:606–613.
42. Wiesner T, Kutzner H, Cerroni L, et al. Genomic aberrations in spitzoid mela- 13. Rutten A, Huschka U, Requena C, et al. Primary cutaneous signet-ring cell mel-
nocytic tumours and their implications for diagnosis, prognosis and therapy. anoma: a clinico-pathologic and immunohistochemical study of two cases. Am
Pathology. 2016;48:113–131. J Dermatopathol. 2003;25:418–422.
43. Kiuru M, Jungbluth A, Kutzner H, et al. Spitz tumors: comparison of histo- 14. Kacerovska D, Sokol L, Michal M, et al. Primary cutaneous signet-ring cell
logical features in relationship to immunohistochemical staining for ALK and melanoma with pseudoglandular features, spindle cells and oncocytoid changes.
NTRK1. Int J Surg Pathol. 2016;24:200–206. Am J Dermatopathol. 2009;31:81–83.
44. Yeh I, de la Fouchardiere A, Pissaloux D, et al. Clinical, histopathologic, 15. Kocovski L, Alowami S. Signet-ring cell melanoma: a potential diagnostic
and genomic features of Spitz tumors with ALK fusions. Am J Surg Pathol. pitfall. Am J Dermatopathol. 2014;36:985–988.
2015;39:581–591. 16. Ishida M, Iwai M, Yoshida K, et al. Signet-ring cell melanoma with sentinel
45. Busam KJ, Kutzner H, Cerroni L, et al. Clinical and pathologic findings of lymph node metastasis: A case report with immunohistochemical analysis and
Spitz nevi and atypical Spitz tumors with ALK fusions. Am J Surg Pathol. review of the clinicopathological features. Oncol Lett. 2014;7:65–68.
2014;38:925–933. 17. Salviato T, Luzar B, Falconieri G. Lipoblastoid/signet-ring cell-rich Spitz nevus:
46. Yazdan P, Cooper C, Sholl LM, et al. Comparative analysis of atypical spitz a hitherto unreported microscopic variant. J Cutan Pathol. 2014;41:762–
tumors with heterozygous versus homozygous 9p21 deletions for clinical out- 764.
comes, histomorphology, BRAF mutation, and p16 expression. Am J Surg 18. Sabater Marco V, Escutia Muñoz B, Morera Faet A, et al. Signet ring cell mela-
Pathol. 2014;38:638–645. nocytic nevus: report of a case over trichilemmal cyst and review of the litera-
47. Busam KJ, Wanna M, Wiesner T. Multiple epithelioid Spitz nevi or tumors with ture. Am J Dermatopathol. 2012;34:e10–e14.
loss of BAP1 expression: a clue to a hereditary tumor syndrome. JAMA Derma-
tol. 2013;149:335–339. Rhabdoid melanoma
48. 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 1. Kacerovska D, Sokol L, Michal M, et al. Primary cutaneous signet-ring cell
Pathol. 2012;36:818–830. melanoma with pseudoglandular features, spindle cells and oncocytoid changes.
49. Fabrizi G, Massi G. Spitzoid malignant melanoma in teenagers: an entity with Am J Dermatopathol. 2009;31:81–83.
no better prognosis than that of other forms of melanoma. Histopathology. 2. Weeks DA, Beckwith JB, Mierau GW, et al. Rhabdoid tumor of kidney. A
2001;38:448–453. report of 111 cases from the National Wilms’ Study Pathology Center. Am J
50. Zuckerman R, Maier JP, Guiney WB, et al. Pediatric melanoma: confirming the Surg Pathol. 1989;13:439–458.
diagnosis with sentinel node biopsy. Ann Plast Surg. 2001;46:394–399. 3. Bittesini L, Dei Tos AP, Fletcher CDM. Metastatic malignant melanoma showing
51. Lohmann CM, Coit DG, Brady MS, et al. Sentinel lymph node biopsy in a rhabdoid phenotype: further evidence of a non-specific histological pattern.
patients with diagnostically controversial spitzoid melanocytic tumors. Am J Histopathology. 1992;20:167–170.
Surg Pathol. 2002;26:47–55. 4. Weeks DA, Beckwith JB, Mierau GW. Rhabdoid tumor: an entity or a pheno-
52. Su LD, Fullen DR, Sondak VK, et al. Sentinel lymph node biopsy for patients typefi. Arch Pathol Lab Med. 1989;113:113–114.
with problematic Spitzoid melanocytic lesions. Cancer. 2003;97:499–507. 5. Chang ES, Wick MR, Swanson PE, et al. Metastatic malignant melanoma with
53. Lohmann CM, Coit DG, Brady MS, et al. Sentinel lymph node biopsy in ‘rhabdoid’ features. Am J Clin Pathol. 1994;102:426–431.
patients with diagnostically controversial spitzoid melanocytic tumors. Am J 6. Laskin WB, Knittel DR. S100 protein and HMB-45 negative rhabdoid malig-
Surg Pathol. 2002;26:47–55. nant melanoma: a totally dedifferentiated malignant melanomafi. Am J Clin
54. Busam KJ, Pulitzer M. Sentinel lymph node biopsy for patients with diag- Pathol. 1995;103:772–773.
nostically controversial Spitzoid melanocytic tumors? Adv Anat Pathol. 7. Wick MR, Ritter JH, Dehner LP. Malignant rhabdoid tumors: clinicopathologic
2008;15:253–262. review and conceptual discussion. Semin Diagn Pathol. 1995;12:223–248.

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
References 1362.e11

8. Nakamura T, Matsumine A, Kato H, et al. Malignant melanoma with a rhab- 3. Sarode VR, Joshi K, Ravichandran P, et al. Myxoid variant of primary cutane-
doid phenotype exhibiting numerous solid tumor masses: a case report. Oncol ous malignant melanoma. Histopathology. 1992;20:186–187.
Rep. 2009;21:887–891. 4. Prieto VG, Kanik A, Salob S, et al. Primary cutaneous myxoid melanoma:
9. Gavino AC, Gillies EM. Metastatic rhabdoid melanoma: report of a case with a immunohistologic clues to a difficult diagnosis. J Am Acad Dermatol.
comparative review of the literature. J Cutan Pathol. 2008;35:337–342. 1994;30:335–339.
10. Abbott JJ, Amirkhan RH, Hoang MP. Malignant melanoma with a rhabdoid 5. McCluggage WG, Shah V, Toner PG. Primary cutaneous myxoid melanoma.
phenotype: histologic, immunohistochemical, and ultrastructural study of a case Histopathology. 1996;28:179–182.
and review of the literature. Arch Pathol Lab Med. 2004;128:686–688. 6. Collina G, Taccagni GL, Maiorana A. Myxoid metastases of melanoma: report
11. Partham DM, Weeks DA, Beckwith JB. The clinicopathologic spectrum of puta- of three cases and review of the literature. Am J Dermatopathol. 1997;19:52–57.
tive extrarenal rhabdoid tumors. Am J Surg Pathol. 1994;18:1010–1029. 7. Hitchcock MG, White WL. Malicious masquerade: myxoid melanoma. Semin
12. Borek BT, McKee PH, Freeman JA, et al. Primary cutaneous rhabdoid malig- Diagn Pathol. 1998;15:195–202.
nant melanoma: a histologic and immunohistochemical study of three cases. Am 8. Hitchcock MG, McCalmont TH, White WL. Cutaneous melanoma with
J Dermatopathol. 1998;20:123–127. myxoid features: twelve cases with differential diagnosis. Am J Surg Pathol.
13. Tallon B, Bhawan J. Primary rhabdoid melanoma with clonal recurrence. Am J 1999;23:1506–1513.
Dermatopathol. 2009;31:200–204. 9. Inoue T, Misago N, Narisawa Y. Metastatic myxoid melanoma with partial
14. Fernández-Vega I, Santos-Juanes J, Fresno-Forcelledo MF. Primary amelanotic regression of the primary lesion. J Cutan Pathol. 2007;34:508–512.
rhabdoid melanoma of the forehead. Br J Dermatol. 2016;174:1156–1158. 10. Uzuncakmak TK, Zindanci I, Zemheri EI, et al. Primary myxoid melanoma
15. Kaneko T, Korekawa A, Akasaka E, et al. Primary amelanotic rhabdoid with dermoscopic findings. Dermatol Pract Concept. 2017;7:59–61.
melanoma: a case report with review of the literature. Case Rep Dermatol. 11. Harmse D, Saunders S, Evans A. Nonpigmented intradermal malignant mela-
2015;7:292–297. noma with cribiform, myxoid, and spindle cell growth patterns. Am J Dermato-
16. Ishida M, Iwai M, Yoshida K, et al. Rhabdoid melanoma: a case report with pathol. 2010;32:829–831.
review of the literature. Int J Clin Exp Pathol. 2014;7:840–843. 12. Vranic S. Melanoma with second myxoid stromal changes after personally
17. Chung BY, Ahn IS, Cho SI, et al. Primary malignant rhabdoid melanoma. Ann applied prolonged phototherapy. Am J Dermatopathol. 2009;31:723.
Dermatol. 2011;23:S155–S159. 13. Ulamec M, Soldo-Belic A, Vucic M, et al. Melanoma with second myxoid
stromal changes after personally applied prolonged phototherapy. Am J Derma-
topathol. 2008;30:185–187.
Balloon and clear cell melanoma
1. Gardner WA Jr, Vazquez MD. Balloon cell melanoma. Arch Pathol. Melanoma with neuroendocrine differentiation
1970;89:470–472.
2. Kao GF, Helwig EB, Graham JH. Balloon cell melanoma of the skin - a clini- 1. Banerjee SS, Eyden B. Divergent differentiation in malignant melanomas: a
copathologic study of 34 cases with histochemical, immunohistochemical, and review. Histopathology. 2008;52:119–129.
ultrastructural observations. Cancer. 1992;69:2942–2952. 2. Eyden B, Pandit D, Banerjee SS. Malignant melanoma with neuroendocrine
3. Requena L, de la Cruz A, Moreno C, et al. Animal type melanoma: a report differentiation: clinical, histological, immunohistochemical and ultrastructural
of a case with balloon-cell change and sentinel lymph node metastasis. Am J features of three cases. Histopathology. 2005;47:402–409.
Dermatopathol. 2001;23:341–346. 3. Katerji H, Childs JM, Bratton LE, et al. Primary esophageal melanoma with
4. Han JS, Won CH, Chang SE, et al. Primary cutaneous balloon cell melanoma: a aberrant CD56 expression: a potential diagnostic pitfall. Case Rep Pathol.
very rare variant. Int J Dermatol. 2014;53:e535–e536. 2017;2017:9052637.
5. Chavez-Alvarez S, Villarreal-Martinez A, Miranda-Maldonado I, et  al. 4. Ilardi G, Caroppo D, Varricchio S, et al. Anal melanoma with neuroendocrine
Balloon cell melanoma and its metastasis, a rare entity. Am J Dermatopathol. differentiation: report of a case. Int J Surg Pathol. 2015;23:329–332.
2017;39:404–411. 5. Pilozzi E, Cacchi C, Di Napoli A, et al. Primary malignant tumour of the
6. Martinez-Casimiro L, Sánchez Carazo JL, Alegre V. Balloon cell naevus. J Eur lung with neuroendocrine and melanoma differentiation. Virchows Arch.
Acad Dermatol Venereol. 2009;23:236–237. 2011;459:239–243.
7. Perez MT, Suster S. Balloon cell change in cellular blue nevus. Am J Dermato- 6. Coli A, Giacomini PG, Bigotti G, et al. Aberrant neurofilament protein and syn-
pathol. 1999;21:181–184. aptophysin expression in malignant melanoma of the nasal cavity. Histopathol-
8. Terayama K, Hirokawa M, Shimizu M, et al. Balloon melanoma cells mimick- ogy. 2004;44:193–195.
ing foamy histiocytes. Acta Cytol. 1999;43:325–326. 7. Kacerovska D, Michal M, Sosna B, et al. Carcinoid-like pattern in melanoma:
9. August C, Baba HA, Heinig J, et al. Endometrial metastasis of a ‘balloon’ report of 4 cases. Am J Dermatopathol. 2009;31:542–550.
cell melanoma mimicking a ‘xanthomatous endometritis. Pathologe. 8. Ishida M, Iwai M, Yoshida K, et al. A distinct histopathological variant of a
2001;22:145–150. malignant melanoma with perivascular pseudorosettes: A case report. Oncol
10. Northcutt AD. Epidermotropic xanthoma mimicking balloon cell melanoma. Lett. 2013;6:673–675.
Am J Dermatopathol. 2000;22:176–178.
11. Macak J, Krc I, Elleder M, et al. Clear cell melanoma of the skin with regressive Adenoid and pseudopapillary melanoma
changes. Histopathology. 1991;18:276–277.
12. Mourad WA, Mackay B, Ordonez NG, et al. Clear cell melanoma of the 1. Nakhleh RE, Wick MR, Rocamora A, et al. Morphologic diversity in malignant
bladder. Ultrastruct Pathol. 1993;17:463–468. melanoma. Am J Clin Pathol. 1990;93:731–740.
13. Ehara H, Takahashi Y, Saitoh A, et al. Clear cell melanoma of the renal pelvis 2. Valero-Torres A, Prieto VG, Nagarajan P, et al. Metastatic melanoma with pap-
presenting as a primary tumor. J Urol. 1997;157:634. illary features: a mimic and possible diagnostic pitfall. Am J Dermatopathol.
14. Nowak MA, Fatteh SM, Campbell TE. Glycogen-rich malignant melanoma 2017;39:468–470.
and glycogen-rich balloon cell malignant melanoma: frequency and pattern of 3. Young RH, Scully RE. Malignant melanoma metastatic to the ovary: a clinico-
PAS positivity in primary and metastatic melanomas. Arch Pathol Lab Med. pathologic analysis of 20 cases. Am J Surg Pathol. 1991;15:849–860.
1998;122:353–360. 4. Kartono F, Shitabata PK, Magro CM, et al. Discohesive malignant melanoma
15. Banerjee SS, Harris M. Morphological and immunophenotypic variations in simulating a bullous dermatoses. J Cutan Pathol. 2009;36:274–279.
malignant melanomas. Histopathology. 2000;36:387–402. 5. Ohnishi T, Hamano M, Watanabe S. Clinically verrucous and histologically dis-
16. Baehner FL, Ng B, Sudilovsky D. Metastatic balloon cell melanoma: a case cohesive melanoma. A case report with dermoscopic and immunohistochemical
report. Acta Cytol. 2005;49:543–548. observations. Australas J Dermatol. 2014;55:e21–e23.

Myxoid melanoma Blue nevus-like melanoma (balignant blue nevus)


1. Bhuta S, Mirra JM, Cochran AJ. Myxoid malignant melanoma: a previous 1. Murali R, McCarthy SW, Scolyer RA. Blue nevi and related lesions: a review
undescribed histologic pattern noted in metastatic lesions and a report of four highlighting atypical and newly described variants, distinguishing features and
cases. Am J Surg Pathol. 1986;10:203–211. diagnostic pitfalls. Adv Anat Pathol. 2009;16:365–382.
2. Urso C, Giannotti B, Bondi R. Myxoid melanoma of the skin. Arch Pathol Lab 2. Zembowicz A, Mihm MC. Dermal dendritic melanocytic proliferations: an
Med. 1990;114:527–528. update. Histopathology. 2004;45:433–451.

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
1362.e12 References

3. Borgenvik TL, Karlsvik TM, Ray S, et al. Blue nevus-like and blue nevus-as- 9. Hung T, Morin J, Munday WR, et al. Angiotropism in primary cutaneous
sociated melanoma: a comprehensive review of the literature. ANZ J Surg. melanoma with brain metastasis: a study of 20 cases. Am J Dermatopathol.
2017;87:345–349. 2013;35:650–654.
4. Toledo-Pastrana T, Rodríguez Pérez I, Eguino Gorrochategui P. Malignant 10. Wilmott J, Haydu L, Bagot M, et al. Angiotropism is an independent predic-
blue nevus: a challenge for dermatologists and dermatopathologists. [Article in tor of microscopic satellites in primary cutaneous melanoma. Histopathology.
English, Spanish]. Actas Dermosifiliogr. 2018;109:181–183. 2012;61:889–898.
5. Sugianto JZ, Ralston JS, Metcalf JS, et al. Blue nevus and “malignant blue 11. Lugassy C, Barnhill RL. Angiotropic melanoma and extravascular migratory
nevus:” A concise review. Semin Diagn Pathol. 2016;33:219–224. metastasis: a review. Adv Anat Pathol. 2007;14:195–201.
6. Nakamura Y, Shibata-Ito M, Nakamura Y, et al. Malignant blue nevus arising 12. Barnhill RL, Lugassy C. Angiotropic malignant melanoma and extravascular
in a giant congenital cellular blue nevus in an infant. Pediatr Dermatol. migratory metastasis: description of 36 cases with emphasis on a new mecha-
2012;29:651–655. nism of tumour spread. Pathology. 2004;36:485–490.
7. García-Rabasco A, Marín-Bertolín S, Esteve-Martínez A, et al. Dermal melano- 13. Gerami P, Shea C, Stone MS. Angiotropism in epidermotropic metastatic mela-
cytosis of the scalp associated to intracranial melanoma: malignant blue nevus, noma: another clue to the diagnosis. Am J Dermatopathol. 2006;28:429–433.
neurocutaneous melanosis, or neurocristic cutaneous hamartoma? Am J Derma- 14. Bald T, Quast T, Landsberg J, et al. Ultraviolet-radiation-induced inflamma-
topathol. 2012;34:177–181. tion promotes angiotropism and metastasis in melanoma. Nature. 2014;507:
8. Yeh I, Fang Y, Busam KJ. Melanoma arising in a large plaque-type blue nevus 109–113.
with subcutaneous cellular nodules. Am J Surg Pathol. 2012;36:1258–1263. 15. Lugassy C, Zadran S, Bentolila LA, et al. Angiotropism, pericytic mimicry and
9. Rodriguez HA, Ackerman LV. Cellular blue nevus: clinicopathologic study of extravascular migratory metastasis in melanoma: an alternative to intravascular
forty-five cases. Cancer. 1968;21:393–405. cancer dissemination. Cancer Microenviron. 2014;7:139–152.
10. Sterchi JM, Muss HB, Weidner N. Cellular blue nevus simulating metastatic 16. Kokta V, Hung T, Al Dhaybi R, et al. High prevalence of angiotropism in
melanoma: report of an unusually large lesion associated with nevus-cell aggre- congenital melanocytic nevi: an analysis of 53 cases. Am J Dermatopathol.
gates in regional lymph nodes. J Surg Oncol. 1987;36:71–75. 2013;35:180–183.
11. Temple-Camp CRE, Saxe N, King H. Benign and malignant cellular blue nevus: 17. Alder MJ, Beckstead J, White CR. Angiomatoid melanoma: a case of meta-
a clinicopathologic study of 30 cases. Am J Dermatopathol. 1988;10:289–296. static melanoma mimicking a vascular malignancy. Am J Dermatopathol.
12. Speakman JS, Phillips MJ. Cellular and malignant blue nevus complicat- 1997;19:606–609.
ing oculodermal melanosis (nevus of Ota syndrome). Can J Ophthalmol. 18. Banerjee SS, Harris M. Morphological and immunophenotypic variations in
1973;8:539–547. malignant melanoma. Histopathology. 2000;36:387–402.
13. Avidar I, Kessler E. ‘Atypical’ blue nevus: a benign variant of cellular blue 19. Baron JA, Monzon F, Galaria N, et al. Angiomatoid melanoma: a novel pattern
nevus. Presentation of three cases. Dermatologica. 1977;154:39–44. of differentiation in invasive periocular desmoplastic malignant melanoma.
14. Griewank KG, Müller H, Jackett LA, et al. SF3B1 and BAP1 mutations in blue Hum Pathol. 2000;31:1520–1522.
nevus-like melanoma. Mod Pathol. 2017;30:928–939. 20. Ramos-Rodríguez G, Ortiz-Hidalgo C. Primary angiomatoid melanoma as an
15. Lambert WC, Brodkin RH. Nodal and subcutaneous cellular blue nevi: a pseu- exceptional morphologic pattern in cutaneous melanoma. A case report and
dometastasizing pseudomelanoma. Arch Dermatol. 1984;120:367–370. review of the literature. [Article in English, Spanish]. Actas Dermosifiliogr.
16. Martin RC, Murali R, Scolyer RA, et al. So-called ‘malignant blue nevus’: a 2015;106:e13–e17.
clinicopathologic study of 23 patients. Cancer. 2009;115:2949–2955. 21. Fonda-Pascual P, Moreno-Arrones OM, Alegre-Sanchez A, et al. Primary cuta-
17. Zembowicz A. Blue nevi and related tumors. Clin Lab Med. 2017;37: neous angiomatoid melanoma. J Dtsch Dermatol Ges. 2018;16:345–347.
401–415. 22. Paolino G, Muscardin LM, Buccini P, et al. Angiomatoid melanoma: a dermo-
18. Barnhill RL, Argenyi Z, Berwick M, et al. Atypical cellular blue nevi (cellular scopic and pathologic challenge. G Ital Dermatol Venereol. 2018;[Epub ahead
blue nevi with atypical features): lack of consensus for diagnosis and distinction of print].
from cellular blue nevi and malignant melanoma (‘malignant blue nevus’). Am
J Surg Pathol. 2008;32:36–44. Metaplastic melanoma (melanoma with
19. Van Raamsdonk CD, Bezrookove V, Green G, et al. Frequent somatic muta- heterologous differentiation)
tions of GNAQ in uveal melanoma and blue naevi. Nature. 2009;457:
599–602. 1. Grunwald MH, Rothem A, Feuerman EJ. Metastatic malignant melanoma with
20. Dai J, Tetzlaff MT, Schuchter LM, et al. Histopathologic and mutational analy- cartilaginous metaplasia. Dermatologica. 1985;170:249–252.
sis of a case of blue nevus-like melanoma. J Cutan Pathol. 2016;43:776–780. 2. Brisigotti M, Moreno A, Llistosella E, et al. Malignant melanoma with osteo-
21. Hung T, Argenyi Z, Erickson L, et al. Cellular blue nevomelanocytic lesions: cartilagenous differentiation. Surg Pathol. 1989;1:73–80.
analysis of clinical, histological, and outcome data in 37 cases. Am J Dermato- 3. Moreno A, Lamarca J, Martínez R, et al. Osteoid and bone formation in des-
pathol. 2016;38:499–503. moplastic malignant melanoma. J Cutan Pathol. 1986;13:128–134.
22. Gammon B, Beilfuss B, Guitart J, et al. Fluorescence in situ hybridization 4. Lucas DR, Tazelaar HD, Unni KK, et al. Osteogenic melanoma: a rare variant
for distinguishing cellular blue nevi from blue nevus-like melanoma. J Cutan of malignant melanoma. Am J Surg Pathol. 1993;17:400–409.
Pathol. 2011;38:335–341. 5. Fukunaga M. Osteogenic melanoma. APMIS. 2005;113:296–300.
6. Ali AM, Wang WL, Lazar AJ. Primary chondro-osseous melanoma (chondro-
Angiotropic and angiomatoid sarcomatous and osteosarcomatous melanoma). J Cutan Pathol. 2018;45:146–
150.
(pseudovascular) melanoma
7. Emanuel PO, Idrees MT, Leytin A, et al. Aggressive osteogenic desmoplastic
1. Moreno A, Español I, Romagosa V. Angiotropic malignant melanoma: report of melanoma: a case report. J Cutan Pathol. 2007;34:423–426.
two cases. J Cutan Pathol. 1992;19:325–329. 8. Joana Devesa P, Labareda JM, Bártolo EA, et al. Cartilaginous melanoma: case
2. Shea CR, Kline MA, Lugo J, et al. Angiotropic metastatic malignant melanoma. report and review of the literature. An Bras Dermatol. 2013;88:403–407.
Am J Dermatopathol. 1995;17:58–62. 9. McKay KM, Deavers MT, Prieto VG. Chondroid melanoma metastatic to lung
3. Barnhill RL, Sagebiel RW, Lugassy C. Angiotropic melanoma: report of seven and skin showing SOX-9 expression: a potential diagnostic pitfall. Int J Surg
additional cases (abstract). J Cutan Pathol. 2000;27:548. Pathol. 2012;20:169–172.
4. Siegel DM, McClain SA. Angiotropic malignant melanoma: more common than 10. Ghadially FN. Osteosarcomatous changes in malignant melanoma. Am J Der-
we think? J Am Acad Dermatol. 2001;44:870–871. matopathol. 1992;14:180–181.
5. Moy AP, Duncan LM, Kraft S. Lymphatic invasion and angiotropism in primary 11. Nakagawa H, Imakado S, Nogita T, et al. Osteosarcomatous changes in malig-
cutaneous melanoma. Lab Invest. 2017;97:118–129. nant melanoma. Immunohistochemical and ultrastructural studies of a case. Am
6. Barnhill RL, Gupta K. Unusual variants of malignant melanoma. Clin Derma- J Dermatopathol. 1990;12:162–168.
tol. 2009;27:564–587. 12. Tscheudschilsuren G, Bosserhoff AK, Schlegel J, et al. Regulation of mes-
7. Barnhill RL, Busam KJ, From L, et al. Inter-observer concordance for the rec- enchymal stem cell and chondrocyte differentiation by MIA. Exp Cell Res.
ognition of angiotropism in human melanoma. Pigment Cell Melanoma Res. 2006;312:63–72.
2011;24:582–583. 13. Banerjee SS, Bishop PW, Nicholson CM, et al. Malignant melanoma showing
8. Kashani-Sabet M, Sagebiel RW, Ferreira CMM, et al. Vascular involve- smooth muscle differentiation. J Clin Pathol. 1996;49:950–951.
ment in the prognosis of primary cutaneous melanoma. Arch Dermatol. 14. Banerjee SS, Eyden B. Divergent differentiation in malignant melanomas: a
2001;137:1169–1173. review. Histopathology. 2008;52:119–129.

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
References 1362.e13

15. Shenjere P, Fisher C, Rajab R, et al. Melanoma with rhabdomyosarcomatous 15. Gavriilidis P, Michalopoulou I, Chatzikakidou K, et al. Pigmented epithelioid
differentiation: two further cases of a rare pathologic pitfall. Int J Surg Pathol. melanocytoma: a new concept encompassing animal-type melanoma and epithe-
2014;22:512–519. lioid blue nevus. BMJ Case Rep. 2013;2013:pii. bcr-2013-008865.
16. Reilly DJ, Volchek M, Ting JW, et al. Rhabdomyoblastic differentiation in met- 16. Phadke PA, Zembowicz A. Blue nevi and related tumors. Clin Lab Med.
astatic melanoma: making sense of a rare but complex form of mimicry. Int J 2011;31:345–358.
Surg Pathol. 2014;22:520–524. 17. Cohen JN, Joseph NM, North JP, et al. Genomic analysis of pigmented epithe-
17. Slaughter JC, Hardman JM, Kempe LG, et al. Neurocutaneous melanosis and lioid melanocytomas reveals recurrent alterations in PRKAR1A, and PRKCA
leptomeningeal melanomatosis in children. Arch Pathol. 1969;88:298–304. genes. Am J Surg Pathol. 2017;41:1333–1346.
18. Wahlström T, Saxén L. Malignant skin tumors of neural crest origin. Cancer. 18. Battistella M, Prochazkova-Carlotti M, Berrebi D, et al. Two congenital cases of
1976;38:2022–2026. pigmented epithelioid melanocytoma studied by fluorescent in situ hybridization
19. Banerjee SS, Menasce LP, Eyden BP, et al. Malignant melanoma showing gan- for melanocytic tumors: case reports and review of these recent topics. Derma-
glioneuroblastic differentiation: report of a unique case. Am J Surg Pathol. tology. 2010;221:97–106.
1999;23:582–588. 19. Muhlbauer A, Momtahen S, Mihm MC, et al. The correlation of the standard
20. Pellegrini AE, Scalamogna PA. Malignant melanoma with osteoid formation. 5 probe FISH assay with melanocytic tumors of uncertain malignant potential.
Am J Dermatopathol. 1990;12:607–611. Ann Diagn Pathol. 2017;28:30–36.
21. Denton KJ, Stretch J, Athanasou N. Osteoclast-like giant cells in malignant mel-
anoma. Histopathology. 1992;20:179–181. Epidermotropic metastatic melanoma
22. Li X, Savory S, Vandergriff T, et al. Osteoclast-like giant cells in malignant mel-
anoma: unintentional accident or strategic recruitment? Am J Dermatopathol. 1. Kornberg R, Harris M, Ackerman AB. Epidermotropically metastatic malig-
2018;40:303–304. nant melanoma: differentiating malignant melanoma metastatic to the epi-
23. Wasserman JK, Sekhon HS, Ayroud Y. Malignant melanoma with osteoclast-like dermis from malignant melanoma primary in the epidermis. Arch Dermatol.
differentiation. Int J Surg Pathol. 2015;23:478–482. 1978;114:67–69.
24. Houang M, Castillo C, La Marca S, et al. An unusual case of desmoplastic 2. Heenan PJ, Clay CD. Epidermotropic metastatic melanoma simulating multiple
melanoma containing an osteoclast-like giant cell-rich nodule. Am J Dermato- primary melanomas. Am J Dermatopathol. 1991;13:396–402.
pathol. 2015;37:299–304. 3. Abernathy JL, Soyer HP, Kerl H, et al. Epidermotropic metastatic malignant
25. Goel G, Rao S, Khurana N. Malignant melanoma with osteoclast-like giant melanoma simulating melanoma in situ: a report of 10 examples from two
cells: A report of two cases. J Cancer Res Ther. 2011;7:336–338. patients. Am J Surg Pathol. 1994;18:1140–1149.
26. Schmitt FC, Bittencourt A, Mendonca N, et al. Rhabdomyosarcoma in a con- 4. White WL, Hitchcock MG. Dying dogma: the pathological diagnosis of
genital pigmented nevus. Pediatr Pathol. 1992;12:93–98. epidermotropic metastatic malignant melanoma. Semin Diagn Pathol.
27. Kovalyshyn I, Braun R, Marghoob A. Congenital melanocytic naevi. Australas 1998;15:176–188.
J Dermatol. 2009;50:231–240, quiz 241–232. 5. Wick MR, Patterson JW. Cutaneous melanocytic lesions: selected problem
28. Christman MP, Kerner JK, Cheng C, et al. Rhabdomyosarcoma arising in a areas. Am J Clin Pathol. 2005;124(suppl):S52–S83.
giant congenital melanocytic nevus. Pediatr Dermatol. 2014;31:584–587. 6. Lestre S, João A, Ponte P, et al. Intraepidermal epidermotropic metastatic mela-
noma: a clinical and histopathological mimicker of melanoma in situ occurring
Pigment synthesizing melanoma in multiplicity. J Cutan Pathol. 2011;38:2011, 514–520.
7. Doshi B, Mahajan S, Khopkar US, et al. Epidermotropic metastatic mela-
(animal-type melanoma)
noma with perilesional depigmentation in an Indian male. Indian J Dermatol.
1. Levene A. Equine melanotic disease. Tumori. 1971;57:133–168. 2013;58:396–399.
2. Lerner AB, Cage GW. Melanoma in horses. Yale J Biol Med. 1973;46:646–649. 8. Kim HJ, So BJ, You MG, et al. Epidermotropic metastatic melanoma clinically
3. Vyas R, Keller JJ, Honda K, et al. A systematic review and meta-analysis of resembling agminated spitz nevi. Ann Dermatol. 2014;26:628–631.
animal-type melanoma. J Am Acad Dermatol. 2015;73:1031–1039. 9. Ratushny V, Kraft S, Moschella SL, et al. A symmetric eczematous eruption
4. Levene A. Disseminated dermal melanocytosis terminating in melanoma: harboring thousands of melanocytic lesions. JAMA Dermatol. 2016;152:1021–
a human condition resembling equine melanotic disease. Br J Dermatol. 1024.
1979;101:197–205. 10. Skala SL, Arps DP, Zhao L, et al. Comprehensive histopathological compari-
5. Crowson AN, Magro CM, Mihm MC. Malignant melanoma with prominent son of epidermotropic/dermal metastatic melanoma and primary nodular mela-
pigment synthesis. ‘Animal type’ melanoma - a clinical study of six cases with a noma. Histopathology. 2018;72:472–480.
consideration of other melanocytic neoplasms with prominent pigment synthe- 11. Bahrami S, Cheng L, Wang M, et al. Clonal relationships between epidermo-
sis. Hum Pathol. 1999;30:543–550. tropic metastatic melanomas and their primary lesions: a loss of heterozygosity
6. Antony FC, Sanclemente G, Shaikh H, et al. Pigment synthesizing mela- and X-chromosome inactivation-based analysis. Mod Pathol. 2007;20:821–827.
noma (so-called animal type melanoma): a clinicopathological study of 14 12. Gershenwald JE, Scolyer RA, Hess KR, et al. Melanoma staging: Evidence-based
cases of a poorly known distinctive variant of melanoma. Histopathology. changes in the American Joint Committee on Cancer eighth edition cancer
2006;48:754–762. staging manual. CA Cancer J Clin. 2017;67:472–492.
7. Bax MJ, Brown MD, Rothberg PG, et al. Pigmented epithelioid melanocytoma
(animal-type melanoma): An institutional experience. J Am Acad Dermatol. Blue nevus-like metastatic melanoma
2017;77:328–332.
8. Zembowicz A, Carney JA, Mihm MC. Pigmented epithelioid melanocytoma: 1. Busam KJ. Metastatic melanoma to the skin simulating blue nevus. Am J Surg
a low-grade melanocytic tumor with metastatic potential indistinguishable Pathol. 1999;23:276–282.
from animal-type melanoma and epithelioid blue nevus. Am J Surg Pathol. 2. Pouryazdanparast P, Newman M, Mafee M, et al. Distinguishing epithelioid
2004;28:31–40. blue nevus from blue nevus-like cutaneous melanoma metastasis using fluores-
9. Liu Q, Tong D, Liu G, et al. Carney complex with PRKAR1A gene mutation: A cence in-situ hybridization. Am J Surg Pathol. 2009;33:1396–1400.
case report and literature review. Medicine (Baltimore). 2017;96:e8999. 3. Baird DS, Ioffreda MD, Helm K, et al. Lentigo maligna melanoma with local and
10. Stratakis CA. Carney complex: A familial lentiginosis predisposing to a variety distant blue nevus-like metastases. Am J Dermatopathol. 2015;37:e126–e128.
of tumors. Rev Endocr Metab Disord. 2016;17:367–371. 4. Busam KJ, Fang Y, Jhanwar S, et al. Diagnosis of blue nevus-like metastatic
11. Kirschner LS, Carney JA, Pack SD, et al. Mutations of the gene encoding the uveal melanoma confirmed by fluorescence in situ hybridization (FISH) for
protein kinase A type I-alpha regulatory subunit in patients with the Carney monosomy 3. J Cutan Pathol. 2012;39:621–625.
complex. Nat Genet. 2000;26:89–92.
12. Settas N, Faucz FR, Stratakis CA. Succinate dehydrogenase (SDH) deficiency, Desmoplastic and neurotropic melanoma
Carney triad and the epigenome. Mol Cell Endocrinol. 2018;469:107–111.
13. Schaefer IM, Hornick JL, Bovée JVMG. The role of metabolic enzymes in mes- 1. Conley L, Lattes A, Orr W. Desmoplastic melanoma. Cancer. 1971;28:914–936.
enchymal tumors and tumor syndromes: genetics, pathology, and molecular 2. Reed RJ, Leonard DD. Neurotropic melanoma: a variant of desmoplastic mela-
mechanisms. Lab Invest. 2018;98:414–426. noma. Am J Surg Pathol. 1979;3:301–311.
14. Mandal RV, Murali R, Lundquist KF, et al. Pigmented Epithelioid Mela- 3. Quinn MJ, Crotty KA, Thompson JF, et al. Desmoplastic and desmoplas-
nocytoma: Favorable Outcome After 5-year Follow-up. Am J Surg Pathol. tic neurotropic melanoma. Experience with 280 patients. Cancer. 1998;83:
2009;33:1778–1782. 1128–1135.

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
1362.e14 References

4. Ward WH, Lambreton F, Goel N, et al. Clinical presentation and staging of 35. George E, McClain SE, Slingluff CL, et al. Subclassification of desmoplastic
melanoma. In: Ward WH, Farma JM, eds. Cutaneous Melanoma: Etiology and melanoma: pure and mixed variants have significantly different capacities for
Therapy [Internet]. Brisbane (AU): Codon Publications; 2017:[Chapter 6]. lymph node metastasis. J Cutan Pathol. 2009;36:425–432.
5. Mauer S, Vogt T, Pföhler C, et al. Errors in dermatology: desmoplastic mela- 36. Scolyer RA, Thompson JF. Desmoplastic melanoma: a heterogeneous entity in
noma - the amazing monster. J Dtsch Dermatol Ges. 2017;15:334–336. which subclassification as ‘pure’ or ‘mixed’ may have important prognostic sig-
6. Busam KJ. Desmoplastic melanoma. Clin Lab Med. 2011;31:321–330. nificance. Ann Surg Oncol. 2005;12:197–199.
7. Chen LL, Jaimes N, Barker CA, et al. Desmoplastic melanoma: a review. J Am 37. Livestro DP, Muzikansky A, Kaine EM, et al. Biology of desmoplastic mel-
Acad Dermatol. 2013;68:825–833. anoma: a case-control comparison with other melanomas. J Clin Oncol.
8. McCarthy SW, Scolyer RA, Palmer AA. Desmoplastic melanoma: a diagnostic 2005;23:6739–6746.
trap for the unwary. Pathology. 2004;36:445–451. 38. Shaw HM, Quinn MJ, Scolyer RA, et al. Survival in patients with desmoplastic
9. Jain S, Allen PW. Desmoplastic melanoma and its variants: a study of 45 cases. melanoma. J Clin Oncol. 2006;24:e12. author reply e13.
Am J Surg Pathol. 1989;13:358–373. 39. Conic RZ, Ko J, Allam SH, et al. Mixed versus pure variants of desmoplastic
10. Jaroszewski DE, Pockaj BA, DiCaudo DJ, et al. The clinical behavior of desmo- melanoma: the Cleveland Clinic experience. Ann Plast Surg. 2018;80:277–281.
plastic melanoma. Am J Surg. 2001;182:590–595. 40. Quinn MJ, Crotty KA, Thompson JF, et al. Desmoplastic and desmoplastic neu-
11. Beenken S, Byers R, Smith JL, et al. Desmoplastic melanoma. Histologic cor- rotropic melanoma: experience with 280 patients. Cancer. 1998;83:1128–1135.
relation with behavior and treatment. Arch Otolaryngol Head Neck Surg. 41. Busam KJ. Cutaneous desmoplastic melanoma. Adv Anat Pathol. 2005;12:
1989;115:374–379. 92–102.
12. Devara JVS, Moss ALH, Briggs JC. Desmoplastic melanoma: a clinico-patho- 42. Bryant E, Ronan S, Feliz E, et al. Desmoplastic melanoma: a study by conven-
logical review. Brit J Plast Surg. 1992;45:595–598. tional and electron microscopy. Am J Dermatopathol. 1982;4:467–474.
13. Egbert B, Kempson R, Sagebiel R. Desmoplastic melanoma. A clinicopathologic 43. Maurichi A, Miceli R, Camerini T, et al. Pure desmoplastic melanoma: a mela-
study of 25 cases. Cancer. 1988;62:2033–2041. noma with distinctive clinical behavior. Ann Surg. 2010;252:1052–1057.
14. Walsh N, Roberts J, Orr W, et al. Desmoplastic melanoma. A clinicopathologic 44. Frolow GR, Shapiro L, Brownstein MH. Desmoplastic melanoma. Arch Derma-
study of 14 cases. Arch Pathol Lab Med. 1988;112:922–927. tol. 1975;111:753–754.
15. Bruijn JA, Mihm MC Jr, Barnhill RL. Desmoplastic melanoma. Histopathology. 45. Wharton JM, Carlson JA, Mihm MC Jr. Desmoplastic malignant melanoma:
1992;20:197–205. diagnosis of early clinical lesions. Hum Pathol. 1999;33:537–542.
16. Bruijn JA, Salasche S, Sober AJ, et al. Desmoplastic melanoma: clinicopatho- 46. de Almeida LS, Requena L, Rutten A, et al. Desmoplastic malignant mel-
logic aspects of six cases. Dermatology. 1992;185:3–8. anoma: a clinicopathologic analysis of 113 cases. Am J Dermatopathol.
17. Chen JY, Hruby G, Scolyer RA, et al. Desmoplastic neurotropic melanoma: a 2008;30:207–215.
clinicopathologic analysis of 128 cases. Cancer. 2008;113:2770–2778. 47. Banerjee SS, Harris M. Morphological and immunophenotypic variations in
18. Smithers BM, McLeod GR, Little JH. Desmoplastic, neural transforming malignant melanoma. Histopathology. 2000;36:387–402.
and neurotropic melanoma: a review of 45 cases. Aust N Z J Surg. 1990;60: 48. Ackerman AB, Godomski J. Neurotropic melanoma and other neurotropic neo-
967–972. plasms in the skin. Am J Dermatopathol. 1984;6(suppl 1):63–80.
19. Anstey A, McKee PH, Wilson-Jones E. Desmoplastic melanoma: a clinicopath- 49. Van Raamsdonk CD, Deo M. Links between Schwann cells and melanocytes in
ological study of 25 cases. Br J Dermatol. 1993;129:359–371. development and disease. Pigment Cell Melanoma Res. 2013;26:634–645.
20. Smithers BM, McLeod GR, Little JH. Desmoplastic melanoma: patterns of 50. Moreno A, Español I, Romagosa V. Angiotropic melanoma: report of two cases.
recurrence. World J Surg. 1992;16:186–190. J Cutan Pathol. 1992;19:325–329.
21. Valensi QJ. Desmoplastic melanoma: a report of two additional cases. Cancer. 51. Moreno A, Lamarca J, Martínez R, et al. Osteoid and bone formation in des-
1977;39:286–292. moplastic malignant melanoma. J Cutan Pathol. 1986;13:128–134.
22. Lee JY-Y, Kapadia SB, Musgrave RH, et al. Neurotropic melanoma occurring in 52. Anstey A, Cerio C, Ramnarain N, et al. Desmoplastic melanoma: an immuno-
a stable burn scar. J Cutan Pathol. 1992;19:145–150. histochemical study of 25 cases. Am J Dermatopathol. 1994;16:14–22.
23. Khan F, Strohl A, Allen PD, et al. Desmoplastic melanoma of the head and 53. Warner TFCS, Lloyd AV, Hafex GR, et al. Immunocytochemistry of neurotropic
neck: incidence and survival, 1992-2013. Otolaryngol Head Neck Surg. melanoma. Cancer. 1984;53:254–257.
2017;157:648–656. 54. Carlston JA, Dickersin GR, Sober AJ, et al. Desmoplastic neurotropic mela-
24. Pawlik TM, Ross MI, Prieto VG, et al. Assessment of the role of sentinel noma: a clinicopathologic analysis of 28 cases. Cancer. 1995;75:478–494.
lymph node biopsy for primary cutaneous desmoplastic melanoma. Cancer. 55. Longacre TA, Egbert BM, Rouse RV. Desmoplastic and spindle cell melanoma:
2006;106:900–906. an immunohistochemical study. Am J Surg Pathol. 1996;20:1489–1500.
25. Ariyan CE, Coit DG. Clinical aspects of sentinel lymph node biopsy in mela- 56. Clarkson KS, Sturdgess IC, Molyneux AJ. The usefulness of tyrosinase in the
noma. Semin Diagn Pathol. 2008;25:86–94. immunohistochemical assessment of melanocytic lesions: a comparison of the
26. Sassen S, Shaw HM, Colman MH, et al. The complex relationships between novel T311 antibody (anti-tyrosinase) with S100, HMB45 and A103 (anti-
sentinel node positivity, patient age, and primary tumor desmoplasia: anal- melan-A). J Clin Pathol. 2001;54:196–200.
ysis of 2303 melanoma patients treated at a single center. Ann Surg Oncol. 57. Ohsie SJ, Sarantopoulos GP, Cochran AJ, et al. Immunohistochemical charac-
2008;15:630–637. teristics of melanoma. J Cutan Pathol. 2008;35:433–444.
27. Gyorki DE, Busam K, Panageas K, et al. Sentinel lymph node biopsy for patients 58. Busam KJ, Chen YT, Old LJ, et al. Expression of melan-A (MART1) in
with cutaneous desmoplastic melanoma. Ann Surg Oncol. 2003;10:403–407. benign melanocytic and primary cutaneous melanoma. Am J Surg Pathol.
28. Mohebati A, Ganly I, Busam KJ, et al. The role of sentinel lymph node biopsy 1998;22:976–982.
in the management of head and neck desmoplastic melanoma. Ann Surg Oncol. 59. Mehregan DR, Hamzavi I. Staining of melanocytic neoplasms by melanoma
2012;19:4307–4313. antigen recognized by T cells. Am J Dermatopathol. 2000;22:247–250.
29. Eppsteiner RW, Swick BL, Milhem MM, et al. Sentinel node biopsy for head 60. Weissinger SE, Lennerz JK. Comparison of MelanA/MART-1 and HMB45
and neck desmoplastic melanoma: not a given. Otolaryngol Head Neck Surg. labeling in desmoplastic melanoma. Mod Pathol. 2014;27:1421–1423.
2012;147:271–274. 61. Busam KJ, Iversen K, Coplan KC, et al. Analysis of microphthalmia transcrip-
30. Dunne JA, Powell BWEM. Re: Sentinel node biopsy in desmoplastic thin tion factor expression in normal tissues and tumors, and comparison of its
melanoma: Histogenetic recommendations. J Plast Reconstr Aesthet Surg. expression with S100 protein, gp100, and tyrosinase in desmoplastic melanoma.
2018;71:440. Am J Surg Pathol. 2001;25:197–204.
31. Broer PN, Walker ME, Goldberg C, et al. Desmoplastic melanoma: a 12-year 62. Koch MB, Shih I-M, Weiss SW, et al. Microphthalmia transcription factor and
experience with sentinel lymph node biopsy. Eur J Surg Oncol. 2013;39: melanoma adhesion molecule expression distinguish desmoplastic/spindle cell
681–685. melanoma from morphologic mimics. Am J Surg Pathol. 2001;25:58–64.
32. Han D, Zager JS, Yu D, et al. Desmoplastic melanoma: is there a role for senti- 63. Nonaka D, Chiriboga L, Rubin BP. Sox10: a pan-schwannian and melanocytic
nel lymph node biopsy? Ann Surg Oncol. 2013;20:2345–2351. marker. Am J Surg Pathol. 2008;32:1291–1298.
33. Martinez SR, Shah DR, Yang AD, et al. Sentinel lymph node biopsy in patients 64. Palla B, Su A, Binder S, et al. SOX10 expression distinguishes desmoplastic
with thick primary cutaneous melanoma: patterns of use and underuse utilizing melanoma from its histologic mimics. Am J Dermatopathol. 2013;35:576–
a population-based model. ISRN Dermatol. 2013;2013:315609. 581.
34. Dunne JA, Wormald JC, Steele J, et al. Is sentinel lymph node biopsy warranted 65. Mohamed A, Gonzalez RS, Lawson D, et al. SOX10 expression in malig-
for desmoplastic melanoma? A systematic review. J Plast Reconstr Aesthet Surg. nant melanoma, carcinoma, and normal tissues. Appl Immunohistochem Mol
2017;70:274–280. Morphol. 2013;21:506–510.

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
References 1362.e15

66. Tacha D, Qi W, Ra S, et al. A newly developed mouse monoclonal SOX10


antibody is a highly sensitive and specific marker for malignant melanoma, Melanoma in children
including spindle cell and desmoplastic melanomas. Arch Pathol Lab Med.
2015;139:530–536. 1. Urist M, Hynds Karnell L. The National Cancer Data Base: report on mela-
67. Nonaka D, Chiriboga L, Rubin BP. Differential expression of S100 protein noma. Cancer. 1994;74:782–788.
subtypes in malignant melanoma, and benign and malignant peripheral nerve 2. Whitehead D, Valery P, McWhirter W, et al. Incidence of cutaneous childhood
sheath tumors. J Cutan Pathol. 2008;35:1014–1019. melanoma in Queensland. Int J Cancer. 1995;63:765–768.
68. Skelton HG, Smith KJ, Laskin WB, et al. Desmoplastic malignant melanoma. J 3. Hoang M-TR, Friedlander SF. Rare cutaneous malignancies of childhood. Curr
Am Acad Dermatol. 1995;32:717–725. Opin Pediatr. 1999;11:464–470.
69. Riccioni L, Di Tommaso L, Collina G. Actin-rich desmoplastic malignant mela- 4. Ceballos P, Ruiz-Maldonaldo R, Mihm M Jr. Melanoma in children. N Engl J
noma: report of three cases. Am J Dermatopathol. 1999;21:537–541. Med. 1995;332:656–662.
70. Reiman HM, Goellner JR, Woods JE, et al. Desmoplastic melanoma of the head 5. Ruiz-Maldonado R, de la Luz Orozco-Covarrubius M. Malignant melanoma in
and neck. Cancer. 1987;60:2269–2274. children. Arch Dermatol. 1997;133:363–371.
71. Labrecque PG, Chung-Wong H, Winkelmann RK. On the nature of desmoplas- 6. Scalzo DA, Hida CA, Toth G, et al. Childhood melanoma: a clinicopathological
tic melanoma. Cancer. 1976;38:1205–1213. study of 22 cases. Melanoma Res. 1997;7:63–68.
72. Hilliard NJ, Krahl D, Sellheyer K. p16 expression differentiates between 7. Tate PS, Ronan SG, Feucht KA, et al. Melanoma in childhood and adoles-
desmoplastic Spitz nevus and desmoplastic melanoma. J Cutan Pathol. cence: clinical and pathological features of 48 cases. J Pediatr Surg. 1993;28:
2009;36:753–759. 217–222.
73. Blokhin E, Pulitzer M, Busam KJ. Immunohistochemical expression of p16 in 8. DeDavid M, Orlow SJ, Provost N, et al. A study of large congenital melano-
desmoplastic melanoma. J Cutan Pathol. 2013;40:796–800. cytic nevi and associated malignant melanomas: review of cases in the New
74. Lazova R, Tantcheva-Poor I, Sigal AC. P75 nerve growth factor receptor stain- York University Registry and the world literature. J Am Acad Dermatol.
ing is superior to S100 in identifying spindle cell and desmoplastic melanoma. J 1997;36:409–416.
Am Acad Dermatol. 2010;63:852–858. 9. McCarthy SW, Crotty KA, Palmer AA, et al. Cutaneous malignant melanoma in
75. Plaza JA, Bonneau P, Prieto V, et al. Desmoplastic melanoma: an updated teenagers. Histopathology. 1994;24:453–461.
immunohistochemical analysis of 40 cases with a proposal for an additional 10. Mehregan AH, Mehregan DA. Malignant melanoma in childhood. Cancer.
panel of stains for diagnosis. J Cutan Pathol. 2016;43:313–323. 1993;71:4096–4103.
76. Miller DD, Emley A, Yang S, et al. Mixed versus pure variants of desmoplas- 11. Barnhill RL, Flotte TJ, Fleischi M, et al. Cutaneous melanoma and atypical
tic melanoma: a genetic and immunohistochemical appraisal. Mod Pathol. Spitz tumors in childhood. Cancer. 1995;76:1833–1845.
2012;25:505–515. 12. Conti EMS, Cercato MC, Gatta G, et al. Childhood melanoma in Europe since
77. Le Guellec S, Macagno N, Velasco V, et al. Loss of H3K27 trimethylation is 1978: a population-based survival study. Eur J Cancer. 2001;37:780–784.
not suitable for distinguishing malignant peripheral nerve sheath tumor from 13. Mones JM, Ackerman AB. Melanomas in prepubescent children. Am J Derma-
melanoma: a study of 387 cases including mimicking lesions. Mod Pathol. topathol. 2003;25:223–238.
2017;30:1677–1687. 14. Handfield-Jones SE, Smith NP. Malignant melanoma in childhood. Br J Derma-
78. Trejo O, Reed JA, Prieto VG. Atypical cells in human cutaneous re-excision tol. 1996;134:607–614.
scars for melanoma express p75NGFR, C56/N-CAM and GAP-43: evidence of 15. Saiyed FK, Hamilton EC, Austin MT. Pediatric melanoma: incidence, treatment,
early Schwann cell differentiation. J Cutan Pathol. 2002;29:397–406. and prognosis. Pediatric Health Med Ther. 2017;8:39–45.
79. Murali R, Loughman NT, McKenzie PR, et al. Cytologic features of metastatic 16. Brecht IB, De Paoli A, Bisogno G, et al. Pediatric patients with cutaneous mela-
and recurrent melanoma in patients with primary cutaneous desmoplastic mel- noma: A European study. Pediatr Blood Cancer. 2018;[Epub ahead of print].
anoma. Am J Clin Pathol. 2008;130:715–723. 17. Carrera C, Scope A, Dusza SW, et al. Clinical and dermoscopic characterization
80. From L, Hanna W, Kahn HJ, et al. Origin of the desmoplasia in desmoplastic of pediatric and adolescent melanomas: Multicenter study of 52 cases. J Am
melanoma. Hum Pathol. 1983;149:1072–1080. Acad Dermatol. 2018;78:278–288.
81. Valensi QJ. Desmoplastic melanoma: a light and electron microscopic study of 18. Eggen CAM, Durgaram VVL, van Doorn R, et al. Incidence and relative sur-
two cases. Cancer. 1979;43:1148–1155. vival of melanoma in children and adolescents in the Netherlands, 1989-2013.
82. Warner TF, Hafez GR, Finch RE, et al. Schwann cell features in neurotropic J Eur Acad Dermatol Venereol. 2018;32:956–961.
melanoma. J Cutan Pathol. 1981;8:177–187. 19. Steliarova-Foucher E, Colombet M, Ries LAG, et al. International incidence of
83. Dimaio SM, Mackay B, Smith JL Jr, et al. Neurosarcomatous transformation in childhood cancer, 2001-10: a population-based registry study. Lancet Oncol.
malignant melanoma: an ultrastructural study. Cancer. 1982;50:2345–2354. 2017;18:719–731.
84. Shain AH, Garrido M, Botton T, et al. Exome sequencing of desmoplastic mel- 20. Wong JR, Harris JK, Rodriguez-Galindo C, et al. Incidence of childhood and
anoma identifies recurrent NFKBIE promoter mutations and diverse activating adolescent melanoma in the United States: 1973-2009. Pediatrics. 2013;131:
mutations in the MAPK pathway. Nat Genet. 2015;47:1194–1199. 846–854.
85. Kiuru M, Busam KJ. The NF1 gene in tumor syndromes and melanoma. Lab 21. Lambie M, Nadler C, Glat P, et al. Infantile melanoma–a triple threat: diagnosis
Invest. 2017;97:146–157. and management. Ann Plast Surg. 2011;67:85–89.
86. Mahalingam M. NF1 and neurofibromin: emerging players in the genetic land- 22. Marghoob AA, Schoenbach SP, Kopf AW, et al. Large congenital melanocytic
scape of desmoplastic melanoma. Adv Anat Pathol. 2017;24:1–14. nevi and risk for the development of malignant melanoma: a prospective study.
87. Wiesner T, Kiuru M, Scott SN, et al. NF1 mutations are common in desmoplas- Arch Dermatol. 1996;132:170–175.
tic melanoma. Am J Surg Pathol. 2015;39:1357–1362. 23. Alvarez-Mendoza A, Reyes-Esparza J, Ruiz-Maldonado R, et al. Malignant
88. Kadokura A, Frydenlund N, Leone DA, et al. Neurofibromin protein loss in melanoma in children and congenital melanocytic nevi: DNA content and cell
desmoplastic melanoma subtypes: implicating NF1 allelic loss as a distinct cycle analysis by flow cytometry. Dev Pathol. 2001;4:73–81.
genetic driver? Hum Pathol. 2016;53:82–90. 24. Jen M, Murphy M, Grant-Kels JM. Childhood melanoma. Clin Dermatol.
89. Yang S, Leone D, Frydenlund N, et al. Frequency of telomerase reverse tran- 2009;27:529–536.
scripter promoter mutations in desmoplastic melanoma subtypes: analyses of 76 25. Kollipara R, Cooley LD, Horii KA, et al. Spitzoid melanoma in a child with
cases. Melanoma Res. 2016;26:361–366. Li-Fraumeni syndrome. Pediatr Dev Pathol. 2014;17:64–69.
90. Weissinger SE, Frick M, Möller P, et al. Performance testing of RREB1, 26. Magnusson S, Gisselsson D, Wiebe T, et al. Prevalence of germline TP53
MYB, and CCND1 fluorescence in situ hybridization in spindle-cell and des- mutations and history of Li-Fraumeni syndrome in families with childhood
moplastic melanoma argues for a two-step test algorithm. Int J Surg Pathol. adrenocortical tumors, choroid plexus tumors, and rhabdomyosarcoma: a pop-
2017;25:148–157. ulation-based survey. Pediatr Blood Cancer. 2012;59:846–853.
91. Kaneishi NK, Cockerell CJ. Histologic differentiation of desmoplastic mela- 27. Gironi LC, Colombo E, Farinelli P, et al. Germline CDKN2A mutations in
noma from cicatrices. Am J Dermatopathol. 1998;20:128–134. childhood melanoma: a case of melanoma-pancreatic cancer syndrome. Int J
92. Robson A, Allen P, Hollowood K. S100 expression in cutaneous scars: a poten- Dermatol. 2015;54:e553–e555.
tial diagnostic pitfall in the diagnosis of desmoplastic melanoma. Histopathol- 28. Tannous ZS, Mihm MC Jr, Sober AJ, et al. Congenital melanocytic nevi: clinical
ogy. 2001;38:135–140. and histopathologic features, risk of melanoma, and clinical management. J Am
93. Jackett LA, McCarthy SW, Scolyer RA. SOX10 expression in cutaneous scars: a Acad Dermatol. 2005;52:197–203.
potential diagnostic pitfall in the evaluation of melanoma re-excision specimens. 29. Kinsler VA, O’Hare P, Bulstrode N, et al. Melanoma in congenital melanocytic
Pathology. 2016;48:626–628. naevi. Br J Dermatol. 2017;176:1131–1143.

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
1362.e16 References

30. Vergier B, Laharanne E, Prochazkova-Carlotti M, et al. Proliferative nodules vs 5. Rodić N, Taube JM, Manson P, et al. Locally invasive dermal squamomela-
melanoma arising in giant congenital melanocytic nevi during childhood. JAMA nocytic tumor with matrical differentiation: a peculiar case with review of the
Dermatol. 2016;152:1147–1151. literature. Am J Dermatopathol. 2013;35:e72–e76.
31. Pappo AS. Melanoma in children and adolescents. Eur J Cancer. 2003;39: 6. Amerio P, Carbone A, Auriemma M, et al. Metastasizing dermal squamomelano-
2651–2661. cytic tumour: more evidences. J Eur Acad Dermatol Venereol. 2011;25:734–735.
32. Pustišek N, Šitum M, Mataija M, et al. Malignant melanoma in childhood and 7. Amerio P, Tracanna M, Di Rollo D, et al. Metastasizing dermal squamomelano-
adolescence. J Eur Acad Dermatol Venereol. 2013;27:e256–e257. cytic tumour. J Eur Acad Dermatol Venereol. 2011;25:489–491.
33. Cordoro KM, Gupta D, Frieden IJ, et al. Pediatric melanoma: results of a large
cohort study and proposal for modified ABCD detection criteria for children. J Basomelanocytic tumor
Am Acad Dermatol. 2013;68:913–925.
34. Smith KJ, Barrett TL, Skelton HGIII, et al. Spindle and epithelioid cell 1. Amin SM, Cooper C, Yélamos O, et al. Combined cutaneous tumors with a
nevi with atypia and metastasis (malignant Spitz nevus). Am J Surg Pathol. melanoma component: A clinical, histologic, and molecular study. J Am Acad
1989;13:931–939. Dermatol. 2015;73:451–460.
35. Ghorbani Z, Dowlati Y, Mehregan AH. Amelanotic Spitzoid melanoma in the 2. Charlton RE. A melanomatous carcinoma. A case report and commentary. Am
burn scar of a child. Pediatr Dermatol. 1996;13:285–287. J Dermatopathol. 1984;6(suppl):221–229.
36. Fabrizi G, Massi G. Spitzoid malignant melanoma in teenagers: an entity with 3. Erickson LA, Myers JL, Mihm MC, et al. Malignant basomelanocytic tumor
no better prognosis than that of other forms of melanoma. Histopathology. manifesting as metastatic melanoma. Am J Surg Pathol. 2004;28:1393–1396.
2001;38:448–453. 4. Rodriguez J, Nonaka D, Kuhn E, et al. Combined high-grade basal cell car-
37. Aldrink JH, Selim MA, Diesen DL, et al. Pediatric melanoma: a single-institu- cinoma and malignant melanoma of the skin (‘malignant basomelanocytic
tion experience of 150 patients. J Pediatr Surg. 2009;44:1514–1521. tumor’): report of two cases and review of the literature. Am J Dermatopathol.
38. Neville HL, Andrassy RJ, Lally KP, et al. Lymphatic mapping with sentinel node 2005;27:314–318.
biopsy in pediatric patients. J Pediatr Surg. 2000;35:961–964. 5. Busam KJ, Halpern A, Marghoob AA. Malignant melanoma metastatic to a
39. Kogut KA, Flemming M, Pappo AS, et al. Sentinel lymph node biopsy for mel- basal cell carcinoma simulating the pattern of a basomelanocytic tumor. Am J
anoma in young children. J Pediatr Surg. 2000;35:965–966. Surg Pathol. 2006;30:133–136.
40. Zuckerman R, Maier JP, Gulney WB, et al. Pediatric melanoma: confirming the 6. Goeser M, DiMaio DJ. A colonization of basal cell carcinoma by malignant
diagnosis with sentinel node biopsy. Ann Plast Surg. 2001;46:394–399. melanoma in situ resembling a malignant basomelanocytic tumor. Am J Derma-
41. Paradela S, Fonseca E, Pita S, et al. Spitzoid melanoma in children: clinicopath- topathol. 2014;36:e179–e182.
ological study and application of immunohistochemistry as an adjunct diagnos-
tic tool. J Cutan Pathol. 2009;36:740–752. Lentiginous melanoma
42. Paradela S, Fonseca E, Pita-Fernandez S, et al. Prognostic factors for melanoma
in children and adolescents: a clinicopathologic, single-center study of 137 1. Weedon D. Lentiginous melanoma. J Cutan Pathol. 2009;36:1232.
patients. Cancer. 2010;116:4334–4344. 2. Davis T, Zembowicz A. Histological evolution of lentiginous melanoma: a
43. Paradela S, Fonseca E, Pita-Fernández S, et al. Spitzoid and non-spitzoid mela- report of five new cases. J Cutan Pathol. 2007;34:296–300.
noma in children: a prognostic comparative study. J Eur Acad Dermatol Vene- 3. King R, Page RN, Googe PB, et al. Lentiginous melanoma: a histologic
reol. 2013;27:1214–1221. pattern of melanoma to be distinguished from lentiginous nevus. Mod Pathol.
44. Stanelle EJ, Busam KJ, Rich BS, et al. Early-stage non-Spitzoid cutaneous mela- 2005;18:1397–1401.
noma in patients younger than 22 years of age at diagnosis: long-term follow-up 4. Milette F. Comment on the ‘histological evolution of lentiginous melanoma. J
and survival analysis. J Pediatr Surg. 2015;50:1019–1023. Cutan Pathol. 2008;35:88, author reply 89.
45. Busam KJ, Murali R, Pulitzer M, et al. Atypical spitzoid melanocytic tumors 5. Sabater-Marco V, García-Rabasco A, García-García JA, et al. Lentiginous mela-
with positive sentinel lymph nodes in children and teenagers, and comparison noma. A clinically malignant entity that histopathologically seems benign. Case
with histologically unambiguous and lethal melanomas. Am J Surg Pathol. study harbouring BRAF(V) (600R) mutation. J Eur Acad Dermatol Venereol.
2009;33:1386–1395. 2016;30:1197–1198.
46. Ferrara G, Gianotti R, Cavicchini S, et al. Spitz nevus, Spitz tumor, and spit- 6. King R. Lentiginous melanoma. Arch Pathol Lab Med. 2011;135:337–341.
zoid melanoma: a comprehensive clinicopathologic overview. Dermatol Clin. 7. Kossard S. Atypical lentiginous junctional naevi of the elderly and melanoma.
2013;31:589–598, viii. Australas J Dermatol. 2002;43:93–101.
47. Granter S, McKee PH, Calonje E, et al. Melanoma associated with blue nevus 8. Kossard S, Commens C, Symons M, et al. Lentiginous dysplastic naevi in the
and melanoma mimicking cellular blue nevus: a clinicopathologic study of 10 elderly: a potential precursor for malignant melanoma. Australas J Dermatol.
cases on the spectrum of so-called ‘malignant blue nevus. Am J Surg Pathol. 1991;32:27–37.
2001;25:316–323. 9. Weedon D. Lentiginous melanoma. J Cutan Pathol. 2009;36:1232.
48. Verzì AE, Bubley JA, Haugh AM, et al. A single-institution assessment of
superficial spreading melanoma (SSM) in the pediatric population: Molecular Primary dermal melanoma
and histopathologic features compared with adult SSM. J Am Acad Dermatol.
2017;77:886–892. 1. Bowen GM, Chang AE, Lowe L, et al. Solitary melanoma confined to the
49. Lu C, Zhang J, Nagahawatte P, et al. The genomic landscape of childhood and dermal and/or subcutaneous tissue: evidence for revisiting the staging classifica-
adolescent melanoma. J Invest Dermatol. 2015;135:816–823. tion. Arch Dermatol. 2000;136:1397–1399.
50. Navid F. Genetic alterations in childhood melanoma. Am Soc Clin Oncol Educ 2. Swetter SM, Ecker PM, Johnson DL, et al. Primary dermal melanoma: a distinct
Book. 2012;589–592. subtype of melanoma. Arch Dermatol. 2004;140:99–103.
51. Prieto-Granada CN, Lezcano C, Scolyer RA, et al. Lethal melanoma in children: 3. Cabrera R, Pulgar C, Daza F, et al. Dermatoscopy of a primary dermal mela-
a clinicopathological study of 12 cases. Pathology. 2016;48:705–711. noma. Am J Dermatopathol. 2009;31:574–577.
4. Hida Y, Kubo Y, Miyajima O, et al. Primary dermal melanoma: A case report
and molecular characterization. J Dermatol. 2009;36:346–352.
Dermal squamomelanocytic tumor
5. Lee CC, Faries MB, Ye X, et al. Solitary dermal melanoma: beginning or end of
1. Rosen LB, Benson J, Williams WD, et al. A malignant neoplasm with features the metastatic process? Ann Surg Oncol. 2009;16:578–584.
of both squamous cell carcinoma and malignant melanoma. Am J Dermatopa- 6. Teow J, Chin O, Hanikeri M, et al. Primary dermal melanoma: a West Austra-
thol. 1984;6(suppl 1):213–219. lian cohort. ANZ J Surg. 2015;85:664–667.
2. Pool SE, Manleel F, Clark WH, et al. Dermal squamo-melanocytic tumor: a 7. Sidiropoulos M, Obregon R, Cooper C, et al. Primary dermal melanoma: a
unique biphenotypic neoplasm of uncertain biological potential. Hum Pathol. unique subtype of melanoma to be distinguished from cutaneous metastatic
1999;30:525–529. melanoma: a clinical, histologic, and gene expression-profiling study. J Am Acad
3. Miteva M, Herschthal D, Ricotti C, et al. A rare case of a cutaneous squa- Dermatol. 2014;71:1083–1092.
momelanocytic tumor: revisiting the histogenesis of combined neoplasms. Am J 8. Cassarino DS, Cabral ES, Kartha RV, et al. Primary dermal melanoma: distinct
Dermatopathol. 2009;31:599–603. immunohistochemical findings and clinical outcome compared with nodular
4. Rongioletti F, Baldari M, Carli C, et al. Squamomelanocytic tumor: a new case and metastatic melanoma. Arch Dermatol. 2008;144:49–56.
of a unique biphenotypic neoplasm of uncertain biological potential. J Cutan 9. Helm TN, Helm KF. More evidence for a new melanoma paradigm. Arch Der-
Pathol. 2009;36:477–481. matol. 2008;144:946–947.

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
References 1362.e17

10. Katz KA, Jonasch E, Hodi FS, et al. Melanoma of unknown primary: expe- 19. Curtin JA, Fridlyand J, Kageshita T, et al. Distinct sets of genetic alterations in
rience at Massachusetts General Hospital and Dana-Farber Cancer Institute. melanoma. N Engl J Med. 2005;353:2135–2147.
Melanoma Res. 2005;15:77–82. 20. Shain AH, Yeh I, Kovalyshyn I, et al. The genetic evolution of melanoma from
11. Schlagenhauff B, Stroebel W, Ellwanger U, et al. Metastatic melanoma of unknown precursor lesions. N Engl J Med. 2015;373:1926–1936.
primary origin shows prognostic similarities to regional metastatic melanoma: 21. Bauer J, Buttner P, Wiecker TS, et al. Effect of sunscreen and clothing on the
recommendations for initial staging examinations. Cancer. 1997;80:60–65. number of melanocytic nevi in 1,812 German children attending day care. Am
12. Reintgen DS, McCarty KS, Woodard B, et al. Metastatic malignant melanoma J Epidemiol. 2005;161:620–627.
with an unknown primary. Surg Gynecol Obstet. 1983;156:335–340. 22. Rivers JK. Is there more than one road to melanoma? Lancet. 2004;363:728–
13. Giuliano AE, Moseley HS, Morton DL. Clinical aspects of unknown primary 730.
melanoma. Ann Surg. 1980;191:98–104. 23. Maldonado JL, Fridlyand J, Patel H, et al. Determinants of BRAF mutations in
14. Anbari KK, Schuchter LM, Bucky LP, et al. Melanoma of unknown primary primary melanoma. J Natl Cancer Inst. 2003;95:1878–1890.
site: presentation, treatment, and prognosis–a single institution study. University 24. Viros A, Fridlyand J, Bauer J, et al. Improving melanoma classification by inte-
of Pennsylvania Pigmented Lesion Study Group. Cancer. 1997;79:1816–1821. grating genetic and morphologic features. PLoS Med. 2008;5:e120.
15. Cormier JN, Xing Y, Feng L, et al. Metastatic melanoma to lymph nodes in 25. Flaherty KT, Puzanov I, Kim KB, et al. Inhibition of mutated, activated BRAF
patients with unknown primary sites. Cancer. 2006;106:2012–2020. in metastatic melanoma. N Engl J Med. 2010;363:809–819.
16. Sun BK, Wang H2, Kim J, et al. Mutational profile of primary dermal mela- 26. Hodi FS, Friedlander P, Corless CL, et al. Major response to imatinib mesylate
noma: A case series. J Am Acad Dermatol. 2016;75:1263–1265. in KIT-mutated melanoma. J Clin Oncol. 2008;26:2046–2051.
17. Cellier L, Perron E, Pissaloux D, et al. Cutaneous melanocytoma with CRTC1- 27. Lutzky J, Bauer J, Bastian BC. Dose-dependent, complete response to imatinib
TRIM11 fusion: report of 5 cases resembling clear cell sarcoma. Am J Surg of a metastatic mucosal melanoma with a K642E KIT mutation. Pigment Cell
Pathol. 2018;42:382–391. Melanoma Res. 2008;21:492–493.
18. Hantschke M, Mentzel T, Rütten A, et al. Cutaneous clear cell sarcoma: 28. Quintas-Cardama A, Lazar AJ, Woodman SE, et al. Complete response of stage
a clinicopathologic, immunohistochemical, and molecular analysis of 12 IV anal mucosal melanoma expressing KIT Val560Asp to the multikinase inhib-
cases emphasizing its distinction from dermal melanoma. Am J Surg Pathol. itor sorabenib. Nat Clin Pract Oncol. 2008;5:737–740.
2010;34:216–222. 29. Friedman RJ, Rigel DS, Kopf AW, et al. Favorable prognosis for malig-
nant melanomas associated with acquired melanocytic nevi. Arch Dermatol.
1983;119:455–462.
Molecular classification of melanoma
30. Chiba K, Lorbeer F, Shain A. Mutations in the promoter of the telomerase
1. Bastian BC, Pinkel D. Expanding the genetic spectrum of pigmentation. Pigment gene TERT contribute to tumorigenesis by a two-step mechanism. Science.
Cell Melanoma Res. 2008;21:507–508. 2017;357:1416–1420.
2. Yakubu A, Mabogunje OA. Skin cancer in African albinos. Acta Oncol. 31. Hayward NK, Wilmott JS, Waddell N, et al. Whole-genome landscapes of
1993;32:621–622. major melanoma subtypes. Nature. 2017;545:175–180.
3. Premi S, Wallisch S, Mano CM, et al. Chemiexcitation of melanin deriv- 32. Tsao H, Goel V, Wu H, et al. Genetic interaction between NRAS and BRAF
atives induces DNA photoproducts long after UV exposure. Science. mutations and PTEN/MMAC1 inactivation in melanoma. J Invest Dermatol.
2015;347:842–847. 2004;122:337–341.
4. Noonan FP, Zaidi MR, Wolnicka-Glubisz A, et al. Melanoma induction by 33. Curtin JA, Stark MS, Pinkel D, et al. PI3-kinase subunits are infrequent somatic
ultraviolet A but not ultraviolet B radiation requires melanin pigment. Nat targets in melanoma. J Invest Dermatol. 2006;126:1660–1663.
Commun. 2012;3:884. 34. Maldonado JL, Fridlyand J, Patel H, et al. Determinants of BRAF mutations in
5. Davies H, Bignell GR, Cox C, et al. Mutations of the BRAF gene in human primary melanoma. J Natl Cancer Inst. 2003;95:1878–1890.
cancer. Nature. 2002;417:949–954. 35. Willmore-Payne C, Holden JA, Hirschowitz S, et al. BRAF and c-kit gene
6. Albino AP, Le Strange R, Oliff AI, et al. Transforming ras genes from human copy number in mutation-positive malignant melanoma. Hum Pathol.
melanoma: a manifestation of tumour heterogeneity? Nature. 1984;308: 2006;37:520–527.
69–72. 36. Sauter ER, Herlyn M, Liu SC, et al. Prolonged response to antisense cyclin D1 in
7. Wikonkal NM, Brash DE. Ultraviolet radiation induced signature mutations in a human squamous cancer xenograft model. Clin Cancer Res. 2000;6:654–660.
photocarcinogenesis. J Investig Dermatol Symp Proc. 1999;4:6–10. 37. Stadelmeyer E, Heitzer E, Resel M, et al. The BRAF V600K mutation is more
8. Pollock PM, Yu F, Qiu L, et al. Evidence for u.v. induction of CDKN2 muta- frequent than the BRAF V600E mutation in melanoma in situ of lentigo maligna
tions in melanoma cell lines. Oncogene. 1995;11:663–668. type. J Invest Dermatol. 2014;134:548–550.
9. Hocker T, Tsao H. Ultraviolet radiation and melanoma: a systematic review and 38. Saldanha G, Purnell D, Fletcher A, et al. High BRAF mutation frequency does
analysis of reported sequence variants. Hum Mutat. 2007;28:578–588. not characterize all melanocytic tumor types. Int J Cancer. 2004;111:705–710.
10. Elwood JM. Epidemiology and control of melanoma in white populations and 39. Curtin JA, Busam K, Pinkel D, et al. Somatic activation of KIT in distinct sub-
in Japan. J Invest Dermatol. 1989;92:214S–221S. types of melanoma. J Clin Oncol. 2006;24:4340–4346.
11. McGovern VJ, Cochran AJ, Van der Esch EP, et al. The classification of malig- 40. van Elsas A, Zerp SF, van der Flier S, et al. Relevance of ultraviolet-induced
nant melanoma, its histological reporting and registration: a revision of the N-ras oncogene point mutations in development of primary human cutaneous
1972 Sydney classification. Pathology. 1986;18:12–21. melanoma. Am J Pathol. 1996;149:883–893.
12. Ackerman AB, David KM. A unifying concept of malignant melanoma: biologic 41. Shain AH, Garrido M, Botton T, et al. Exome sequencing of desmoplastic mel-
aspects. Hum Pathol. 1986;17:438–440. anoma identifies recurrent NFKBIE promoter mutations and diverse activating
13. Weyers W, Euler M, Diaz-Cascajo C, et al. Classification of cutaneous malig- mutations in the MAPK pathway. Nat Genet. 2015;47:1194–1199.
nant melanoma: a reassessment of histopathologic criteria for the distinction of 42. Wiesner T, Kiuru M, Scott SN, et al. NF1 mutations are common in desmoplas-
different types. Cancer. 1999;86:288–299. tic melanoma. Am J Surg Pathol. 2015;39:1357–1362.
14. Lachiewicz AM, Berwick M, Wiggins CL, et al. Epidemiologic support for mel- 43. Fuse N, Yasumoto K, Takeda K, et al. Molecular cloning of cDNA encoding a
anoma heterogeneity using the Surveillance, Epidemiology, and End Results novel microphthalmia- associated transcription factor isoform with a distinct
Program. J Invest Dermatol. 2008;128:243–245. amino-terminus. J Biochem. 1999;126:1043–1051.
15. Whiteman DC, Watt P, Purdie DM, et al. Melanocytic nevi, solar kerato- 44. Omholt K, Platz A, Ringborg U, et al. Cytoplasmic and nuclear accumulation
ses, and divergent pathways to cutaneous melanoma. J Natl Cancer Inst. of beta-catenin is rarely caused by CTNNB1 exon 3 mutations in cutaneous
2003;95:806–812. malignant melanoma. Int J Cancer. 2001;92:839–842.
16. van der Pols JC, Xu C, Boyle GM, et al. Expression of p53 tumor suppressor 45. Demunter A, Libbrecht L, Degreef H, et al. Loss of membranous expression of
protein in sun-exposed skin and associations with sunscreen use and time spent beta-catenin is associated with tumor progression in cutaneous melanoma and
outdoors: a community-based study. Am J Epidemiol. 2006;163:982–988. rarely caused by exon 3 mutations. Mod Pathol. 2002;15:454–461.
17. Olsen CM, Zens MS, Stukel TA, et al. Nevus density and melanoma risk in 46. Aoude LG, Pritchard AL, Robles-Espinoza CD, et al. Nonsense mutations in
women: A pooled analysis to test the divergent pathway hypothesis. Int J the shelterin complex genes ACD and TERF2IP in familial melanoma. J Natl
Cancer. 2009;124:937–944. Cancer Inst. 2014;107(2).
18. Bauer J, Büttner P, Murali R, et al. BRAF mutations in cutaneous melanoma are 47. Huang F, Hodis E, Xu MJ, et al. Highly recurrent TERT promoter mutations in
independently associated with age, anatomic site of the primary tumor, and the human melanoma. Science. 2013;339:957–959.
degree of solar elastosis at the primary tumor site. Pigment Cell Melanoma Res. 48. Horn S, Figl A, Rachakonda PS, et al. TERT promoter mutations in familial
2011;24:345–351. and sporadic melanoma. Science. 2013;339:959–961.

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
1362.e18 References

49. Shain AH, Bastian BC. From melanocytes to melanomas. Nat Rev Cancer. 77. Alexis JB, Martinez AE, Lutzky J. An immunohistochemical evaluation of c-kit
2016;16:345–358. (CD-117) expression in malignant melanoma, and results of imatinib mesylate
50. Bastian BC, Kashani-Sabet M, Hamm H, et al. Gene amplifications characterize (Gleevec) therapy in three patients. Melanoma Res. 2005;15:283–285.
acral melanoma and permit the detection of occult tumor cells in the surround- 78. Alexeev V, Yoon K. Distinctive role of the cKit receptor tyrosine kinase signal-
ing skin. Cancer Res. 2000;60:1968–1973. ing in mammalian melanocytes. J Invest Dermatol. 2006;126:1102–1110.
51. Bastian BC, Leboit PE, Hamm H, et al. Chromosomal gains and losses in 79. Cruz F 3rd, Rubin BP, Wilson D, et al. Absence of BRAF and NRAS mutations
primary cutaneous melanomas detected by comparative genomic hybridization. in uveal melanoma. Cancer Res. 2003;63:5761–5766.
Cancer Res. 1998;58:2170–2175. 80. Singh AD, Bergman L, Seregard S. Uveal melanoma: epidemiologic aspects.
52. Bastian BC, Olshen A, LeBoit PE, et al. Classification of melanocytic tumors by Ophthalmol Clin North Am. 2005;18:75–84, viii.
DNA copy number changes. Am J Pathol. 2003;163:1765–1770. 81. Horsman DE, White VA. Cytogenetic analysis of uveal melanoma. Consistent
53. van Dijk M, Sprenger S, Rombout P, et al. Distinct chromosomal aberrations occurrence of monosomy 3 and trisomy 8q. Cancer. 1993;71:811–819.
in sinonasal mucosal melanoma as detected by comparative genomic hybridiza- 82. Zembowicz A, Mihm MC. Dermal dendritic melanocytic proliferations: an
tion. Genes Chromosomes Cancer. 2003;36:151–158. update. Histopathology. 2004;45:433–451.
54. Brodeur GM, Hogarty MD. Gene amplifications in human cancers: Biological 83. Van Raamsdonk CD, Fitch KR, Fuchs H, et al. Effects of G-protein mutations
and clinical significance. In: Vogelstein B, Kinzler KW, eds. The genetic basis of on skin color. Nat Genet. 2004;36:961–968.
human cancer. New York: McGraw-Hill. 84. Wettschureck N, Offermanns S. Mammalian G proteins and their cell type spe-
55. North JP, Kageshita T, Pinkel D, et al. Distribution and significance of occult cific functions. Physiol Rev. 2005;85:1159–1204.
intraepidermal tumor cells surrounding primary melanoma. J Invest Dermatol. 85. Van Raamsdonk CD, Bezrookove V, Green G, et al. Frequent somatic muta-
2008;128:2024–2030. tions of GNAQ in uveal melanoma and intradermal melanocytic proliferations.
56. Yang HM, Hsiao SJ, Schaeffer DF, et al. Identification of recurrent mutational Nature. 2009;457.
events in anorectal melanoma. Mod Pathol. 2017;30:286–296. 86. Chen X, Wu Q, Depeille P, et al. RasGRP3 mediates MAPK pathway activation
57. Iwama E, Fujimura T, Kusakari Y, et al. Multifocal BRAF(V600E)-mutated mel- in GNAQ mutant uveal melanoma. Cancer Cell. 2017;31:685–696.
anoma in situ on the foot. Case Rep Dermatol. 2015;7:322–327. 87. Harbour JW, Onken MD, Roberson ED, et al. Frequent mutation of BAP1 in
58. Vazquez Vde L, Vicente AL, Carloni A, et al. Molecular profiling, including metastasizing uveal melanomas. Science. 2010;330:1410–1413.
TERT promoter mutations, of acral lentiginous melanomas. Melanoma Res. 88. Costa S, Byrne M, Pissaloux D, et al. Melanomas associated with blue nevi or
2016;26:93–99. mimicking cellular blue nevi: clinical, pathologic, and molecular study of 11
59. Cohen Y, Rosenbaum E, Begum S, et al. Exon 15 BRAF mutations are cases displaying a high frequency of GNA11 mutations, BAP1 expression loss,
uncommon in melanomas arising in nonsun-exposed sites. Clin Cancer Res. and a predilection for the scalp. Am J Surg Pathol. 2016;40:368–377.
2004;10:3444–3447. 89. Harbour JW, Roberson ED, Anbunathan H, et al. Recurrent mutations at
60. Edwards RH, Ward MR, Wu H, et al. Absence of BRAF mutations in UV-pro- codon 625 of the splicing factor SF3B1 in uveal melanoma. Nat Genet.
tected mucosal melanomas. J Med Genet. 2004;41:270–272. 2013;45:133–135.
61. Wong CW, Fan YS, Chan TL, et al. BRAF and NRAS mutations are uncommon 90. Robertson AG, Shih J, Yau C, et al. Integrative analysis identifies four
in melanomas arising in diverse internal organs. J Clin Pathol. 2005;58:640–644. molecular and clinical subsets in uveal melanoma. Cancer Cell. 2017;32:
62. Lyu J, Wu Y, Li C, et al. Mutation scanning of BRAF, NRAS, KIT, and GNAQ/ 204–220.
GNA11 in oral mucosal melanoma: a study of 57 cases. J Oral Pathol Med. 91. Maize JC Jr, McCalmont TH, Carlson JA, et al. Genomic analysis of blue
2016;45:295–301. nevi and related dermal melanocytic proliferations. Am J Surg Pathol.
63. Öztürk Sari Ş, Yilmaz İ, Taşkin OÇ, et al. BRAF, NRAS, KIT, TERT, GNAQ/ 2005;29:1214–1220.
GNA11 mutation profile analysis of head and neck mucosal melanomas: a 92. Groussin L, Kirschner LS, Vincent-Dejean C, et al. Molecular analysis of
study of 42 cases. Pathology. 2017;49:55–61. the cyclic AMP-dependent protein kinase A (PKA) regulatory subunit 1A
64. Shim JH, Shin HT, Park J, et al. Mutational profiling of acral melanomas in (PRKAR1A) gene in patients with Carney complex and primary pigmented
Korean populations. Exp Dermatol. 2017;26:883–888. nodular adrenocortical disease (PPNAD) reveals novel mutations and clues for
65. Zebary A, Omholt K, Vassilaki I, et al. KIT, NRAS, BRAF and PTEN mutations pathophysiology: augmented PKA signaling is associated with adrenal tumori-
in a sample of Swedish patients with acral lentiginous melanoma. J Dermatol genesis in PPNAD. Am J Hum Genet. 2002;71:1433–1442.
Sci. 2013;72:284–289. 93. Cohen JN, Joseph NM, North JP, et al. Genomic analysis of pigmented epithe-
66. Diaz A, Puig-Butillé JA, Valera A, et al. TERT and AURKA gene copy number lioid melanocytomas reveals recurrent alterations in PRKAR1A, and PRKCA
gains enhance the detection of acral lentiginous melanomas by fluorescence in genes. Am J Surg Pathol. 2017;41:1333–1346.
situ hybridization. J Mol Diagn. 2014;16:198–206. 94. Bastian BC, LeBoit PE, Pinkel D. Mutations and copy number increase of
67. Beadling C, Jacobson-Dunlop E, Hodi FS, et al. KIT gene mutations and copy HRAS in Spitz nevi with distinctive histopathological features. Am J Pathol.
number in melanoma subtypes. Clin Cancer Res. 2008;14:6821–6828. 2000;157:967–972.
68. Omholt K, Grafström E, Kanter-Lewensohn L, et al. KIT pathway alter- 95. Wiesner T, He J, Yelensky R, et al. Kinase fusions are frequent in Spitz tumours
ations in mucosal melanomas of the vulva and other sites. Clin Cancer Res. and spitzoid melanomas. Nat Commun. 2014;5:3116.
2011;17:3933–3942. 96. Yeh I, Botton T, Talevich E, et al. Activating MET kinase rearrangements in
69. Hou JY, Baptiste C, Hombalegowda RB, et al. Vulvar and vaginal melanoma: melanoma and Spitz tumours. Nat Commun. 2015;6:7174.
A unique subclass of mucosal melanoma based on a comprehensive molecular 97. Yeh I, Tee MK, Botton T, et al. NTRK3 kinase fusions in Spitz tumours. J
analysis of 51 cases compared with 2253 cases of nongynecologic melanoma. Pathol. 2016;240:282–290.
Cancer. 2017;123:1333–1344. 98. Lazova R, Pornputtapong N, Halaban R, et al. Spitz nevi and Spitzoid mela-
70. Santi R, Simi L, Fucci R, et al. KIT genetic alterations in anorectal melanomas. nomas: exome sequencing and comparison with conventional melanocytic nevi
J Clin Pathol. 2015;68:130–134. and melanomas. Mod Pathol. 2017;30:640–649.
71. Becker JC, Brocker EB, Schadendorf D, et al. Imatinib in melanoma: a selective 99. Gerami P, Scolyer RA, Xu X, et al. Risk assessment for atypical spitzoid mela-
treatment option based on KIT mutation status? J Clin Oncol. 2007;25:e9. nocytic neoplasms using FISH to identify chromosomal copy number aberra-
72. Carvajal RD, Antonescu CR, Wolchok JD, et al. KIT as a therapeutic target in tions. Am J Surg Pathol. 2013;37:676–684.
metastatic melanoma. JAMA. 2011;305:2327–2334. 100. Wu G, Barnhill RL, Lee S, et al. The landscape of fusion transcripts in spitzoid
73. Hodi FS, Corless CL, Giobbie-Hurder A, et al. Imatinib for melanomas har- melanoma and biologically indeterminate spitzoid tumors by RNA sequencing.
boring mutationally activated or amplified KIT arising on mucosal, acral, and Mod Pathol. 2016;29:359–369.
chronically sun-damaged skin. J Clin Oncol. 2013;31:3182–3190. 101. Sanborn JZ, Chung J, Purdom E, et al. Phylogenetic analyses of melanoma
74. Guo J, Si L, Kong Y, et al. Phase II, open-label, single-arm trial of imatinib reveal complex patterns of metastatic dissemination. Proc Natl Acad Sci USA.
mesylate in patients with metastatic melanoma harboring c-Kit mutation or 2015;112:10995–11000.
amplification. J Clin Oncol. 2011;29:2904–2909. 102. Ding L, Kim M, Kanchi KL, et al. Clonal architectures and driver mutations in
75. Antonescu CR, Busam KJ, Francone TD, et al. L576P KIT mutation in anal metastatic melanomas. PLoS ONE. 2014;9:e111153.
melanomas correlates with KIT protein expression and is sensitive to specific 103. Chao T, Hsieh E, Hwang B. A study of the effect of humidification on tempera-
kinase inhibition. Int J Cancer. 2007;121:257–264. ture of incubators in the nursery in Taiwan. Zhonghua Yi Xue Za Zhi (Taipei).
76. Torres-Cabala CA, Wang WL, Trent J, et al. Correlation between KIT expres- 1989;44:25–30.
sion and KIT mutation in melanoma: a study of 173 cases with emphasis on the 104. Kim MY, Oskarsson T, Acharyya S, et al. Tumor self-seeding by circulating
acral-lentiginous/mucosal type. Mod Pathol. 2009;22:1446–1456. cancer cells. Cell. 2009;139:1315–1326.

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
References 1362.e19

105. Wagle N, Emery C, Berger MF, et al. Dissecting therapeutic resistance to 109. Turajlic S, Furney SJ, Lambros MB, et al. Whole genome sequencing of matched
RAF inhibition in melanoma by tumor genomic profiling. J Clin Oncol. primary and metastatic acral melanomas. Genome Res. 2011;22:196–207.
2011;29:3085–3096. 110. Ding L, Kim M, Kanchi KL, et al. Clonal architectures and driver mutations in
106. Damsky WE, Curley DP, Santhanakrishnan M, et al. β–catenin signaling metastatic melanomas. PLoS ONE. 2014;9:e111153.
controls metastasis in Braf-activated Pten-deficient melanomas. Cancer Cell. 111. Watson IR, Gershenwald JE, Chin L, et al. Genomic classification of cutaneous
2011;20:741–754. melanoma. Cell. 2015;161:1681–1696.
107. Garraway LA, Widlund HR, Rubin MA, et al. Integrative genomic analyses 112. Iglesia MD, Parker JS, Hoadley KA, et al. Genomic analysis of Immune cell
identify MITF as a lineage survival oncogene amplified in malignant melanoma. infiltrates across 11 tumor types. J Natl Cancer Inst. 2016;108(11).
Nature. 2005;436:117–122.
108. Gartner JJ, Davis S, Wei X, et al. Comparative exome sequencing of metastatic
lesions provides insights into the mutational progression of melanoma. BMC
Genomics. 2012;13:505.

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en octubre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.

You might also like