You are on page 1of 12

Pigmented Lesions in

C h i l d re n
Update on Clinical, Histopathologic and
Ancillary Testing
Diana Bartenstein Reusch, MDa, Elena B. Hawryluk, MD, PhDb,c,*

KEYWORDS
 Congenital melanocytic nevus  Atypical spitz tumor  Pediatric melanoma

KEY POINTS
 Congenital melanocytic nevi confer a risk of melanoma that is influenced by size and the number of
nevi.
 Early screening of appropriate patients can provide insight regarding melanoma risk.
 Spitzoid proliferations in the pediatric population generally follow a banal clinical course; molecular
studies have not yet been validated in this age group.
 Melanoma can present diversely in the pediatric population compared with adults.

INTRODUCTION for. Tardive CMN, which are sometimes referred


to as congenital nevus-like nevi, present within
Patients are commonly referred to pediatric the first few years of life and may be clinically, der-
dermatology for evaluation of pigmented lesions. moscopically, and histologically indistinguishable
For families, pediatricians, and dermatologists from earlier onset CMN.2,3 Research is underway
alike, malignancy is the main fear. An evolving to elucidate the origin of CMN cells, because
body of literature is available to inform diagnosis many hypotheses for development exist4,5
and management. In this article, we provide an up- A careful clinical inspection of CMN can inform
date on the clinical, histopathologic, and ancillary expectations regarding a child’s risk of developing
testing for 3 categories of particularly challenging malignant melanoma and central nervous system
pigmented lesions: congenital melanocytic nevi involvement, which includes neurocutaneous
(CMN), spitzoid neoplasms, and pediatric melanosis or neurocutaneous melanocytosis, as
melanoma. well as other pathologies. The term “congenital
melanocytic nevus syndrome” has been proposed
CONGENITAL MELANOCYTIC NEVI for children with CMN in addition to extracutane-
ous features. Mosaic postzygotic mutation of
CMN are pigmented proliferations that may be NRAS underlies many CMN and associated cen-
observed either at birth or within the first few tral nervous system changes.6 It has been noted
weeks of life. The estimated incidence varies, but that some children with larger and multiple CMN
is generally reported at less than 6%.1 Among all possess certain facial characteristics, such as a
CMN, smaller birthmarks are much more common prominent forehead and long philtrum, and it has
than larger lesions (Fig. 1). The overall incidence been proposed that these features also be consid-
increases when tardive CMN are also accounted ered syndromic.7
derm.theclinics.com

a
Harvard Combined Dermatology Residency Training Program, 50 Staniford Street, Suite 200, Boston, MA
02114, USA; b Department of Dermatology, Massachusetts General Hospital, Boston, MA 02114, USA;
c
Dermatology Section, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
* Corresponding author.
E-mail address: ehawryluk@mgh.harvard.edu

Dermatol Clin - (2022) -–-


https://doi.org/10.1016/j.det.2021.09.003
0733-8635/21/Ó 2021 Elsevier Inc. All rights reserved.
Descargado para Emeric Jorge Macedo Vendezu (emeric_macedo@usmp.pe) en San Martin de Porres University de ClinicalKey.es por Elsevier en noviembre 10,
2021. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
2 Bartenstein Reusch & Hawryluk

to 15% risk of melanoma.11,12 Patients with small


and medium sized CMN may have a mildly
increased risk of developing melanoma, but previ-
ous calculations have likely been skewed by the
largest of lesions in heterogenous cohorts.
For benign appearing, clinically stable CMN of
all sizes, we recommend conservative monitoring.
All patients should be taught to inspect and
palpate their birthmarks routinely. For patients
with a CMN greater than 20 cm (Fig. 2), dermatol-
ogists can additionally monitor patients using pho-
tographs and dermoscopy.
A biopsy for histopathologic analysis may be
considered if evolution concerning for melanoma
occurs. Warning signs include ulceration, pain,
rapid growth, and other unexpected evolution.13
However, whereas it was historically recommen-
ded to excise each and every CMN, this practice
is no longer the standard of care.14 Malignant
degeneration is quite uncommon in small and me-
dium CMN and excision does not seem to
decrease the risk of melanoma for patients with
large or giant CMNs. Excision, which may cause
substantial morbidity and suboptimal cosmetic re-
Fig. 1. Medium sized congenital melanocytic nevus of sults, does not prevent melanoma from occurring
the chin on a child. in other cutaneous sites, underneath skin grafts,
or internally.11 It remains to be determined whether
When evaluating a patient with CMN, it is impor- providers have an easier time detecting melanoma
tant to assess for size, satellite or multiple nevi, that develops in preintervention versus postinter-
and evolution. Imaging, histopathology, and ge- vention skin, but anecdotally, it can be challenging
netic testing can also be helpful in selected cir- to evaluate for malignant transformation in pa-
cumstances, as detailed elsewhere in this article. tients who have undergone serial excisions, derm-
abrasion, and skin grafts. Families should be
Clinical Assessment counseled extensively about the risks and benefits
of any invasive intervention before pursuing
Size treatment.
The classification of CMN is based on measure-
ment by projected adult size,8 with groupings as Satellite and multiple nevi
follows: Many patients with large and giant CMN have
 Small CMN: less than 1.5 cm small CMN surrounding their largest lesion.
 Medium CMN: 1.5–20.0 cm Customary terminology designates these smaller
 Large CMN: greater than 20–40 cm CMN as “satellite nevi,” and the presence or
 Giant CMN: greater than 40 cm absence of satellite nevi has been evaluated as a
prognostic factor for the development of CMN
Measuring the CMN size is important for risk complications. However, it has alternatively been
stratification. As CMN size increases, so too proposed that classification of CMN as single
does melanoma risk. In a comprehensive review versus multiple (>1 CMN regardless of size and
of the literature from 1966 to 2011, it was found configuration) may be more appropriate.15 The
that the incidence of melanoma in patients with a presence of multiple CMNs is a risk factor for the
CMN greater than 20 cm (large and giant CMNs) development of melanoma, central nervous sys-
was approximately 2%.9 In a recent prospective tem involvement, and adverse outcomes, regard-
cohort, 8% of patients with a CMN greater than less of configuration.9
60 cm developed melanoma, whereas melanoma
incidence remained at 1% for all other patients.10 Location
Notably, there is variability between reported re- Historically, CMN in a posterior axial distribution
sults, and patients with giant CMN and many sat- were thought to be associated with neurocutane-
ellites remain at greatest risk with upwards of 10% ous melanosis and adverse outcomes.16 It is now

Descargado para Emeric Jorge Macedo Vendezu (emeric_macedo@usmp.pe) en San Martin de Porres University de ClinicalKey.es por Elsevier en noviembre 10,
2021. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Pigmented Lesions in Children 3

1. Complete a thorough clinical examination that


is documented with high-quality photography.
2. Reassess the patient in 4 weeks.
a. If the CMN is unchanged, continue regular
monitoring until the provider is assured of le-
sional stability.
b. If the lesion has changed or if any red flag
signs develop, perform an excisional biopsy
for further characterization.

Ancillary Testing
MRI
MRI is the preferred modality to screen for central
nervous system involvement in patients with CMN.
MRI screening of the central nervous system (brain
and spine) should be considered for the following
patient groups:

1. Infants with 2 or more CMN (of any size)19


2. Infants with large or giant CMN (>20 cm)
3. Any patient who demonstrates neurologic
change (in the context of any sized CMN)
Patients with multiple as well as large or giant
Fig. 2. Large congenital melanocytic nevus of the CMN are at an increased risk of central nervous
torso on a child. system abnormality, whether owing to melano-
cytic proliferation or other process; risk increases
as size and number increases.19 Alternative guide-
questioned, however, whether an axial location for lines proposed by Krengel and Marghoob recom-
large and giant CMN simply represents the most mend central nervous system imaging for the
common site of involvement without indepen- following patient groups only20:
dently portending a poor prognosis.12
1. Patients with giant CMN greater than 40 cm
2. Patients with multiple medium CMN
Evolution 3. Patients with multiple satellite nevi
During early infancy, a CMN may grow rapidly.17 In 4. Any patient with a concerning neurologic
childhood, it is expected that a CMN will grow pro- change
portionally as a child grows. It is also common for
CMN to develop changes in thickness, texture, An MRI of the full central nervous system is
hair growth, and color over time. Although some ideally performed for high-risk patients within the
CMN darken, others substantially lighten without first 6 months of life, before myelination occurs,
treatment. and an when MRI can be performed without gen-
Benign proliferative nodules and neuroid over- eral anesthesia using the “swaddle and bottle”
growth may be present at birth in large and giant approach, to serve as a baseline. After initial imag-
CMN, or develop later in childhood. There are var- ing, or if imaging is not pursued, routine clinical
iable clinical presentations for these nonmalignant monitoring for neurologic changes should be per-
growths and reassuring features include a soft or formed by the patient’s pediatrician and dermatol-
slightly firm texture, multiple nodules, a lack of ul- ogist. Abnormal neurologic signs or symptoms
ceration, and stability after an initial period of should prompt repeat MRI and referral to
growth.10,18 neurology.
Of course, any change in a CMN must be scru- In 1 prospective study, on baseline imaging,
tinized so as to not miss malignant degeneration. If high-risk patients with CMN were found to have
there are no red flag signs warranting excision at intraparenchymal melanosis as well as other cen-
the initial evaluation (eg, rock hard texture, ulcera- tral nervous system pathology, including but not
tion, or associated lymphadenopathy), providers limited to cysts, tumors, and white matter
may follow Kinsler and colleagues’10 algorithm changes.19 Whereas the MRI finding of intraparen-
for the management of new bumps noticed within chymal melanosis was associated with seizures
a patient’s CMN: and neurodevelopmental abnormalities, it seemed

Descargado para Emeric Jorge Macedo Vendezu (emeric_macedo@usmp.pe) en San Martin de Porres University de ClinicalKey.es por Elsevier en noviembre 10,
2021. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
4 Bartenstein Reusch & Hawryluk

to carry a low risk of central nervous system mela- patients with CMN who suffered from fatal mela-
noma; this finding is in contrast with the increased noma, and BRAF hotspot genotyping can also be
risk of melanoma observed in association with used.22
other central nervous system abnormalities identi-
fied on baseline imaging.19 As data emerge, our CLINICS CARE POINTS
understanding of neurocutaneous melanosis con-
tinues to evolve. Parents must be counseled that  Evaluating patients with CMN by the size and
individual management and outcomes are unique number of CMN is important for risk stratifica-
for each patient. Central nervous system findings tion. CMN greater than 60 cm and multiple
on a baseline scan are neither fully predictive nor CMN confer an unambiguously increased
prognostic; rather, they provide clinicians with risk for melanoma and central nervous system
additional information that will inform overall sur- disease.
veillance and management.  For CMN of all sizes, clinical examination re-
mains the gold standard for melanoma sur-
Histopathology veillance. Surgical excision is not
CMN are diagnosed clinically and sampling for his- recommended for primary malignancy
topathology is typically reserved for evaluation of prevention.
lumps and bumps that grow within a CMN, for  Baseline imaging of the central nervous sys-
example, to distinguish benign proliferative nodule tem should be obtained for high-risk patients.
from melanoma. Differentiating these diagnoses
can be challenging for dermatopathologists SPITZ NEVI AND SPITZOID NEOPLASMS
because it is not uncommon to see mitoses, aty-
Spitz nevi are epithelioid and spindled cell prolifer-
pia, and other traditionally concerning features in
ations that were first identified and named (“benign
benign CMN growths.18 Further research is under-
juvenile melanoma”) by Dr Sophie Spitz in 1948.23
way, but it has been proposed that ulceration,
Since then, a spectrum from benign to malignant
high-grade nuclear atypia, and high mitotic counts
has been recognized and lesions are now classi-
may be particularly concerning for melanoma
fied on the spitzoid neoplasm spectrum as benign
when arising in an expansile nodule of epithelioid
Spitz nevi, atypical Spitz tumors, or spitzoid mela-
cells, as opposed to a small round blue cell or
nomas (Fig. 3). Spitz melanoma is a subtype of
complex pattern.18 For these challenging lesions,
spitzoid melanoma that is distinguished by the
it is important to have an experienced dermatopa-
presence of typical kinase fusions or HRAS muta-
thologist reviewing the patient’s slides. Genetic
tions, despite overlapping histopathologic
testing can also provide key information.
features.24
Recommendations for the evaluation and man-
Genetic testing
agement of adults with spitzoid tumors vary
Most CMN result from postzygotic NRAS and
greatly from those for pediatric patients. Clinically
BRAF gene mutations,21 and mosaic NRAS
banal appearing spitzoid lesions in adults carry
expression is responsible for the majority of pa-
significant risk, in contrast with pediatric lesions.25
tients with multiple CMN, large CMN, and/or neu-
This article focuses on the care of children and ad-
rocutaneous melanosis.6,21 Our current
olescents only.
understanding is that additional mutations are
required for malignant transformation. Therefore,
Identification
for patients with CMN, molecular diagnostics can
help to inform evaluation of benign proliferative Benign, nonpigmented Spitz nevi present as pink
nodule versus malignant melanoma. Fluorescence to red papules that are symmetric, may be dome
in situ hybridization (FISH) and comparative shaped, and are asymptomatic. They are typically
genomic hybridization can demonstrate copy less than 6 mm in size. At initial onset, they may
number gains and losses, which may occur in undergo a period of rapid growth, but then achieve
both benign and malignant lesions, but are typi- stability. Many lesions ultimately undergo involu-
cally thought to be more concerning if partial as tion.26 Differential diagnosis includes vascular
opposed to full chromosomes are affected.18 Hot- lesion, xanthogranuloma, and molluscum,
spot genotyping is another adjunctive tool that although close clinical inspection can usually
may be used to evaluate for melanoma, because distinguish these entities.
multiple foci of melanoma within a patient with Spitz nevi can also be pigmented. These lesions
CMN patient may demonstrate the same mutation are brown to black and can be quite striking in fair-
and provide distinction from proliferative nodules. skinned patients. However, dermoscopic exami-
NRAS hotspot mutations have been identified in nation will reveal the classic starburst pattern,

Descargado para Emeric Jorge Macedo Vendezu (emeric_macedo@usmp.pe) en San Martin de Porres University de ClinicalKey.es por Elsevier en noviembre 10,
2021. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Pigmented Lesions in Children 5

Fig. 3. Clinical appearance of pediatric typical Spitz nevus, atypical Spitz tumor, and spitzoid melanoma.27 (From
Bartenstein, D., Fisher, J., Stamoulis, C., Weldon, C., Huang, J., Gellis, S., Liang, M., Schmidt, B. and Hawryluk, E.
(2019), Clinical features and outcomes of spitzoid proliferations in children and adolescents. Br J Dermatol, 181:
366–372.)

which is pathognomonic. Features suggestive of at 1- to 3-month intervals. If the lesion achieves


atypia or melanoma include persistent evolution, stability, monitoring can be spaced to a less
ulceration, bleeding, pain, and irritation of the sur- frequent interval such as every 6 to 12 months.33
rounding skin. If the lesion develops any concerning features
such as ulceration, bleeding, asymmetric growth,
Natural History and Clinical Outcomes pain, crust or persistent growth/change, a biopsy
should be pursued. Asymmetric or unexpected
Traditionally, it has been thought that Spitz nevi do
growth in a 1- to 3-month interval, or growth out
not carry malignant potential, atypical spitzoid tu-
of proportion with the patient’s growth, should
mors have uncertain risk, and malignant spitzoid
prompt a biopsy for histopathologic confirmation.
melanoma can lead to a fatal outcome.27 Howev-
er, spitzoid tumors of all subtypes have yielded Histopathology
excellent outcomes in children and adolescents Histopathology remains the gold standard for
specifically. This factor is a key difference from differentiating spitzoid tumor subtypes, although
the adult population. their evaluation can be exceedingly challenging.
In a survey study of pediatric dermatologists Spitzoid proliferations that are biopsied should un-
who had evaluated approximately 10,000 Spitz dergo evaluation by a dermatopathologist who is
nevi and atypical Spitz tumors in their practices, an expert in pediatric pigmented lesions before
no deaths were observed.28 To date, the 2 largest any treatment decisions are made to avoid over-
retrospective cohorts for pediatric spitzoid neo- diagnosis and undue treatment morbidity.
plasms come from Boston Children’s Hospital
and Children’s Hospital of Wisconsin.29,30 Cumu- Molecular testing
latively, these cohorts capture 888 spitzoid le- Molecular testing has gained traction for diagnosis
sions, without any known deaths occurring. of adult melanomas. Exciting research is under-
There were 3 patients diagnosed with spitzoid way to determine the best use of molecular tools
melanomas at Boston Children’s Hospital, the to characterize Spitz spectrum tumors and in
youngest of whom was 14.7 years. At Children’s some cases, attempt to differentiate benign from
Hospital of Wisconsin, 1 spitzoid melanoma was malignant. For several approaches described
diagnosed in a 10-year-old. elsewhere in this article, testing is reported in adult
Although fatal outcome from a spitzoid tumor is or mixed cohorts and has not been validated in
exceedingly uncommon in pediatric patients, it children34–36; therefore, any results obtained
has been reported,31,32 and there is not sufficient must be interpreted with caution.
evidence at present to forego standard melanoma The demonstration of kinase fusion is useful to
treatment in those diagnosed with spitzoid mela- distinguish a Spitz spectrum tumor from non-
noma. It is impossible to say whether favorable Spitz pigmented lesion.
outcomes observed have been confounded by
 Kinase fusions
treatments received or publication bias.
 The characterizing molecular feature of a
Spitz tumor is kinase fusion, such as
Clinical Approach and Workup
ROS1, NTRK1, ALK, BRAF, and RET.37
For healthy patients who present with banal,  Although these genomic changes can help
typical appearing Spitz nevi, it is reasonable to to distinguish Spitz tumors from mimickers,
clinically monitor the lesion with repeat evaluation kinase fusions do not differentiate benign

Descargado para Emeric Jorge Macedo Vendezu (emeric_macedo@usmp.pe) en San Martin de Porres University de ClinicalKey.es por Elsevier en noviembre 10,
2021. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
6 Bartenstein Reusch & Hawryluk

from malignant spitzoid tumors owing to that this ancillary test was not capable of
substantial overlap. In 1 study, kinase fu- distinguishing spitzoid subtypes. A 9p21
sions were found in 55% of Spitz nevi, FISH was performed on 37 atypical Spitz tu-
56% of atypical Spitz tumors, and 39% of mor cases, demonstrating homozygous
spitzoid melanomas.37 deletion in 2 cases and heterozygous dele-
tion in 3 cases. The only fatal case in this
For patients with atypical Spitz tumors or spit- cohort occurred in a patient with an atypical
zoid melanomas, molecular testing may provide Spitz tumor who had heterozygous 9p21
investigational and supportive information to help loss, indicating that homozygous 9p21
guide management decisions; existing data are deletion was neither fully sensitive nor spe-
summarized here: cific to predict spitzoid behavior in this pe-
 Telomerase reverse transcriptase (TERT) pro- diatric cohort.
moter (Tert-p) mutations  A larger, prospective pediatric study of spit-
 Promoter mutations in the TERT gene are zoid proliferations was published in 2017
common in conventional adult melanomas. that did find a significant association be-
 In 1 study of atypical Spitz tumors (n 5 23) tween homozygous 9p21 deletion and dis-
and spitzoid melanomas (n 5 33), which ease recurrence, though the signal was
included children and adults, all 4 patients less strong than in adult patients.34 Among
with identified TERT-p mutations died 85 patients with follow-up data, 9 had ho-
from metastatic disease.38 Two were adults mozygous 9p21 deletions including 2 pa-
and 2 were adolescents, ages 11 and 14.39 tients with disease recurrence and/or
No patients in this cohort without the TERT- distant metastasis.34 Although this study
p mutation experienced death. suggests that homozygous 9p21 deletions
 Although TERT-p mutations represent a may provide complementary information
promising area of future study, there is not for management decisions, the sample
yet sufficient data to rely on this marker as size remains too low for widespread extrap-
a reliable predictor of malignancy or olation and standard use in pediatric patient
outcome in pediatric patients with spitzoid care.
neoplasms.  PReferentially expressed Antigen in MEla-
 FISH noma (PRAME)
 FISH uses specialized probes to detect  PRAME can be detected by performing
chromosomal aberrations. immunohistochemical analysis on
 In 2009, the original 4-probe FISH assay formalin-fixed and paraffin-embedded tis-
including RREB1, MYB, centromere 6, and sue blocks. It is a noninvasive adjunct that
CCND1 was reported to have 86.7% sensi- has been developed to distinguish benign
tivity and 95.4% specificity in distinguishing melanocytic lesions from malignant mela-
benign nevi from melanoma.40 However, noma.43 In a recent analysis of 35 spitzoid
this cohort was not pediatric specific. proliferations, PRAME performed poorly.36
 In 2012, an enhanced FISH assay with PRAME was expressed in 1 of 20 Spitz
higher sensitivity and specificity as well as nevi, 1 of 13 atypical Spitz tumors, and 1
better targeting of spitzoid melanoma was of 2 spitzoid melanomas, suggesting this
proposed. This assay included RREB1, tool is not helpful in the evaluation of spit-
CCND1, C-MYC, and CDKN2A and re- zoid lesions.
ported that homozygous 9p21 (CKDN2A)
deletions were significantly associated Sentinel lymph node biopsy
with aggressive behavior in spitzoid tu- Sentinel lymph node biopsy (SLNB) should be pur-
mors.41,42 This cohort was not pediatric sued only for patients diagnosed with spitzoid mel-
specific. anoma as a top-line diagnosis. However, given the
 The first pediatric-specific Spitz cohort excellent outcomes observed for preadolescent
analyzed with FISH emerged from Italy in patients diagnosed with Spitz melanoma (even in
2014. This retrospective study included 20 the presence of positive sentinel nodes), individual
Spitz nevi and 50 atypical Spitz tumors in clinicians and families must carefully weigh the
patients aged 18 years and younger.35 Re- risks and benefits of SLNB in this patient popula-
searchers found that the traditional 4-probe tion. The implications of a positive test include
FISH was positive in 20% of Spitz nevi and consideration of completion lymph node dissec-
30% of atypical Spitz tumors, indicating tion versus imaging of the lymph node basin and
consideration of adjuvant medical treatment. If

Descargado para Emeric Jorge Macedo Vendezu (emeric_macedo@usmp.pe) en San Martin de Porres University de ClinicalKey.es por Elsevier en noviembre 10,
2021. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Pigmented Lesions in Children 7

completion lymph node dissection is pursued, the PEDIATRIC MELANOMA


patient undergoes a major surgery the with risk of Epidemiology
permanent complications, such as lymphedema.
Melanoma is an exceedingly rare diagnosis in the
Adjuvant medical therapy might include chemo-
pediatric population. It is estimated to occur in
therapy agents with substantial morbidity.
less than 6 per 1 million children and adoles-
Historically, SLNB was additionally recommen-
cents.46 Its incidence increases with age and
ded for patients with atypical Spitz tumors, and
therefore for young children, the risk is even lower.
positive SLNB outcome resulted in upgrading of
Data from the Surveillance, Epidemiology and End
lesional classification to that of Spitz melanoma.
Results database suggests that between 2000 and
However, research has failed to demonstrate a
2010, less than 1 per 1 million children under the
prognostic or therapeutic benefit from this prac-
age of 4 years was affected.46 The incidence
tice.44,45 In a large cohort of children and adults,
was highest for patients 15 to 19 years old, at
survival was 99% for patients with SLNB positive
more than 17 per million, although this rate seems
atypical Spitz tumors.45 Histopathology remains
to be decreasing with time, perhaps owing to an
sufficient for tumor classification.
increased awareness about the importance of
Re-excision photoprotection.46
It is not necessary to re-excise a biopsy-proven Risk factors for pediatric melanoma include
benign Spitz nevus given the excellent clinical out- excessive sun and indoor tanning exposure, ge-
comes, but shared-decision making should be netic susceptibility, immunosuppression, prior
used with a patient’s family. Incompletely excised cancer, and the presence of giant and/or multiple
Spitz nevi can recur with atypia29 and re-excision CMN.
with the goal of complete removal may be pursued
to avoid future diagnostic confusion. Whether a Outcomes
Spitz nevus is documented as fully or incompletely
Fortunately, fatal melanoma is quite uncommon. A
excised on initial biopsy, recurrent nevus phenom-
multicenter, retrospective study characterized pe-
enon should be discussed with families given that
diatric patients who experienced fatal outcomes
margin evaluation is imperfect in standard histo-
from their melanoma.47 Queries at 16 academic in-
pathologic evaluation.
stitutions between the years of 1994 and 2017
It is important to re-excise atypical Spitz tumors
identified 38 fatal cases. Twenty-four percent
to fully evaluate lesional histopathology and to
were diagnosed before the age of 11 and 76%
avoid potential future confusion and overtreatment
were diagnosed between the ages of 11 and 20.
of a recurrent lesion.
The average survival time after diagnosis was
Patients with Spitz melanoma should undergo
35.0  29.7 months. Among the 16 cases with re-
re-excision with standard melanoma margins and
ported subtypes, nodular was the most common
additional management as dictated by tumor
(50%), followed by superficial spreading (31%),
features.
and spitzoid (19%). All 3 patients who died from
their Spitz melanomas were diagnosed postpu-
bertally (ages 13, 15, and 19 years).
CLINICS CARE POINTS Many melanoma studies have replicated the
finding that a favorable outcome is inversely corre-
 Conservative monitoring may be pursued for lated with age.29,47,48 It remains to be determined
pediatric patients presenting with banal ap- whether a distinct pathophysiology underlies adult
pearing Spitz nevi. and pediatric melanomas, and perhaps even be-
 If a biopsy is pursued, complete sampling is tween childhood and adolescent melanomas,
ideal and the specimen should be reviewed and whether other factors are at play.
by a dermatopathologist experienced in pedi-
atric pigmented lesions.
Identification
 Ancillary testing has not been validated for pe-
diatric Spitz tumors. Although it may provide Pediatric patients tend to be diagnosed with
additional information in challenging cases, it thicker melanomas than adult patients48 and de-
should be interpreted with caution and in the lays in identification may be contributory. Pediatric
context of all available data. melanoma often presents differently than adult
 Re-excision may be pursued for benign Spitz melanoma. The conventional ABCDs (Asymmetry,
nevi on an individual basis. It is recommended Border irregularity, Color variegation,
for atypical Spitz tumors and spitzoid Diameter >6 mm) apply more frequently to lesions
melanomas. in postpubertal patients, whereas these detection

Descargado para Emeric Jorge Macedo Vendezu (emeric_macedo@usmp.pe) en San Martin de Porres University de ClinicalKey.es por Elsevier en noviembre 10,
2021. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
8 Bartenstein Reusch & Hawryluk

criteria will miss a substantial subset of melanoma  Blue white veil


in prepubertal patients. For example, amelanotic  Atypical vessels
lesions are common in young children, and may  Negative network
be mistaken for a benign lesion such as a wart or
molluscum.49,50 Additionally, the dermoscopic findings of atyp-
ical vessels and shiny white structures in amela-
notic lesions may be red flags for spitzoid
Visual inspection
melanoma (Fig. 5).52 For pigmented Spitz nevi,
Pediatric-specific ABCD criteria, intended to be
the classic starburst pattern is benign (Fig. 6),
used in concert with standard melanoma detec-
but other findings may indicate malignancy
tion strategies, have been proposed as follows51:
including black, blue-gray, and dark brown colors,
 A: amelanosis a well as peripheral streaks and dark blotches.52
 B: bleeding, bumps
 C: uniform color Melanoma Subtypes
 D: variable diameter, de novo development
Pediatric melanoma subtypes differ in their demo-
A high index of suspicion is required to identify pe- graphics, characteristics, risk factors and clinical
diatric melanoma. At the end of the day, evolution is course. A brief review is included, with asterisks
likely the most sensitive predictor (Fig. 4). If a parent representing the most common subtypes.
presents to clinic with high anxiety and concern
about a child’s lesion that clinically seems to be  *Conventional melanoma
benign, close monitoring is a reasonable approach.  Older teenagers are most likely to develop
For lesions with questionable characteristics, after the same types of melanomas that develop
performing a close visual inspection, dermoscopic in adults, including melanoma in situ, super-
examination, measurement, and photography, the ficial spreading, and nodular subtypes.
patient should be brought back to clinic within 1 to These melanomas often display single
3 months, depending on the clinician’s level of nucleotide variations consistent with UV
concern. Whereas benign lesions demonstrate no damage, BRAF V600 activating mutations,
change or subtle, symmetric growth, malignant le- and TERT promoter mutations.53
sions will display prominent changes that might  Blistering sunburns and artificial indoor tan-
include asymmetric advancement of a border ning should be avoided to prevent the
edge, ulceration, or focal color change. With this development of conventional melanomas,
approach, clinicians can be confident in their clinical which can occur even before adulthood.
assessment, parents are reassured, and children  Lentigo maligna may be seen in patients
are saved from unnecessary biopsies. with xeroderma pigmentosum.54
 *CMN-associated melanoma
Dermoscopy  The most common fatal melanomas in
Dermoscopy is a noninvasive tool that can aid in young children are CMN associated.
the diagnosis of atypical appearing melanocytic  CMN-associated melanoma may present
proliferations. The following dermoscopic findings within CMN, in other cutaneous locations,
should raise concern for the diagnosis of pediatric or internally (central nervous system or
melanoma52: visceral organs). Surgical intervention for
multiple or large or giant CMN does not reli-
 Irregular globules ably decrease the melanoma risk and is not
 Atypical network recommended for malignancy prophylaxis.

Fig. 4. Evolution of a spitzoid melanoma in a 3-year-old girl before presentation to dermatology, as documented
by parent.46 (From Bartenstein DW, Song JS, Nazarian RM, Hawryluk EB. Evolving Childhood Melanoma Moni-
tored by Parental Photodocumentation. J Pediatr. 2017 Jul;186:205–205.e1.)

Descargado para Emeric Jorge Macedo Vendezu (emeric_macedo@usmp.pe) en San Martin de Porres University de ClinicalKey.es por Elsevier en noviembre 10,
2021. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Pigmented Lesions in Children 9

Fig. 5. Pattern 3, pink vascular spitzoid pattern. (A) Clinically, hypomelanotic rapid growing tumor on a 14 year
old girl. (B) Dermoscopy. Polymorphic vascular pattern, with dotted vessels, milky red areas, and the remnants of
pigment at the periphery, central shiny white structures. Spitzoid melanoma, Breslow 1.9 mm. (C) Clinically, ame-
lanotic pink bump on the lower limb of a 3-year-old girl. (D) Dermoscopy. Polymorphic vascular pattern, milky red
areas and globules and shiny white structures. Spitzoid melanoma Breslow 5.5 mm. (E) Clinically ulcerated ame-
lanotic bump on the lower limb of a 17-year-old girl. (F) Dermoscopy. Vascular pattern showing dotted vessels
and milky red globules, and ulceration. Spitzoid melanoma, Breslow 1.9 mm. (From Carrera C, Scope A, Dusza
SW, et al. Clinical and dermoscopic characterization of pediatric and adolescent melanomas: Multicenter study
of 52 cases. J Am Acad Dermatol. 2018;78(2):278–288.)

 Melanoma risk increases with CMN size pronounced architectural disorder and
and the presence of multiple lesions. cytologic abnormalities. Lentiginous mela-
 All patients with CMN should be taught the nocytic proliferation and upward migration
importance of inspecting and palpating of melanocytes are nonspecific findings in
their birthmarks over the course of their life- the pediatric population.39
time, and presenting urgently to derma-  The pretest probability of this exceptionally
tology with any observed change. rare entity should be taken into account
 *Spitz melanoma during evaluation.
 Spitz melanoma is explored in further detail  Ocular melanoma
elsewhere in this article.  Risk factors include oculodermal melano-
 Spitz melanoma may be amelanotic (pink or cytosis, choroidal nevi, neurofibromatosis
red) or pigmented. Malignant tumors often type 1, familial atypical multiple mole and
grow rapidly, ulcerate, and bleed. melanoma syndrome, and BAP1
 Ancillary genomic testing has yet to be vali- mutation.55–57
dated in the pediatric population and histo-  Ocular melanoma should be considered in
pathology remains the gold standard for the differential diagnosis for children with
diagnosis. new ocular symptoms, regardless of risk
 Fatality from Spitz melanoma has not been factors.
reported in a child less than 13 years of age.  Congenital melanoma
 Acral melanoma  Congenital melanoma occurring within the
 It can be quite challenging to histologically first year of life may occur via transplacental
distinguish benign pediatric acral nevus transmission from an affected mother,58 in
from acral melanoma. The key features of association with CMN, or de novo.59
malignant acral melanoma include

Descargado para Emeric Jorge Macedo Vendezu (emeric_macedo@usmp.pe) en San Martin de Porres University de ClinicalKey.es por Elsevier en noviembre 10,
2021. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
10 Bartenstein Reusch & Hawryluk

age, and these tumors are likely characterized by


a distinct biology. Furthermore, results are likely
confounded by cohorts with atypical spitzoid tu-
mors masquerading as spitzoid melanoma. In the
past, SLNB was performed to distinguish atypical
spitzoid tumors from spitzoid melanomas, but we
now know that SLNB does not predict patient sur-
vival for patients with atypical spitzoid tumors.
For pediatric patients presenting with nonspit-
zoid melanomas, it is prudent to recommend
SLNB. For very young patients diagnosed with a
pediatric spitzoid melanoma, that is unlikely to
result in fatality, the decision to pursue SLNB or
subsequent treatment is more complex and must
be made in careful collaboration with the patient’s
family and multidisciplinary providers.

CLINICS CARE POINTS


 Pediatric melanoma is exceedingly rare and a
Fig. 6. Pigmented Spitz nevus demonstrating symme- high level of suspicion is required for identifi-
try, lack of ulceration, and characteristic starburst cation. Evolution is likely the most sensitive in-
pattern on dermoscopy. dicator and close observation can be used for
borderline or questionable lesions without
acutely concerning features.
 Nevi in pregnant women should be evalu-  Melanoma presentations and subtypes vary
ated for atypia, malignancy, and sampled by age group.
promptly if there are any concerning  SLNB should be pursued for patients with
features.60 conventional melanomas, but the decision is
 Melanoma of unknown primary complex for prepubertal patients diagnosed
 In general, it is less likely for children to pre- with spitzoid melanoma.
sent with malignancies of unknown primary  Ancillary genomic testing has not been vali-
site than adults.61 dated for melanoma diagnosis in the pediatric
population.
Ancillary Testing
ACKNOWLEDGEMENTS
Molecular testing
Molecular tests including but not limited to FISH, Dr E.B. Hawryluk acknowledges the Dermatology
comparative genomic hybridization, and PRAME Foundation (Pediatric Dermatology Career Devel-
must be interpreted with caution if used for pediat- opment Award) for support.
ric melanocytic lesions. Despite promising ad-
vances in the adult population, studies in DISCLOSURE
pediatrics are limited, as highlighted in the section
on spitzoid tumors elsewhere in this article. Molec- D.W. Bartenstein: MRK (stock, self); DVA (stock,
ular studies have neither been validated nor spouse); ANTM (stock, spouse); PRSC (stock,
approved in the pediatric population, making re- spouse); AMAG (stock, spouse). E.B. Hawryluk:
sults challenging to interpret and use for manage- UpToDate, Inc. (royalty, author/reviewer); Purity
ment decisions. Brands, LLC (consultant); Gritstone Oncology,
Inc. (salary, stock spouse); PathAI (stock and advi-
Sentinel lymph node biopsy sory board, spouse).
The reliability of SLNB for pediatric melanoma
staging is uncertain. Several studies have demon- REFERENCES
strated that SLNB positivity is not necessarily
indicative of a poor prognosis,62 but the results 1. Ingordo V, Gentile C, Iannazzone SS, et al. Congen-
are likely confounded by cohorts with young spit- ital melanocytic nevus: an epidemiologic study in
zoid melanoma patients. As mentioned elsewhere Italy. Dermatology 2007;214(3):227–30.
in this article, a fatality has not been reported in the 2. Stinco G, Argenziano G, Favot F, et al. Absence of
context of spitzoid melanoma under 13 years of clinical and dermoscopic differences between

Descargado para Emeric Jorge Macedo Vendezu (emeric_macedo@usmp.pe) en San Martin de Porres University de ClinicalKey.es por Elsevier en noviembre 10,
2021. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Pigmented Lesions in Children 11

congenital and noncongenital melanocytic naevi in a 17. Rhodes AR, Albert LS, Weinstock MA. Congenital
cohort of 2-year-old children. Br J Dermatol 2011; nevomelanocytic nevi: proportionate area expansion
165(6):1303–7. during infancy and early childhood. J Am Acad Der-
3. Clemmensen OJ, Kroon S. The histology of "congen- matol 1996;34(1):51–62.
ital features" in early acquired melanocytic nevi. 18. Yelamos O, Arva NC, Obregon R, et al.
J Am Acad Dermatol 1988;19(4):742–6. A comparative study of proliferative nodules and le-
4. Krengel S. Nevogenesis–new thoughts regarding a thal melanomas in congenital nevi from children. Am
classical problem. Am J Dermatopathol 2005; J Surg Pathol 2015;39(3):405–15.
27(5):456–65. 19. Waelchli R, Aylett SE, Atherton D, et al. Classification
5. Kinsler VA, Anderson G, Latimer B, et al. Immuno- of neurological abnormalities in children with congen-
histochemical and ultrastructural features of ital melanocytic naevus syndrome identifies magnetic
congenital melanocytic naevus cells support a resonance imaging as the best predictor of clinical
stem-cell phenotype. Br J Dermatol 2013;169(2): outcome. Br J Dermatol 2015;173(3):739–50.
374–83. 20. Krengel S, Marghoob AA. Current management ap-
6. Kinsler VA, Thomas AC, Ishida M, et al. Multiple congen- proaches for congenital melanocytic nevi. Dermatol
ital melanocytic nevi and neurocutaneous melanosis Clin 2012;30(3):377–87.
are caused by postzygotic mutations in codon 61 of 21. Charbel C, Fontaine RH, Malouf GG, et al. NRAS
NRAS. J Invest Dermatol 2013;133(9):2229–36. mutation is the sole recurrent somatic mutation in
7. Kinsler V, Shaw AC, Merks JH, et al. The face in large congenital melanocytic nevi. J Invest Dermatol
congenital melanocytic nevus syndrome. Am J 2014;134(4):1067–74.
Med Genet A 2012;158a(5):1014–9. 22. Gerami P, Paller AS. Making a mountain out of a
8. Krengel S, Scope A, Dusza SW, et al. New recom- molehill: NRAS, mosaicism, and large congenital
mendations for the categorization of cutaneous fea- nevi. J Invest Dermatol 2013;133(9):2127–30.
tures of congenital melanocytic nevi. J Am Acad 23. Spitz S. Melanomas of childhood. Am J Pathol 1948;
Dermatol 2013;68(3):441–51. 24(3):591–609.
9. Vourc’h-Jourdain M, Martin L, Barbarot S. Large 24. Raghavan SS, Peternel S, Mully TW, et al. Spitz mel-
congenital melanocytic nevi: therapeutic manage- anoma is a distinct subset of spitzoid melanoma.
ment and melanoma risk: a systematic review. Mod Pathol 2020;33(6):1122–34.
J Am Acad Dermatol 2013;68(3):493–8.e1-4. 25. Lallas A, Apalla Z, Ioannides D, et al. International
10. Kinsler VA, O’Hare P, Bulstrode N, et al. Melanoma in Dermoscopy Society. Update on dermoscopy of
congenital melanocytic naevi. Br J Dermatol 2017; Spitz/Reed naevi and management guidelines by
176(5):1131–43. the International Dermoscopy Society. Br J Dermatol
11. Krengel S, Hauschild A, Schafer T. Melanoma risk in 2017;177(3):645–55.
congenital melanocytic naevi: a systematic review. 26. Argenziano G, Agozzino M, Bonifazi E, et al. Natural
Br J Dermatol 2006;155(1):1–8. evolution of Spitz nevi. Dermatology 2011;222(3):
12. Kinsler VA, Chong WK, Aylett SE, et al. Complica- 256–60.
tions of congenital melanocytic naevi in children: 27. Hawryluk EB, Duncan LM. The evolving nomencla-
analysis of 16 years’ experience and clinical prac- ture of spitzoid proliferations-pediatric outcomes
tice. Br J Dermatol 2008;159(4):907–14. are favorable, regardless of name. Pediatr Dermatol
13. Kim JY, Kim SM, Park KD, et al. A retrospective case 2020;37(6):1083–4.
series of 10 patients with malignant melanomas 28. Tlougan BE, Orlow SJ, Schaffer JV. Spitz nevi: be-
arising from small- and medium-sized congenital liefs, behaviors, and experiences of pediatric der-
melanocytic nevi in South Koreans. Indian J Derma- matologists. JAMA Dermatol 2013;149(3):283–91.
tol Venereol Leprol 2021;87(2):293–7. 29. Davies OMT, Majerowski J, Segura A, et al.
14. Ott H, Krengel S, Beck O, et al. Multidisciplinary A sixteen-year single-center retrospective chart re-
long-term care and modern surgical treatment of view of Spitz nevi and spitzoid neoplasms in pediat-
congenital melanocytic nevi - recommendations by ric patients. Pediatr Dermatol 2020;37(6):1073–82.
the CMN surgery network. J Dtsch Dermatol Ges 30. Bartenstein DW, Fisher JM, Stamoulis C, et al. Clin-
2019;17(10):1005–16. ical features and outcomes of spitzoid proliferations
15. Kinsler V. Satellite lesions in congenital melanocytic in children and adolescents. Br J Dermatol 2019;
nevi–time for a change of name. Pediatr Dermatol 181(2):366–72.
2011;28(2):212–3. 31. Gerami P, Scolyer RA, Xu X, et al. Risk assessment
16. DeDavid M, Orlow SJ, Provost N, et al. Neurocutane- for atypical spitzoid melanocytic neoplasms using
ous melanosis: clinical features of large congenital FISH to identify chromosomal copy number aberra-
melanocytic nevi in patients with manifest central tions. Am J Surg Pathol 2013;37(5):676–84.
nervous system melanosis. J Am Acad Dermatol 32. Massi D, Cesinaro AM, Tomasini C, et al. Atypical
1996;35(4):529–38. spitzoid melanocytic tumors: a morphological,

Descargado para Emeric Jorge Macedo Vendezu (emeric_macedo@usmp.pe) en San Martin de Porres University de ClinicalKey.es por Elsevier en noviembre 10,
2021. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
12 Bartenstein Reusch & Hawryluk

mutational, and FISH analysis. J Am Acad Dermatol 48. Averbook BJ, Lee SJ, Delman KA, et al. Pediatric
2011;64(5):919–35. melanoma: analysis of an international registry. Can-
33. Brunetti B, Nino M, Sammarco E, et al. Spitz naevus: cer 2013;119(22):4012–9.
a proposal for management. J Eur Acad Dermatol 49. Mitkov M, Chrest M, Diehl NN, et al. Pediatric mela-
Venereol 2005;19(3):391–3. nomas often mimic benign skin lesions: a retrospec-
34. Lee CY, Sholl LM, Zhang B, et al. Atypical spitzoid tive study. J Am Acad Dermatol 2016;75(4):706–11.
neoplasms in childhood: a molecular and outcome e704.
study. Am J Dermatopathol 2017;39(3):181–6. 50. Bartenstein DW, Song JS, Nazarian RM,
35. Massi D, Tomasini C, Senetta R, et al. Atypical Spitz Hawryluk EB. Evolving Childhood Melanoma Moni-
tumors in patients younger than 18 years. J Am tored by Parental Photodocumentation. J Pediatr
Acad Dermatol 2015;72(1):37–46. 2017;186:205–205.e1.
36. Raghavan SS, Wang JY, Kwok S, et al. PRAME 51. Cordoro KM, Gupta D, Frieden IJ, et al. Pediatric
expression in melanocytic proliferations with inter- melanoma: results of a large cohort study and
mediate histopathologic or spitzoid features. proposal for modified ABCD detection criteria
J Cutan Pathol 2020;47(12):1123–31. for children. J Am Acad Dermatol 2013;68(6):
37. Wiesner T, He J, Yelensky R, et al. Kinase fusions are 913–25.
frequent in Spitz tumours and spitzoid melanomas. 52. Carrera C, Scope A, Dusza SW, et al. Clinical and
Nat Commun 2014;5:3116. dermoscopic characterization of pediatric and
38. Lee S, Barnhill RL, Dummer R, et al. TERT promoter adolescent melanomas: multicenter study of 52
mutations are predictive of aggressive clinical cases. J Am Acad Dermatol 2018;78(2):278–88.
behavior in patients with spitzoid melanocytic neo- 53. Lu C, Zhang J, Nagahawatte P, et al. The genomic
plasms. Sci Rep 2015;5:11200. landscape of childhood and adolescent melanoma.
39. Bahrami A, Barnhill RL. Pathology and genomics of J Invest Dermatol 2015;135(3):816–23.
pediatric melanoma: A critical reexamination and 54. Barnhill RL, Flotte TJ, Fleischli M, et al. Cutaneous
new insights. Pediatr Blood Cancer 2018;65(2): melanoma and atypical Spitz tumors in childhood.
10.1002/pbc.26792. Cancer 1995;76(10):1833–45.
40. Gerami P, Jewell SS, Morrison LE, et al. Fluores- 55. Vavvas D, Kim I, Lane AM, et al. Posterior uveal mel-
cence in situ hybridization (FISH) as an ancillary anoma in young patients treated with proton beam
diagnostic tool in the diagnosis of melanoma. Am J therapy. Retina 2010;30(8):1267–71.
Surg Pathol 2009;33(8):1146–56. 56. Al-Jamal RT, Cassoux N, Desjardins L, et al. The pe-
41. Gammon B, Beilfuss B, Guitart J, et al. Enhanced diatric choroidal and ciliary body melanoma study: a
detection of spitzoid melanomas using fluorescence survey by the european ophthalmic oncology group.
in situ hybridization with 9p21 as an adjunctive Ophthalmology 2016;123(4):898–907.
probe. Am J Surg Pathol 2012;36(1):81–8. 57. Soni S, Lee DS, DiVito J Jr, et al. Treatment of pedi-
42. Gerami P, Li G, Pouryazdanparast P, et al. A highly atric ocular melanoma with high-dose interleukin-2
specific and discriminatory FISH assay for distin- and thalidomide. J Pediatr Hematol Oncol 2002;
guishing between benign and malignant melanocytic 24(6):488–91.
neoplasms. Am J Surg Pathol 2012;36(6):808–17. 58. Chrysouli K, Tsakanikos M, Stamataki S. A case
43. Clarke LE, Warf MB, Flake DD 2nd, et al. Clinical report of melanoma as acute mastoiditis in a 10-
validation of a gene expression signature that differ- month-old female child. Case Rep Otolaryngol
entiates benign nevi from malignant melanoma. 2019;2019:9641945.
J Cutan Pathol 2015;42(4):244–52. 59. Richardson SK, Tannous ZS, Mihm MC Jr. Congen-
44. Hung T, Piris A, Lobo A, et al. Sentinel lymph node ital and infantile melanoma: review of the literature
metastasis is not predictive of poor outcome in pa- and report of an uncommon variant, pigment-
tients with problematic spitzoid melanocytic tumors. synthesizing melanoma. J Am Acad Dermatol
Hum Pathol 2013;44(1):87–94. 2002;47(1):77–90.
45. Lallas A, Kyrgidis A, Ferrara G, et al. Atypical Spitz 60. Friedman EB, Scolyer RA, Thompson JF. Manage-
tumours and sentinel lymph node biopsy: a system- ment of pigmented skin lesions during pregnancy.
atic review. Lancet Oncol 2014;15(4):e178–83. Aust J Gen Pract 2019;48(9):621–4.
46. Campbell LB, Kreicher KL, Gittleman HR, et al. Mel- 61. Howman-Giles R, Shaw HM, Scolyer RA, et al.
anoma incidence in children and adolescents: Sentinel lymph node biopsy in pediatric and adoles-
decreasing trends in the United States. J Pediatr cent cutaneous melanoma patients. Ann Surg Oncol
2015;166(6):1505–13. 2010;17(1):138–43.
47. Hawryluk EB, Moustafa D, Bartenstein D, et al. 62. Howman-Giles R, Uren RF, Thompson J. Sentinel
A retrospective multicenter study of fatal pediatric lymph node biopsy in pediatric and adolescent pa-
melanoma. J Am Acad Dermatol 2020;83(5): tients: a proven technique. Ann Surg 2014;259(6):
1274–81. e86.

Descargado para Emeric Jorge Macedo Vendezu (emeric_macedo@usmp.pe) en San Martin de Porres University de ClinicalKey.es por Elsevier en noviembre 10,
2021. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.

You might also like