You are on page 1of 20

See Targeted Therapy available online at studentconsult.

com C H A P T E R

Skin
24 
CHAPTER OUTLINE
Acute Inflammatory Dermatoses  889 Lichen Simplex Chronicus  894 Bullous Pemphigoid  896
Urticaria  889 Infectious Dermatoses  894 Dermatitis Herpetiformis  898
Acute Eczematous Dermatitis  890 Bacterial Infections  894 Tumors of the Skin  899
Erythema Multiforme  891 Fungal Infections  894 Benign and Premalignant Epithelial Lesions  899
Chronic Inflammatory Dermatoses  892 Verrucae (Warts)  895 Malignant Epidermal Tumors  900
Psoriasis  892 Blistering (Bullous) Disorders  895 Melanocytic Proliferations  903
Lichen Planus  893 Pemphigus (Vulgaris and Foliaceus)  895

Skin diseases are common and diverse, ranging from irri- dermatopathology relies on close interactions with clini-
tating acne to life-threatening melanoma. Many are intrin- cians, particularly dermatologists, as the clinical history,
sic to the skin, but some are manifestations of diseases gross appearance, and distribution of lesions are often as
involving many tissues, such as systemic lupus erythe- important as the microscopic findings in arriving at a spe-
matosus or genetic syndromes such as neurofibromatosis. cific diagnosis. Diseases of the skin can be confusing for
In this sense, the skin is a uniquely accessible “window” the student, in part because dermatologists and derma-
through which numerous disorders can be recognized. topathologists communicate using a large, “skin-specific”
Skin is not a mere protective mantle but rather a lexicon that students must become familiar with in order
complex organ, actually the largest in the body. It is con- to understand skin diseases. The most important of these
stantly exposed to microbial and nonmicrobial antigens terms and definitions are listed in Table 24.1.
from the environment. Given this, it is not surprising that
the skin is an active participant in immune responses. Envi-
ronmentally derived antigens are processed by intraepi- ACUTE INFLAMMATORY
thelial Langerhans cells, which bear their antigenic cargo DERMATOSES
to regional lymph nodes and initiate immune responses.
Squamous cells (keratinocytes) help maintain skin homeo- Thousands of inflammatory dermatoses exist, challenging
stasis by providing a physical barrier to environmental the diagnostic acumen of even experienced clinicians. In
insults and by secreting a plethora of cytokines that influ- general, acute lesions, defined as days to several weeks in
ence both the squamous and dermal microenvironments. duration, are characterized by inflammation, edema, and
The dermis contains resident populations of CD4+ helper sometimes epidermal, vascular, or subcutaneous injury.
and CD8+ cytotoxic T lymphocytes, some of which home Acute dermatoses are often marked by infiltrates consist-
to the skin by virtue of specialized receptors such as cuta- ing of mononuclear cells rather than neutrophils, unlike
neous lymphocyte antigen, as well as memory T cells, acute inflammatory disorders at most other sites. Some
regulatory T cells (Tregs) and occasional B cells. The epi- acute lesions may persist, transitioning to a chronic phase,
dermis contains intraepithelial lymphocytes, including γ/δ while others are self-limited.
T cells, while the dermis contains perivascular mast cells
and scattered macrophages, all components of the innate Urticaria
immune system. Responses involving these immune cells
and locally released cytokines account for the morpho- Urticaria (“hives”) is a common disorder mediated by
logic patterns and clinical expressions of inflammatory and localized mast cell degranulation, which leads to dermal
infectious skin disorders. microvascular hyperpermeability. The resulting erythe-
This chapter focuses on common and pathogeni- matous, edematous, and pruritic plaques are termed wheals.
cally illustrative skin diseases. In considering these dis-
eases, it is important to appreciate that the practice of Pathogenesis
In most cases, urticaria stems from an immediate (type
The contributions of Dr. George Murphy to this chapter in previ- 1) hypersensitivity reaction (Chapter 5), in which anti-
ous editions are gratefully acknowledged. gens trigger mast cell degranulation by binding to
889
http://ebooksmedicine.net
890 C H A P T E R 24 Skin

Table 24.1  Nomenclature of Skin Lesions


dermal edema causes splaying of collagen bundles, making them
Macroscopic appear to be more widely spaced than normal. Degranulation of
Lesions Definition mast cells, which reside around superficial dermal venules, is dif-
Excoriation Traumatic lesion breaking the epidermis ficult to appreciate with routine hematoxylin-eosin (H&E) stains
and causing a raw linear area (i.e., deep but can be highlighted using a Giemsa stain.
scratch); often self-inflicted
Lichenification Thickened, rough skin (similar to a lichen
on a rock); usually the result of repeated
rubbing
Clinical Features
Macule, patch Circumscribed, flat lesion distinguished Urticaria typically affects individuals between 20 and 40
from surrounding skin by color. Macules years of age, but no age is immune. Individual lesions
are 5 mm in diameter or less, while usually develop and fade within hours, but episodes can
patches are greater than 5 mm in size. persist for days or even months. Lesions range in size and
Papule, nodule Elevated dome-shaped or flat-topped lesion. nature from small, pruritic papules to large, edematous,
Papules are 5 mm in diameter or less, erythematous plaques. They may be localized to a par-
while nodules are greater than 5 mm in ticular part of the body or generalized. In a specific type
size.
of urticaria termed pressure urticaria, lesions are found
Plaque Elevated flat-topped lesion, usually greater only in areas exposed to pressure (such as the feet or but-
than 5 mm in diameter (may be formed
by coalescence of papules)
tocks). Although not life-threatening, severe pruritus and
the social embarrassment of urticaria can compromise
Pustule Discrete, pus-filled, raised lesion
quality of life. Most cases respond to antihistamines, but
Scale Dry, horny, platelike excrescence; usually more severe, refractory disease may require treatment with
the result of imperfect cornification
leukotriene antagonists, monoclonal antibodies that block
Vesicle, bulla, blister Fluid-filled raised lesion 5 mm or less in the action of IgE, or immunosuppressive drugs.
diameter (vesicle) or greater than 5 mm
in diameter (bulla). Blister is the common
term for both lesions. Acute Eczematous Dermatitis
Wheal Itchy, transient, elevated lesion with variable
blanching and erythema formed as the Eczema is a clinical term that embraces a number of con-
result of dermal edema ditions with varied underlying etiologies. New lesions
Microscopic take the form of erythematous papules, often with overly-
Lesions Definition ing vesicles, which ooze and become crusted. Pruritus is
characteristic. With persistence, these lesions coalesce into
Acanthosis Diffuse epidermal hyperplasia
raised, scaling plaques. The nature and degree of these
Dyskeratosis Abnormal, premature keratinization within changes vary among the clinical subtypes, which include
cells below the stratum granulosum
the following:
Hyperkeratosis Thickening of the stratum corneum, often • Allergic contact dermatitis—stems from topical exposure
associated with a qualitative abnormality
of the keratin
to an allergen and is caused by delayed hypersensitivy
reactions.
Papillomatosis Surface elevation caused by hyperplasia and
enlargement of contiguous dermal
• Atopic dermatitis—formerly attributed to allergen expo-
papillae sure, now thought to often stem from defects in kerati-
Parakeratosis Retention of nuclei in the stratum corneum
nocyte barrier function, defined as skin with increased
of a squamous epithelium. On mucous permeability to substances to which it is exposed, such
membranes, parakeratosis is normal. as potential antigens
Spongiosis Intercellular edema of the epidermis • Drug-related eczematous dermatitis—hypersensitivity
reaction to a drug
• Photoeczematous dermatitis—appears as an abnormal
reaction to UV or visible light
immunoglobulin E (IgE) antibodies attached to the mast • Primary irritant dermatitis—results from exposure to
cell surface through their Fc receptor. Responsible antigens substances that chemically, physically, or mechanically
include viruses, pollens, foods, drugs, and insect venom. damage the skin
IgE-independent urticaria also can result from exposure
to substances that directly incite mast cell degranulation, While atopic dermatitis reflects a genetic predisposi-
such as opiates and certain antibiotics. In the vast majority tion and can persist for years or decades, other forms
of cases, no clinical cause is discovered even with extensive of eczematous dermatitis resolve completely when the
investigation. offending stimulus is removed or exposure is limited,
stressing the importance of investigating the underlying
cause. Only the most common form, contact dermatitis, is
MORPHOLOGY considered here.
The histologic features of urticaria often are subtle. There is
Allergic contact dermatitis is triggered by exposure
usually a sparse superficial perivenular infiltrate of mononuclear
to an environmental contact-sensitizing agent, such as
cells, rare neutrophils, and sometimes eosinophils. Superficial
poison ivy, that chemically reacts with self-proteins, cre-
ating neoantigens that can be recognized by the T cell

http://ebooksmedicine.net
Acute Inflammatory Dermatoses 891

arm of the adaptive immune system. The self-proteins


modified by the agent are processed by epidermal Lang-
erhans cells, which migrate to draining lymph nodes and
present the antigen to naïve T cells. This sensitization event
leads to acquisition of immunologic memory; on reexpo-
sure to the antigen, the activated memory CD4+ T lympho-
cytes migrate to the affected skin sites during the course
of normal circulation. There they release cytokines that
recruit additional inflammatory cells and also mediate epi-
dermal damage, as in any delayed-type hypersensitivity
reaction (Chapter 5).

MORPHOLOGY
As the name implies, skin involvement in contact dermatitis
is limited to sites of direct contact with the triggering agent
(Fig. 24.1A), whereas in other forms of eczema, lesions may be
widely distributed. Spongiosis, or epidermal edema, charac- A
terizes all forms of acute eczematous dermatitis—hence the
synonym spongiotic dermatitis. Edema fluid seeps into the
epidermis, where it splays apart keratinocytes (Fig. 24.1B). Inter-
cellular bridges are stretched and become more prominent and
are easier to visualize. This change is accompanied by a super-
ficial perivascular lymphocytic infiltrate, edema of dermal papil-
lae, and mast cell degranulation. Eosinophils may be present
and are especially prominent in spongiotic eruptions provoked
by drugs, but in general the histologic features are similar
regardless of cause, emphasizing the need for careful clinical 
correlation.

Clinical Features
Lesions of acute eczematous dermatitis are pruritic, edem-
atous, oozing plaques, often containing vesicles and bullae.
With persistent antigen exposure, lesions may become
B
scaly (hyperkeratotic) as the epidermis thickens (acan- Fig. 24.1  Eczematous dermatitis. (A) Patterned erythema and scale stem-
thosis). Some changes are produced or exacerbated by ming from a nickel-induced contact dermatitis produced by a necklace. 
scratching of the lesion (see “Lichen Simplex Chronicus”, (B) Microscopically, there is accumulation of fluid (spongiosis) between 
discussed later). The clinical causes of eczema are some- epidermal cells, which may progress to frank blister formation.
times divided into “inside jobs”—reaction to an internal
circulating antigen (such as an ingested food or drug)—
and “outside jobs”—disease resulting from contact with caused by herpes simplex, mycoplasma, and some fungi,
an external antigen (such as poison ivy). while implicated drugs include sulfonamides, penicillin,
Susceptibility to atopic dermatitis is often inherited; salicylates, hydantoins, and anti-malarials. The cytotoxic
the disorder is concordant in 80% of identical twins and T cell attack is focused on the basal cells of cutaneous
20% of fraternal twins. It usually appears in early child- and mucosal epithelia, presumably due to recogni-
hood and remits spontaneously as patients mature into tion of still unknown antigens. Certain human lympho-
adults. Children with atopic dermatitis often have asthma cyte antigen (HLA) haplotypes are associated with the
and allergic rhinitis, termed the atopic triad. Recent genetic disease.
studies have identified polymorphisms associated with
increased risk in genes that encode proteins involved in
keratinocyte barrier function, innate immunity, and T cell MORPHOLOGY
function.
Affected individuals present with a wide array of lesions, which
may include macules, papules, vesicles, and bullae (hence the
Erythema Multiforme term multiforme). Well-developed lesions have a characteristic
“targetoid” appearance (Fig. 24.2A). Early lesions show a super-
Erythema multiforme is characterized by epithelial
ficial perivascular lymphocytic infiltrate associated with dermal
injury mediated by skin-homing CD8+ cytotoxic T lym-
edema and margination of lymphocytes along the dermoepider-
phocytes. It is an uncommon, usually self-limited disorder
mal junction in intimate association with apoptotic keratinocytes
that appears to be a hypersensitivity response to certain
(see Fig. 24.2B). With time, discrete, confluent zones of basal
infections and drugs. Antecedent infections include those

http://ebooksmedicine.net
892 C H A P T E R 24 Skin

dermatoses is roughened as a result of excessive or abnor-


mal scale formation and shedding (desquamation).

Psoriasis
Psoriasis is a common chronic inflammatory dermatosis,
affecting 1% to 2% of individuals residing in the United
States. Recent epidemiologic studies have shown that pso-
riasis is associated with an increased risk for heart attack and
stroke, a relationship that may be related to a chronic inflam-
matory state. Psoriasis also is associated in up to 10% of
A patients with arthritis, which in some cases may be severe.
Pathogenesis
Psoriasis is a T cell-mediated inflammatory disease, pre-
sumed to be autoimmune in origin, although the antigens
are not well described. Both genetic (HLA types and other
susceptibility loci) and environmental factors contribute
to the risk. It is unclear whether the inciting antigens are
self-antigens, environmental antigens, or some combina-
tion of the two. Sensitized populations of T cells home to
the dermis, including CD4+ TH17 and TH1 cells and CD8+
T cells, and accumulate in the epidermis. These cells secrete
cytokines and growth factors that induce keratinocyte
hyperproliferation, resulting in the characteristic lesions.
Psoriatic lesions can be induced in susceptible individuals
by local trauma (Koebner phenomenon), which may induce
a local inflammatory response that promotes lesion devel-
opment. Genome-wide association studies (GWAS) have
B
linked an increased risk for psoriasis to polymorphisms in
Fig. 24.2  Erythema multiforme. (A) Target like lesions consisting of a pale HLA loci and genes affecting antigen presentation, TNF
central blister or zone of epidermal necrosis surrounded by macular ery- signaling, and skin barrier function. Several loci also are
thema. (B) Early lesions show lymphocytes along the dermoepidermal junc- associated with the development of psoriatic arthritis, a
tion (interface dermatitis) associated with scattered apoptotic keratinocytes, more severe complication of this disease.
marked by dark shrunken nuclei and eosinophilic cytoplasm.

MORPHOLOGY
epidermal necrosis appear, with concomitant blister formation. In
The typical lesion is a well-demarcated, pink to salmon–
a rarer and more severe form of this disease, toxic epidermal
colored plaque covered by loosely adherent silver-white
necrolysis, the necrosis extends through the full thickness of
scale (Fig. 24.3A). There is marked epidermal thickening (acan-
the epidermis.
thosis), with regular downward elongation of the rete ridges
(see Fig. 24.3B). The pattern of this downward growth has been
Clinical Features likened to “test tubes in a rack.” Increased epidermal cell turn-
over and lack of maturation results in loss of the stratum
Erythema multiforme has a broad range of severity. The
granulosum and extensive parakeratotic scale. Also seen
forms associated with infection (most often herpesvirus)
is thinning of the epidermal cell layer overlying the tips of dermal
are less severe. Erythema multiforme caused by medica-
papillae (suprapapillary plates), and dilated and tortuous blood
tions may progress to more serious eruptions, such as
vessels within the papillae. These vessels bleed readily when
Stevens-Johnson syndrome or toxic epidermal necrolysis.
the scale is removed, giving rise to multiple punctate bleeding
These forms can be life-threatening, as they may cause
points (Auspitz sign). Neutrophils form small aggregates within
sloughing of large portions of the epidermis, resulting in
both the spongiotic superficial epidermis and the parakeratotic
fluid loss and infections complications similar to those seen
stratum corneum. Similar changes can be seen in superficial
in burn-injured patients.
fungal infections, which need to be excluded with appropriate
special stains.
CHRONIC INFLAMMATORY
DERMATOSES
Clinical Features
Chronic inflammatory dermatoses are persistent skin con- Psoriasis most frequently affects the skin of the elbows,
ditions that exhibit their most characteristic features over knees, scalp, lumbosacral areas, intergluteal cleft, glans
many months to years, although they may begin with an penis, and vulva. Nail changes on the fingers and toes
acute stage. The skin surface in some chronic inflammatory occur in 30% of cases. In most cases psoriasis is limited in

http://ebooksmedicine.net
Chronic Inflammatory Dermatoses 893

A B
Fig. 24.3  Chronic psoriasis. (A) Erythematous psoriatic plaques covered by silvery-white scale. (B) Microscopic examination shows marked epidermal hyper-
plasia, downward extension of rete ridges (psoriasiform hyperplasia), and prominent parakeratotic scale with infiltrating neutrophils.

distribution, but it can be widespread and severe. The clini-


bodies. Although these changes bear some similarities to those
cal subtypes are defined by pattern of involvement and
in erythema multiforme (another type of interface dermatitis
severity. Treatment is aimed at preventing the release or
discussed earlier), lichen planus shows well-developed changes
actions of inflammatory mediators. Mild disease is treated
of chronicity, including epidermal hyperplasia, hypergranulosis,
topically with ointments containing corticosteroids or other
and hyperkeratosis.
immunomodulatory agents, whereas more severe disease
is treated with phototherapy (which has immunosuppres-
sive effects) or systemic therapy with immunosuppressive
agents such as methotrexate or TNF antagonists.

Lichen Planus
“Pruritic, purple, polygonal, planar papules, and plaques”
are the tongue-twisting Ps that describe this disorder of
skin and squamous mucosa. The lesions may result from a
CD8+ T cell–mediated cytotoxic response against antigens
in the basal cell layer and the dermoepidermal junction
that are produced by unknown mechanisms, perhaps as a
consequence of a viral infection or drug exposure.
A

MORPHOLOGY
Cutaneous lesions of lichen planus consist of pruritic, vio-
laceous, flat-topped papules that may coalesce focally to
form plaques (Fig. 24.4A). These papules are highlighted by white
dots or lines termed Wickham striae. Hyperpigmentation may
result from melanin loss into the dermis from damaged keratino-
cytes. Microscopically, lichen planus is a prototypical interface
dermatitis, so called because the inflammation and damage
are concentrated at the interface of the squamous epithelium
and papillary dermis. There is a dense, continuous infiltrate of
lymphocytes along the dermoepidermal junction (see Fig. 24.4B).
The lymphocytes are intimately associated with basal keratino-
cytes, which often atrophy or become necrotic. Perhaps as a
response to damage, the basal cells take on the appearance of B
the more mature cells of the stratum spinosum (squamatization).
This pattern of inflammation causes the dermoepidermal inter- Fig. 24.4  Lichen planus. (A) Flat-topped pink-purple polygonal papule with
face to assume an angulated, zigzag contour (sawtoothing). white lacelike markings referred to as Wickham striae. (B) Microscopic
Anucleate, necrotic basal cells are seen in the inflamed papil- examination shows a bandlike infiltrate of lymphocytes along the dermoepi-
lary dermis and are referred to as colloid bodies or Civatte dermal junction, hyperkeratosis, hypergranulosis, and pointed rete ridges
(“sawtoothing”), which results from chronic injury of the basal cell layer.

http://ebooksmedicine.net
894 C H A P T E R 24 Skin

another (often pruritic) dermatosis. It is therefore impor-


Clinical Features tant to rule out an underlying cause while recognizing that
Lichen planus is an uncommon disorder that usually the lesion may be entirely trauma-related.
presents in middle-aged adults. The cutaneous lesions
are multiple and are usually symmetrically distributed,
particularly on the extremities, and often occur around SUMMARY
the wrists and elbows and on the vulva and glans penis. INFLAMMATORY DERMATOSES
Approximately 70% of cases also involve the oral mucosa,
• Many specific inflammatory dermatoses exist and can be
where the lesions manifest as white papules with a reticu-
mediated by IgE antibodies (urticaria), antigen-specific T cells
late or netlike appearance. The cutaneous lesions of lichen
(eczema, erythema multiforme, and psoriasis), or trauma
planus usually resolve spontaneously within 1 to 2 years,
(lichen simplex chronicus).
but the oral lesions may persist and be of sufficient severity
• Underlying genetic susceptibility plays a role in atopic derma-
to interfere with food intake.
titis and psoriasis.
• These disorders can be grouped based on patterns of inflam-
Lichen Simplex Chronicus mation (e.g., interface dermatitis in lichen planus and erythema
multiforme).
Lichen simplex chronicus manifests as roughening of the
• Clinical correlation is essential to diagnose specific skin dis-
skin, which takes on an appearance reminiscent of lichen
eases, since many have overlapping, nonspecific histologic
on a tree. It is a response to local repetitive trauma, usually
features.
from rubbing or scratching. Nodular forms exist that are
referred to as prurigo nodularis. The pathogenesis of lichen
simplex chronicus is not understood, but the trauma prob-
ably induces epithelial hyperplasia and eventual dermal INFECTIOUS DERMATOSES
scarring.
Bacterial Infections
MORPHOLOGY
Numerous bacterial infections occur in skin. These range
Lichen simplex chronicus is characterized by acanthosis, from superficial infections known as impetigo, to deeper
hyperkeratosis, and hypergranulosis. Also seen are elonga- dermal abscesses associated with puncture wounds that
tion of the rete ridges, fibrosis of the papillary dermis, and a are caused by bacteria such as Pseudomonas aeruginosa. The
dermal chronic inflammatory infiltrate (Fig. 24.5). Of interest, pathogenesis is similar to that for microbial infections else-
these lesions are similar in appearance to normal volar (palms where (Chapter 9). Only impetigo is discussed here.
and soles) skin, in which skin thickening serves as an adaptation
to repetitive mechanical stress.
MORPHOLOGY
Clinical Features Impetigo is characterized by an accumulation of neutrophils
The lesions often are raised, erythematous, and scaly beneath the stratum corneum that often produces a subcorneal
and can be mistaken for keratinocytic neoplasms. Lichen pustule. Nonspecific reactive epidermal alternation and super-
simplex chronicus can be superimposed on and mask ficial dermal inflammation accompany these findings. Bacterial
cocci in the superficial epidermis can be demonstrated by Gram
stain.

Clinical Features
Impetigo, one of the most common bacterial infections of
the skin, is seen primarily in children. The causative organ-
ism is usually Staphylococcus aureus or, less commonly,
Streptococcus pyogenes, and is typically acquired through
direct contact with a source. Impetigo often begins as a
single small macule, usually on the extremities or the face
near the nose or the mouth, which rapidly evolves into a
larger lesion (Fig. 24.6), often with a honey-colored crust
of dried serum. Individuals who are colonized by S. aureus
or S. pyogenes (usually nasal or anal) are more likely to be
affected. A less common bullous form of childhood impe-
tigo may mimic an autoimmune blistering disorder.

Fungal Infections
Fig. 24.5  Lichen simplex chronicus. Note the distinctive acanthosis, hyper-
keratosis, and hypergranulosis. Superficial dermal fibrosis and vascular ectasia, Fungal infections are varied, ranging from superficial
both common features, also are present. infections with Tinea or Candida spp. to life-threatening

http://ebooksmedicine.net
Blistering (Bullous) Disorders 895

low-risk HPV subtypes that lack transforming potential.


Like high-risk HPV, low-risk viruses express viral E6 and
E7 oncoproteins that lead to dysregulated epidermal cell
growth and increased survival. Why low-risk viruses cause
warts instead of cancer is likely due to structural variation
in E6 and E7 proteins that affect their interactions with host
proteins. Because the growth of warts is normally halted
by the immune response, immunodeficiency is associated
with more numerous and larger verrucae.

MORPHOLOGY

Fig. 24.6  Impetigo. A child’s arm involved by a superficial bacterial infection Different kinds of warts are identified on the basis of their
showing the characteristic erythematous scablike lesions crusted with dried gross appearance and location and generally are caused by dis-
serum. (Courtesy of Dr. Angela Wyatt, Bellaire, Texas.) tinct HPV subtypes. Verruca vulgaris (Fig. 24.7A), the most
common type of wart, can occur anywhere but is found most
frequently on the hands, particularly on the dorsal surfaces and
Aspergillus spp. infections in immunosuppressed individu- periungual areas, where it appears as a gray-white to tan, flat to
als. Fungal infections can be superficial (stratum corneum, convex, 0.1- to 1-cm papule with a rough, pebble like surface.
hair, and nails), deep (dermis or subcutis), or systemic, the Verruca plana, or flat wart, is common on the face or dorsal
last type arising through hematogenous spread, often in an surfaces of the hands. These warts are flat, smooth, tan macules.
immunocompromised patient. Verruca plantaris and verruca palmaris occur on the soles
and palms, respectively. These rough, scaly lesions can reach 1 to
2 cm in diameter and may coalesce to form a surface that can
MORPHOLOGY be confused with ordinary calluses. Condyloma acuminatum
The histologic appearance varies depending on the organism, (venereal wart) occurs on the penis, female genitalia, urethra,
host response, and degree of superinfection. Superficial infec- and perianal areas (Chapters 18 and 19). Histologic features
tions are often associated with a neutrophilic infiltrate in the common to verrucae include epidermal hyperplasia, which
epidermis. Deep fungal infections produce greater tissue damage is often undulant in character (so-called verrucous or papil-
and often elicit a granulomatous response. Aspergillus can be lomatous epidermal hyperplasia) (Fig. 24.7B, top panel), and
angioinvasive. Periodic acid–Schiff (PAS) and Gomori methena- cytoplasmic vacuolization (koilocytosis), which preferentially
mine silver stains are helpful in identifying the fungal organisms. involves the more superficial epidermal layers, producing halos
of pallor surrounding infected nuclei. Infected cells also may
demonstrate prominent keratohyalin granules and jagged eosino-
Clinical Features philic intracytoplasmic protein aggregates as a result of impaired
Superficial infections usually produce erythematous maturation (Fig. 24.7B, bottom panel).
macules with superficial scale that can be pruritic, while
deeper infections such as those seen with Aspergillus spp.
are erythematous and often nodular and sometimes associ-
ated with local hemorrhage. Superficial fungal infections BLISTERING (BULLOUS) DISORDERS
may have an annular appearance. However, they also may
induce lesions that mimic other psoriasiform or eczema- Although vesicles and bullae (blisters) occur as secondary
tous dermatoses, so it is important to consider the possi- phenomena in several unrelated conditions (e.g., herpes-
bility of fungal infection when these conditions are in the virus infection, spongiotic dermatitis), there is a group of
differential diagnosis. disorders in which blisters are the primary and most dis-
tinctive feature. Blistering in these diseases tends to occur
Verrucae (Warts) at specific levels within the skin, a morphologic distinction
that is critical for diagnosis (Fig. 24.8).
Verrucae are proliferative lesions of squamous epithelial
cells that are caused by human papillomavirus (HPV). Pemphigus (Vulgaris and Foliaceus)
They are most common in children and adolescents, but
may be encountered in any age group. HPV infection Pemphigus is an uncommon autoimmune blistering dis-
usually stems from direct contact with an infected individ- order resulting from loss of normal intercellular attach-
ual or autoinoculation. Verrucae generally are self-limited, ments within the epidermis and the squamous mucosal
most often regressing spontaneously within 6 months to epithelium. There are three major variants:
2 years. • Pemphigus vulgaris (the most common type)
• Pemphigus foliaceus
Pathogenesis • Paraneoplastic pemphigus
While some members of the HPV family are associated with
preneoplastic and invasive cancers of the anogenital region The last entity is associated with internal malignancy
(Chapters 6 and 18), cutaneous warts are mainly caused by and is not discussed here.

http://ebooksmedicine.net
896 C H A P T E R 24 Skin

determines the location of the lesions. By direct immu-


nofluorescence study, lesional sites show a characteristic
fishnet-like pattern of intercellular IgG deposits (Fig. 24.9).
As with many other autoimmune diseases, pemphigus is
associated with particular HLA alleles.

MORPHOLOGY
Pemphigus vulgaris involves both mucosa and skin, especially
on the scalp, face, axillae, groin, trunk, and points of pressure.
The lesions are superficial flaccid vesicles and bullae that rupture
easily, leaving deep and often extensive erosions covered with a
serum crust (Fig. 24.10A). Pemphigus foliaceus, a rare, milder
A form of pemphigus, results in bullae that are mainly confined
to the skin, with only infrequent involvement of mucous mem-
branes. The blisters in this disorder are superficial, such that
more limited zones of erythema and crusting of ruptured blisters
are seen (Fig. 24.11A).
The common histologic denominator in all forms of pemphi-
gus is acantholysis, lysis of the intercellular adhesive junctions
between neighboring squamous epithelial cells that results in the
rounding up of detached cells. In pemphigus vulgaris, acantholysis
selectively involves the layer of cells immediately above the basal
cell layer, giving rise to a suprabasal acantholytic blister (Fig.
24.10B). In pemphigus foliaceus, acantholysis selectively involves
the superficial epidermis at the level of the stratum granulosum
(Fig. 24.11B). Variable superficial dermal infiltrates composed of
lymphocytes, macrophages, and eosinophils accompany all forms
of pemphigus.

Clinical Features
Pemphigus vulgaris is a rare disorder that occurs most
commonly in older adults and more often in women than
in men. Lesions are painful, particularly when ruptured,
and frequently develop secondary infections. Most affected
patients have oropharyngeal involvement at some point in
their course. The mainstay of treatment is immunosuppres-
sive therapy, sometimes for life. Medications can induce
pemphigus, more often pemphigus foliaceus than pem-
phigus vulgaris. There is also an unusual endemic form of
pemphigus foliaceus in South America (fogo selvagem) that
is putatively associated with the bite of a black fly.
B
Fig. 24.7  Verruca vulgaris. (A) Multiple warts, with characteristic rough, Bullous Pemphigoid
pebble like surfaces. (B) Microscopically, common warts contain zones of
papillary epidermal proliferation that often radiate symmetrically like the Bullous pemphigoid is another distinctive acquired blister-
points of a crown (top). Pallor or halos around nuclei, prominent keratohyalin ing disorder with an autoimmune basis.
granules, and related cytopathic changes are seen at higher magnification
(bottom). Pathogenesis
Blistering in bullous pemphigoid is triggered by the
linear deposition of autoreactive IgG antibodies and
Pathogenesis complement in the epidermal basement membrane (Fig.
Pemphigus vulgaris and pemphigus foliaceus are auto- 24.12A). Reactivity also occurs in the basement membrane
immune diseases caused by antibody-mediated (type II) attachment plaques (hemidesmosomes), where most
hypersensitivity reactions (Chapter 5). The pathogenic bullous pemphigoid antigen (most commonly type XVII
antibodies are IgG autoantibodies that bind to intercel- collagen) is located. The proteins that are recognized by the
lular desmosomal proteins (desmoglein types 1 and 3) autoantibodies have structural roles in dermoepidermal
found in the skin and mucous membranes. The antibod- adhesion. IgG autoantibodies to hemidesmosome compo-
ies disrupt the intercellular adhesive function of desmo- nents fix complement and cause tissue injury by recruit-
somes and may activate intercellular proteases as well. The ing neutrophils and eosinophils. Bullous pemphigoid and
distribution of desmoglein proteins within the epidermis pemphigus vulgaris are thus caused by similar pathogenic

http://ebooksmedicine.net
Blistering (Bullous) Disorders 897

A Subcorneal B Suprabasal C Subepidermal


Fig. 24.8  Levels of blister formation. (A) Subcorneal (as in pemphigus foliaceus). (B) Suprabasal (as in pemphigus vulgaris). (C) Subepidermal (as in bullous
pemphigoid or dermatitis herpetiformis).

mechanisms, but differ in their clinical presentation and Clinical Features


course due to variation in the location of the target antigen The lesions of bullous pemphigoid do not rupture as readily
(hemidesmosomes in bullous pemphigoid, desmosomes in as in pemphigus and, if uncomplicated by infection, heal
pemphigus). without scarring. The disease tends to follow a remitting
and relapsing course and responds to topical or systemic
immunosuppressive agents. Gestational pemphigoid (also
MORPHOLOGY known as herpes gestationis, a misnomer since there is no

Bullous pemphigoid is associated with tense subepidermal


bullae filled with clear fluid (Fig. 24.12B). The overlying epider-
mis characteristically lacks acantholysis. Early lesions show vari-
able numbers of eosinophils at the dermal-epidermal junction,
occasional neutrophils, superficial dermal edema, and associated
basal cell layer vacuolization.The vacuolated basal cell layer even-
tually gives rise to a fluid-filled blister (Fig. 24.12C). The blister
roof consists of full-thickness epidermis with intact intercellular
junctions, a key distinction from the blisters seen in pemphigus.

A B
B
Fig. 24.9  Direct immunofluorescence findings in pemphigus. (A) Pemphigus
vulgaris. Note the uniform deposition of immunoglobulin (green) along kera- Fig. 24.10  Pemphigus vulgaris. (A) An erosion on the leg arising from
tinocyte cell membranes in a characteristic “fishnet” pattern. (B) Pemphigus coalescence of a group of “unroofed” blisters. (B) Suprabasal intraepidermal
foliaceus. Immunoglobulin deposits are confined to superficial layers of the blister in which rounded, dissociated (acantholytic) keratinocytes are plenti-
epidermis. ful (inset).

http://ebooksmedicine.net
898 C H A P T E R 24 Skin

B
Fig. 24.11  Pemphigus foliaceus. (A) A typical blister, which is more superfi-
cial than those seen in pemphigus vulgaris. (B) Microscopic appearance of a
characteristic subcorneal blister.

viral etiology) is a clinically distinct subtype that appears


suddenly during the second or third trimester of preg-
nancy. It also is caused by autoantibodies against bullous
pemphigoid antigen. Gestational pemphigoid typically
resolves after childbirth, but may recur with subsequent
pregnancies.
C
Dermatitis Herpetiformis Fig. 24.12  Bullous pemphigoid. (A) Deposition of IgG antibody (detected
Dermatitis herpetiformis is an autoimmune blistering by direct immunofluorescence) in the subepidermal basement membrane
(epidermis is on the left side of the fluorescent band). (B) Gross appearance
disorder associated with gluten sensitivity that is char- of characteristic tense, fluid-filled blisters. (C) Subepidermal vesicle with an
acterized by extremely pruritic grouped vesicles and eosinophil-rich inflammatory infiltrate. (C, Courtesy of Dr. Victor G. Prieto,
papules. The disease affects predominantly males, often Houston, Texas.)
in the third and fourth decades of life. Up to 80% of cases
are associated with celiac disease; conversely, only a small
fraction of patients with celiac disease develop dermatitis
herpetiformis. Like celiac disease, dermatitis herpetiformis cally predisposed individuals develop IgA antibodies to
responds to a gluten-free diet. dietary gluten (derived from the wheat protein gliadin) as
well as IgA autoantibodies that cross-react with endomy-
Pathogenesis sium and tissue transglutaminases, including epidermal
The strong association of dermatitis herpetiformis with transglutaminase expressed by keratinocytes. By direct
celiac disease provides a clue to its pathogenesis. Geneti- immunofluorescence, the skin shows discontinuous,

http://ebooksmedicine.net
Tumors of the Skin 899

granular deposits of IgA selectively localized in the tips


of dermal papillae (Fig. 24.13A). The resultant injury and
inflammation produce a subepidermal blister.

MORPHOLOGY
The lesions of dermatitis herpetiformis are bilateral, symmetric,
and grouped and preferentially involve the extensor surfaces,
elbows, knees, upper back, and buttocks (Fig. 24.13B). Initially,
neutrophils accumulate selectively at the tips of dermal papil-
lae, forming small microabscesses (Fig. 24.13C). The basal cells
overlying these microabscesses show vacuolization and focal
dermoepidermal separation that ultimately coalesce to form a
true subepidermal blister. A

SUMMARY
BLISTERING DISORDERS
• Blistering disorders are classified based on the level of the
epidermis that is affected.
• These disorders often are caused by autoantibodies specific
for epithelial or basement membrane proteins that lead to
unmooring of keratinocytes (acantholysis).
• Pemphigus is associated with IgG autoantibodies to various
intercellular desmogleins (part of the desmosome), resulting
in bullae that are either subcorneal (pemphigus foliaceus) or
suprabasilar (pemphigus vulgaris).
• Bullous pemphigoid is associated with IgG autoantibodies to
basement membrane proteins (part of the hemidesmosome)
and produces a subepidermal blister.
• Dermatitis herpetiformis is associated with IgA autoantibodies
to transglutaminase, and also is characterized by subepidermal
blisters.
B

TUMORS OF THE SKIN


Benign and Premalignant Epithelial Lesions
Benign epithelial neoplasms are common and probably
arise from stem cells residing in the epidermis and hair
follicles. These tumors grow to a limited size and generally
do not undergo malignant transformation.

Seborrheic Keratosis
These common pigmented epidermal tumors occur most
frequently in middle-age or older individuals. They arise
spontaneously and are particularly numerous on the trunk,
although the extremities, head, and neck also may be sites
of involvement.

Fig. 24.13  Dermatitis herpetiformis. (A) Characteristic selective deposition


of IgA autoantibody at the tips of the dermal papillae. (B) Lesions consist of
intact and eroded (usually scratched) erythematous blisters, often grouped
C
(seen here on elbows and arms). (C) Blisters associated with basal cell layer
injury, initially caused by accumulation of neutrophils (microabscesses) at the
tips of the dermal papillae. (A, Courtesy of Dr. Victor G. Prieto, Houston, Texas.)
900 C H A P T E R 24 Skin

Seborrheic keratoses are caused by acquired activat- sunlight and are associated with hyperkeratosis, they are
ing mutations in growth factor signaling pathways. A called actinic (sun-related) keratoses. The rate of progression
significant fraction of these tumors harbor activating muta- to squamous cell carcinoma is small, varying from 0.1% to
tions in fibroblast growth factor receptor 3 (FGFR3), which 2.6% per year. Most regress or remain stable.
possesses a tyrosine kinase activity that stimulates RAS
and the PI3K/AKT pathway, while others have activat-
MORPHOLOGY
ing mutations in downstream pathway components such
as RAS and PI3K. Except for cosmetic concerns, seborrheic Actinic keratoses usually are less than 1 cm in diameter, tan-
keratoses are usually of little clinical importance. However, brown or red, and rough (sandpaper-like) to the touch (Fig.
in rare patients hundreds of lesions may appear sud- 24.15A). Microscopically, lower portions of the epidermis show
denly as a paraneoplastic syndrome (sign of Leser-Trelat). cytologic atypia, often associated with hyperplasia of basal
Patients with this presentation may harbor internal malig- cells (see Fig. 24.15B) or with atrophy and diffuse thinning of
nancies, most commonly gastrointestinal tract carcinomas, the epidermal surface. The dermis contains thickened, blue-gray
which produce growth factors that stimulate epidermal elastic fibers (solar elastosis), the result of chronic sun damage.
proliferation. The stratum corneum is thickened and shows abnormal reten-
tion of nuclei (parakeratosis). Uncommonly, full-thickness epi-
dermal atypia is seen; such lesions are considered squamous cell
MORPHOLOGY carcinoma in situ (Fig. 24.15C).
Seborrheic keratoses are round, exophytic, coinlike plaques
that vary in diameter from millimeters to centimeters and have Clinical Features
a “stuck-on” appearance (Fig. 24.14, inset). They are tan to Actinic keratoses are very common in fair-skinned individ-
dark brown and have a velvety- to granular-appearing surface. uals and increase in incidence with age and sun exposure.
Occasionally, their dark color is suggestive of melanoma, leading As would be expected, there is a predilection for sun-
to surgical removal. exposed areas (face, arms, dorsum of the hands). Despite
Microscopically, seborrheic keratoses are composed of the low risk for malignant progression, actinic keratoses
monotonous sheets of small cells that resemble the basal cells are often treated, either to prevent progression or for cos-
of the normal epidermis (see Fig. 24.14). Variable melanin pig- metic reasons. Local eradication with cryotherapy (super-
mentation is present within these basaloid cells, accounting for ficial freezing) or topical agents is effective and safe.
the brown coloration seen grossly. Hyperkeratosis occurs at
the surface, and the presence of small keratin-filled cysts (horn
cysts) and downgrowth of keratin into the main tumor mass SUMMARY
(pseudo–horn cysts) are characteristic features.
BENIGN AND PREMALIGNANT EPITHELIAL
LESIONS
Actinic Keratosis • Seborrheic keratosis: Round, flat plaques made up of prolif-
Actinic keratosis is a premalignant lesion caused by erating monotonous epidermal basal cells, which sometimes
UV-induced DNA damage that is associated with muta- contain melanin. Hyperkeratosis and keratin-filled cysts are
tions in TP53 and other genes that also are frequently characteristic.
mutated in squamous cell carcinoma of the skin. Because • Actinic keratosis: Present on sun-exposed skin, these lesions
such lesions usually are the result of chronic exposure to show cytologic atypia in lower parts of the epidermis and
infrequently progress to carcinoma in situ.
• Although both of these lesions are associated with oncogenic
mutations, malignant transformation is exceedingly rare in seb-
orrheic keratoses and occurs in only a small subset of actinic
keratoses.

Malignant Epidermal Tumors


Squamous Cell Carcinoma
Squamous cell carcinoma is a common tumor that typically
arises on sun-exposed sites in older adults. These tumors
have a higher incidence in men than in women.
Pathogenesis
Cutaneous squamous cell carcinoma is mainly caused
by UV light exposure, which leads to widespread DNA
Fig. 24.14  Seborrheic keratosis. A characteristic roughened, brown, waxy
damage and extremely high mutational loads (Chapter 6).
lesion that appears to be “stuck on” the skin (inset). Microscopic examination As one might imagine, patients with the rare disorder xero-
shows an orderly proliferation of uniform, basaloid keratinocytes that tend derma pigmentosum, which disrupts repair of UV-induced
to form keratin microcysts (horn cysts). DNA damage, are at an exceptionally high risk. TP53
Tumors of the Skin 901

*
A B C
Fig. 24.15  Actinic keratosis. (A) Red, rough (sandpaper-like) lesions owing to excessive scale are present on the cheek and nose. (B) Basal cell layer atypia
(dysplasia) with epithelial buds associated with marked hyperkeratosis, parakeratosis, and dermal solar elastosis (asterisk). (C) Squamous cell carcinoma in situ
lesion showing full-thickness epithelial atypia.

mutations are common, as are activating mutations in RAS


and loss-of-function mutations in Notch receptors, which
transmit signals that regulate the orderly differentiation of
normal squamous epithelia.
Immunosuppression, particularly in organ transplant
recipients, is associated with an increased incidence of
cutaneous squamous cell carcinomas that are likely to be
associated with HPV infection. Other predisposing factors
include industrial carcinogens (tars and oils), chronic non-
healing ulcers, old burn scars, ingestion of arsenicals, and
ionizing radiation.

MORPHOLOGY
Squamous cell carcinomas in situ appear as sharply defined,
red, scaling plaques; some appear to arise in association with
prior actinic keratoses. Microscopically, squamous cell carcinoma
in situ is characterized by highly atypical cells at all levels of
A
the epidermis, with nuclear crowding and disorganization. More
advanced, invasive squamous cell carcinomas are nodular, often
scaly lesions that may undergo ulceration (Fig. 24.16A). Such
tumors show variable degrees of differentiation, ranging from
tumors with cells arranged in orderly lobules that exhibit exten-
sive keratinization to neoplasms consisting of highly anaplastic
cells with foci of necrosis and only abortive, single-cell keratiniza-
tion (dyskeratosis) (Fig. 24.16B).

Clinical Features
Invasive squamous cell carcinomas of the skin often are
discovered while small and resectable. Less than 1% will
have metastasized to regional lymph nodes at diagnosis.
The likelihood of metastasis is related to the thickness of
the lesion and degree of invasion into the subcutis. Tumors
arising from actinic keratoses may be locally aggressive but
generally metastasize only after long periods of time, while B
those arising in burn scars, ulcers, and non–sun-exposed
Fig. 24.16  Invasive squamous cell carcinoma. (A) A nodular, hyperkeratotic
skin often behave more aggressively. Squamous cell carci- lesion occurring on the ear, associated with metastasis to a prominent
nomas arising at internal sites (oropharynx, lung, esopha- postauricular lymph node (arrow). (B) Tumor invades the dermal soft tissue
gus, anus) are generally invasive and aggressive, possibly as irregular projections of atypical squamous cells exhibiting acantholysis.
because (unlike in the skin) early lesions go unrecognized.
902 C H A P T E R 24 Skin

Basal Cell Carcinoma (Fig. 24.17A). Some tumors contain melanin pigment and can
have an appearance similar to melanocytic nevi or melanomas.
Basal cell carcinoma is a common slow-growing cancer
Microscopically, the tumor cells resemble the normal epidermal
that rarely metastasizes. It tends to occur at sites subject to
basal cell layer or follicular germinative elements. Because they
chronic sun exposure and in lightly pigmented individuals.
arise from either the epidermis or the follicular epithelium, they
Pathogenesis are not encountered on mucosal surfaces. Two common pat-
terns are seen: multifocal superficial growths, originating
The molecular hallmark of basal cell carcinoma is loss-of-
from the epidermis, and nodular lesions growing downward
function mutations in PTCH1, a tumor suppressor gene
into the dermis as cords and islands of variably basophilic cells
that negatively regulates Hedgehog signaling; hence,
with hyperchromatic nuclei, embedded in a fibrotic or muci-
tumors exhibit constitutive Hedgehog pathway activa-
nous stromal matrix (Fig. 24.17B). Peripheral tumor cell nuclei
tion. Excessive activation of Hedgehog in turn activates a
align in the outermost layer (a pattern termed palisading), which
host of downstream genes implicated in cell growth and
often separates from the stroma, creating a characteristic cleft 
survival and other phenotypes linked to malignant transfor-
(Fig. 24.17C).
mation. In sporadic basal cell carcinoma, PTCH1 mutations
bear the telltale signs of UV light–induced DNA damage.
The central role of increased Hedgehog signaling in basal
cell carcinoma is further emphasized by Gorlin syndrome, an Clinical Features
autosomal dominant disorder caused by inherited defects It is estimated that more than 1 million basal cell carcino-
in PTCH1 that is associated with familial basal cell carci- mas are treated in the United States annually. By far the
noma. The Hedgehog pathway is an important regulator most important risk factor is cumulative sun exposure;
of embryonic development, and patients with Gorlin syn- basal cell carcinoma is more common in warm southern
drome also often manifest subtle developmental anomalies. regions of the United States, and its incidence is 40-fold
Mutations in TP53 caused by UV light–induced damage also higher in sunny climates near the equator, such as Aus-
are common in both familial and sporadic tumors. tralia, than it is in Northern European locales. Individual
tumors usually are cured by local excision, but approx-
imately 40% of patients develop another basal cell car-
cinoma within 5 years. Advanced lesions may ulcerate,
MORPHOLOGY and extensive local invasion of bone or facial sinuses may
occur if the lesions are neglected. Metastasis is exceedingly
Basal cell carcinomas manifest as pearly papules, often with
rare. Hedgehog pathway inhibitors are used to treat locally
prominent, dilated subepidermal blood vessels (telangiectasia)
advanced or metastatic tumors.

A B C
Fig. 24.17  Basal cell carcinoma. (A) A prototypical pearly, smooth-surfaced papule with telangiectatic vessels. (B) Tumor is composed of nests of basaloid
cells infiltrating a fibrotic stroma. (C) Tumor cells with scant cytoplasm and small hyperchromatic nuclei that palisade on the outside of the nest. The cleft
between the tumor cells and the stroma is a highly characteristic artifact of sectioning.
Tumors of the Skin 903

melanomas. Indeed, the growth and migration of nevus


SUMMARY cells from the dermoepidermal junction into the underlying
MALIGNANT EPIDERMAL TUMORS dermis is accompanied by morphologic changes that are
• The incidence of both basal cell and squamous cell carcinoma taken as evidence of cellular senescence (Fig. 24.18). Super-
is strongly correlated with increasing lifetime sun exposure. ficial nevus cells are larger and tend to produce melanin
• Risk factors for cutaneous squamous cell carcinoma include pigment and grow in nests; deeper nevus cells are smaller,
fair skin, UV light exposure, exposure to carcinogenic chemi- produce little or no pigment, and grow in cords or single
cals, chronic skin inflammation and scarring, and HPV infection cells. The deepest nevus cells have fusiform contours and
(in the setting of immunosuppression). grow in fascicles. This sequence of morphologic changes
• Cutaneous squamous cell carcinoma has the potential for metas- is of diagnostic importance, since they are absent from
tasis but usually is recognized and excised before it does so. melanomas.
• Basal cell carcinoma, the most common malignancy worldwide,
is a locally aggressive tumor associated with mutations in the MORPHOLOGY
Hedgehog pathway. Metastasis is very rare.
Common melanocytic nevi are tan-to-brown, uniformly pig-
mented, small papules (5 mm or less across) with well-defined,
rounded borders (Fig. 24.19A). Early lesions are composed of
Melanocytic Proliferations round to oval cells that grow in “nests” along the dermoepi-
dermal junction. Nuclei are uniform and round, and contain
Melanocytic Nevi inconspicuous nucleoli with little or no mitotic activity. Such
Strictly speaking, the term nevus denotes any congenital early-stage lesions are called junctional nevi. Eventually, most
lesion of the skin. Melanocytic nevus, however, refers to any junctional nevi grow into the underlying dermis as nests or
benign congenital or acquired neoplasm of melanocytes. cords of cells (compound nevi), and in older lesions the epi-
dermal nests may be lost entirely, creating intradermal nevi
Pathogenesis (Fig. 24.19B).
Melanocytic nevi are benign neoplasms caused by
somatic gain-of-function mutations in BRAF or RAS.
Nevi are derived from melanocytes, pigment-producing Clinical Features
cells with dendritic projections that are normally inter- There are numerous types of melanocytic nevi, with varied
spersed among basal keratinocytes. You will recall that appearances. Although these lesions usually are of only
BRAF encodes a serine/threonine kinase that lies down- cosmetic concern, they may cause irritation or mimic mela-
stream of RAS in the extracellular regulated kinase (ERK) noma, requiring their surgical removal. Compound and
pathway. Experimental evidence suggests that unbridled intradermal nevi often are more elevated than junctional
BRAF/RAS signaling initially induces melanocytic pro- nevi.
liferation followed by senescence. How these opposing
effects are coordinated is unclear, but it is believed that Dysplastic Nevus
the “brake” on proliferation provided by induced senes- Dysplastic nevi may be sporadic or familial. The latter are
cence explains why very few nevi transform into malignant important clinically because they identify individuals who

A B C D E

Fig. 24.18  Possible steps in the development of melanocytic nevi. (A) Normal skin shows only scattered melanocytes. (B) Junctional nevus. (C) Compound
nevus. (D) Intradermal nevus. (E) Intradermal nevus with extensive cellular senescence.
904 C H A P T E R 24 Skin

A B
Fig. 24.19  Melanocytic nevus. (A) Melanocytic nevi are relatively small, symmetric, and uniformly pigmented. (B) A nevus composed of melanocytes that lose
pigmentation and become smaller and more dispersed as they extend into the dermis—all signs that speak to the benign nature of the proliferation.

have an increased risk of developing melanoma. As with


hyperplasia” (Fig. 24.20B). Cytologic atypia consisting of irreg-
conventional melanocytic nevi, activating RAS or BRAF
ular, often angulated, nuclear contours and hyperchromasia is
mutations are commonly found in dysplastic nevi and are
frequently observed (Fig. 24.20B-C). Associated alterations also
believed to have a pathogenic role.
occur in the superficial dermis. These consist of a sparse lym-
phocytic infiltrate, release of melanin pigment that is phago-
MORPHOLOGY cytosed by dermal macrophages (melanin incontinence), and
linear fibrosis surrounding epidermal nests of melanocytes.These
Dysplastic nevi are larger than most acquired nevi (often more dermal changes are elements of the host response to these 
than 5 mm across) and may number in the hundreds (Fig. lesions.
24.20A). They are flat macules to slightly raised plaques, with a
“pebbly” surface. They usually have variable pigmentation (varie-
gation) and irregular borders (Fig. 24.20A, inset). Clinical Features
Microscopically, dysplastic nevi are mostly compound nevi
Unlike ordinary nevi, dysplastic nevi have a tendency to
that exhibit both architectural and cytologic evidence of
occur on body surfaces not exposed to the sun as well as
abnormal growth. Nevus cell nests within the epidermis may
on sun-exposed sites. Familial dysplastic nevus syndrome is
be enlarged and exhibit abnormal fusion or coalescence with
strongly associated with melanoma, as the lifetime risk
adjacent nests (bridging). As part of this process, single nevus
for the development of melanoma in affected individuals
cells begin to replace the normal basal cell layer along the
is close to 100%. In sporadic cases, only individuals with
dermoepidermal junction, producing so-called “lentiginous
10 or more dysplastic nevi appear to be at an increased

A B C
Fig. 24.20  Dysplastic nevus. (A) Numerous irregular nevi on the back of a patient with dysplastic nevus syndrome. The lesions usually are greater than 5 mm
in diameter and have irregular borders and variable pigmentation (inset). (B) Compound dysplastic nevus featuring a central dermal component and an asym-
metric “shoulder” of exclusively junctional melanocytes (lentiginous hyperplasia). The former corresponds to the raised, more pigmented central zone seen
in A (inset), and the latter corresponds to the less pigmented flat peripheral rim. (C) Other important features are cytologic atypia (irregular, dark-staining
nuclei) and characteristic parallel bands of fibrosis.
Tumors of the Skin 905

risk for melanoma. Transformation of dysplastic nevus regulates the G1-S transition of the cell cycle by maintain-
to melanoma has been documented, both clinically and ing the retinoblastoma (RB) tumor suppressor protein in
histologically. However, such cases are the exception, as its active state; and p14, which augments the activity of
most melanomas appear to arise de novo and not from a the p53 tumor suppressor by preventing its degradation.
preexisting nevus. Thus, the likelihood that any particular Key phases of melanoma development are marked by
nevus, dysplastic or otherwise, will develop into mela- radial and vertical growth. The earliest recognizable phase
noma is low, and these lesions are best viewed as markers of melanoma development is proposed to consist of lateral
of melanoma risk. expansion of melanocytes along the dermoepidermal junc-
tion (lentiginous hyperplasia and lentiginous compound
Melanoma nevus; Fig. 24.21A-C). This then progresses to the phase
Melanoma is less common but much more deadly than of melanoma in situ, which is marked by radial growth
basal or squamous cell carcinoma. Today, as a result of within the epidermis, often for a prolonged period (Fig.
increased public awareness of the earliest signs of skin 24.21D). During this stage, melanoma cells do not have the
melanomas, most melanomas are cured surgically. None- capacity to invade and metastasize. With time, a vertical
theless, the incidence of these lesions has increased dra- growth phase supervenes, in which the tumor grows down-
matically over the past several decades, at least in part as a ward into the deeper dermal layers as an expansile mass
result of increasing sun exposure and/or higher detection lacking cellular maturation (Fig. 24.21E). This event often
rates resulting from vigorous surveillance. is heralded by the development of a nodule in a previously
flat lesion and correlates with the emergence of metastatic
Pathogenesis potential.
As with other cutaneous malignancies, melanoma is DNA sequencing of familial and sporadic cases, includ-
mainly caused by UV light–induced DNA damage that ing cases that appear to have arisen from benign nevi, has
leads to the stepwise acquisition of driver mutations. The provided important insights into the molecular pathogen-
incidence is highest in sun-exposed skin and in geographic esis of melanoma (Fig. 24.22). The initiating event appears
locales such as Australia, where sun exposure is high and to be an activating mutation in BRAF or (less commonly)
much of the population is fair-skinned. Intense intermit- RAS. In the vast majority of cases, this produces only a
tent exposure at an early age is particularly harmful. benign nevus unless other mutations are superimposed.
Hereditary predisposition also plays a role in an estimated Sequencing of nevi with “atypical” morphologic features
5% to 10% of cases, as already discussed under familial suggestive of melanoma as well as melanomas in the radial
dysplastic nevus syndrome. For example, germ-line muta- phase of growth (melanoma in situ) has shown that they
tions in the CDKN2A locus (located on 9p21) are found in commonly harbor mutations that activate the expression of
as many as 40% of the rare individuals who suffer from telomerase, which is proposed to serve as an antidote to
familial melanoma. This complex locus encodes two tumor senescence (the usual fate of benign nevi). With additional
suppressors: p16, a cyclin-dependent kinase inhibitor that mutations or epigenetic aberrations that lead to loss of

A B C D E
Time
Fig. 24.21  Possible steps in the development of melanoma. (A) Normal skin shows only scattered melanocytes. (B) Lentiginous melanocytic hyperplasia. (C)
Lentiginous compound nevus with abnormal architecture and cytologic features (dysplastic nevus). (D) Early or radial growth phase melanoma (large dark
cells in epidermis) arising in a nevus. (E) Melanoma in vertical growth phase with metastatic potential. Note that no melanocytic nevus precursor is identified
in most cases of melanoma. They are believed to arise de novo, perhaps all using the same pathway.
906 C H A P T E R 24 Skin

Initiating mutation
BRAF Telomerase Loss of Loss of
RAS activation p16 p53, PTEN
Melanoma, Melanoma,
Benign lesion Lesion with
Histology radial growth vertical growth Metastasis
(e.g., nevus) atypia
phase phase

Point
mutations

Copy-number
alterations

Mutation
Ultraviolet radiation
signature

Fig. 24.22  Molecular evolution of cutaneous melanoma. The most important driver mutations and the overall mutational burden (point mutations and
genomic copy-number variations) at various histologic phases of melanocytic lesion progression are indicated. Note that as the tumor metastasizes to internal
sites, UV light–induced DNA damage leading to point substitutions ceases, and copy number changes related to aneuploidy increase.

CDNK2A and its encoded tumor suppressor p16, the tumor


worse biologic behavior of melanomas (termed Breslow
shifts to the invasive vertical phase of growth. Through-
thickness). By recording and using these and other variables in
out this cutaneous phase of tumor evolution, exposure
aggregate, accurate prognostication is possible.
to UV light adds to the mutational burden and increases
Individual melanoma cells usually are considerably larger
the chances of tumor progression. Finally, with additional
than nevus cells. They have large nuclei with irregular contours,
mutations in genes such as tumor suppressors TP53 and
chromatin that is characteristically clumped at the periphery of
PTEN, the tumor acquires the capacity for metastasis. This
the nuclear membrane, and prominent “cherry red” eosinophilic
phase is marked by the appearance of aneuploidy and
nucleoli (Fig. 24.23D). Immunohistochemical stains can be helpful
genomic copy number alterations, which add to the genetic
in identifying metastatic deposits (Fig. 24.23D, inset).
heterogeneity of the evolving tumor.
By contrast, the less common melanomas that arise in
non–sun-exposed acral and mucosal sites follow different
molecular courses. The most common initiating mutation Clinical Features
in these tumors is a gain-of-function mutation in the KIT Although most of these lesions arise in the skin, they also
receptor tyrosine kinase. Similarly, melanomas arising the may occur in the oral and anogenital mucosal surfaces,
uvea of the eye also have a distinct set of driver gene the esophagus, the meninges, and the eye. The following
mutations, most notably mutually exclusive mutations that discussion applies to cutaneous melanomas.
activate the GTP-binding proteins GNAQ or GNA11. Melanoma of the skin usually is asymptomatic, although
In addition, it has long been speculated that melanomas pruritus may be an early manifestation. The most important
express neoantigens that should be subject to recognition clinical sign is a change in the color or size of a pigmented lesion.
by the immune system. It follows that for melanoma to The main clinical warning signs are as follows:
develop, tumor cells must acquire the ability to either sup- 1. Rapid enlargement of a preexisting nevus
press or evade the host immune response. The importance 2. Itching or pain in a lesion
of immune evasion has been proven by the response of 3. Development of a new pigmented lesion during adult
many advanced melanomas to immune checkpoint inhib- life
itors, agents that unleash muzzled melanoma-specific T 4. Irregularity of the borders of a pigmented lesion
cells, allowing them to attack the tumor (described later). 5. Variegation of color within a pigmented lesion

These principles are expressed in the so-called “ABCs”


MORPHOLOGY of melanoma: asymmetry, border, color, diameter, and evo-
Unlike benign nevi, melanomas often exhibit striking variations
lution (change of an existing nevus). It is vitally important
in pigmentation, including shades of black, brown, red, dark
to recognize melanomas and intervene as rapidly as pos-
blue, and gray (Fig. 24.23A). The borders are irregular and
sible. The vast majority of superficial lesions are curable
often “notched.” Microscopically, malignant cells grow as poorly
surgically, while metastatic melanoma has a very poor
formed nests or as individual cells at all levels of the epider-
prognosis.
mis (pagetoid spread) and in expansile dermal nodules; these
The probability of metastasis is predicted by measuring
constitute the radial and vertical growth phases, respectively
the depth of invasion in millimeters of the vertical growth
(Fig. 24.23B-C). Of note, superficial spreading melanomas are
phase nodule from the top of the granular cell layer of the
often associated with a brisk lymphocytic infiltrate (Fig. 24.23B),
overlying epidermis (Breslow thickness). Metastasis risk
a feature that may reflect a host response to tumor-specific
also is increased in tumors with a high mitotic rate and in
antigens. Increasing thickness strongly correlates with
those that fail to induce a local immune response. When
metastases occur, they involve not only regional lymph
Tumors of the Skin 907

A B

C D
Fig. 24.23  Melanoma. (A) Lesions tend to be larger than nevi, with irregular contours and variable pigmentation. Macular areas indicate superficial (radial)
growth, while elevated areas indicate dermal invasion (vertical growth). (B) Radial growth phase, with spread of nested and single melanoma cells within the
epidermis. (C) Vertical growth phase, with nodular aggregates of infiltrating tumor cells within the dermis. (D) Melanoma cells with hyperchromatic irregular
nuclei of varying size that have prominent nucleoli. An atypical mitotic figure is present in the center of the field). The inset shows a sentinel lymph node
containing a tiny cluster of metastatic melanoma (arrow), detected by staining for the melanocytic marker HMB-45.

nodes but also liver, lungs, brain, and virtually any other well as checkpoint inhibitors together with other targeted
site that can be seeded hematogenously. Sentinel lymph therapies such as BRAF inhibitors.
node biopsy (of the first draining node[s] of a primary
melanoma) at the time of surgery provides additional
information on biologic aggressiveness. S UMMARY
Agents that selectively inhibit mutant BRAF and KIT MELANOCYTIC LESIONS, BENIGN AND
have produced dramatic responses in patients with meta- MALIGNANT
static tumors with BRAF and KIT mutations, respectively,
• Most melanocytic nevi have activating mutations in BRAF or less
an encouraging development in a previously hopeless
often NRAS, but the vast majority never undergoes malignant
disease. More recently, immune checkpoint inhibitors
transformation.
have been shown to be effective at stabilizing metastatic
• Most sporadic dysplastic nevi are best regarded as markers
disease and in some instances causing remarkable tumor
of melanoma risk rather than premalignant lesions. They are
regression and even clinical remissions. Immune check-
characterized by architectural disorder and cytologic atypia.
point inhibitors are antibodies that interfere with the func-
• Melanoma is a highly aggressive malignancy; tumors only a few
tion of proteins found on the surface of T lymphocytes
millimeters in thickness can give rise to deadly metastases.
that abrogate cytotoxic T cell responsiveness. By blocking
• In most cases, melanoma progresses from an intraepithelial
these pathways, checkpoint inhibitors reactivate the host
(in situ) to an invasive (dermal) form. Characteristics of the
T-cell response, which otherwise is held at bay. Current
dermal tumor such as depth of invasion and mitotic activity
efforts are focused on building upon these successes by
correlate with survival.
using combinations of different checkpoint inhibitors, as
908 C H A P T E R 24 Skin

Nestle FO, Kaplan DH, Barker J: Psoriasis, N Engl J Med 361:496, 2009.
SUGGESTED READINGS [A discussion of the pathogenesis, clinical features, and targeted treatment
Bastian BC: The molecular pathology of melanoma: an integrated options for psoriasis.]
taxonomy of melanocytic neoplasia, Ann Rev Pathol 9:239, 2014. [A Ratushny V, Gober MD, Hick R, et al: From keratinocyte to cancer:
modified classification of melanoma based on both clinical and genetic the pathogenesis and modeling of cutaneous squamous cell carci-
features. Such molecular classification schemes are critical for progress in noma, J Clin Invest 122:464, 2012. [Models of human epidermal carcino-
targeted therapy.] genesis indicate that multiple mutations in specific pathways are required
Cancer Genome Atlas Network: Genomic classification of cutaneous for malignant transformation.]
melanoma, Cell 161:1681, 2015. [A genomic study of cutaneous mela- Ujiie H, Shibaki A, Nishie W, et al: What’s new in bullous pemphi-
noma that highlights the different molecular subtypes and common patho- goid, J Dermatol 37:194, 2010. [A review of bullous pemphigoid
genic pathways.] pathogenesis.]
Elder DE: Dysplastic nevi: an update, Histopathology 56:112, 2010. [A Wargo JA, Cooper ZA, Flaherty KT: Universes collide: combining
balanced presentation of the histology and pathogenesis of dysplastic nevi immunotherapy with targeted therapy for cancer, Cancer Discov
and their relationship to melanoma.] 4:1377, 2014. [A presentation of melanoma signaling pathways with
Epstein EH: Basal cell carcinomas: attack of the hedgehog, Nat Rev therapeutic interventions and the role of the immune system.]
Cancer 8:743, 2008. [A succinct review of the epidemiology, clinical pre- Yokoyama T, Amagai M: Immune dysregulation of pemphigus in
sentation, molecular pathogenesis, and novel treatment options for basal humans and mice, J Dermatol 37:205, 2010. [A review of immune dis-
cell carcinoma.] turbances that may underlie pemphigus.]

You might also like