You are on page 1of 7

DOI: 10.1111/exd.

12880
www.wileyonlinelibrary.com/journal/EXD
Review

Keratoacanthoma: a distinct entity?


Tobias Gleich, Elena Chiticariu, Marcel Huber and Daniel Hohl
Service of Dermatology, University Hospital Center and University of Lausanne, Lausanne, Switzerland
Correspondence: Daniel Hohl, CHUV, Service de Dermatologie, 1011 Lausanne, Switzerland, Tel.: +41 21 3140353, Fax: +41-21-314 0382,
e-mail: daniel.hohl@chuv.ch

Abstract: Keratoacanthoma (KA) are common but exceptional signature caused by alterations in the TGFb signalling pathway.
benign tumors, often appearing on sun-exposed areas of light These recent findings will help to understand the nature of KA
skinned people and showing spontaneous resolution. The goal of and to develop new reliable diagnostic tools, simplifying the
this study was to review existing literature, to point out the discrimination of the histologically similar KA and SCC.
etiological complexity of KA biology and to answer the
Key words: keratoacanthoma – paradoxical activation – squamous cell
controversial debate if or not KA is a distinct entity or a variant
carcinoma – TGFb – tumor
of squamous cell carcinoma (SCC). Relying on recent results, we
highlight that KA is an individual lesion with a unique molecular Accepted for publication 12 October 2015

Introduction Classification
The first description of the ‘crateriform ulcer of the face’ was Several forms of KA exist, with solitary KA as the most common
given by Jonathan Hutchinson in 1889 (1), and since then many variant. Multiple KA have been described by Ferguson Smith,
different synonyms like molluscum sebaceum, self healing-epithe- Grzybowski and Witten-Zak.
lioma and keratocarcinoma have been used (2–4). In the 19400 s Solitary KA
Freudenthal proposed the name Keratoacanthoma (KA) to high- 1. The typical solitary KA is rapidly growing up to 2 cm,
light acanthosis and keratosis as major histological changes of the appearing as a rose nodule with a stretched shiny epidermis
tumor (4–7). KA however is a benign tumor, characterized by and a keratotic centre. After this mature phase with a dura-
rapid growth over 4–5 weeks followed by spontaneous regression tion around 6 months, the lesions becomes smaller until
(5,8). KA occurs mainly as a solitary lesion on sun-exposed loca- complete regression (5).
tions (9–11), but multiple tumors appear in several syndromes 2. Giant KA, usually grow larger than 2 cm, predominantly
such as familial self-healing epithelioma of Ferguson-Smith, Xero- appears on eyelids and nose (35–37). Unusual vertical
derma pigmentosum or Muir-Torre syndrome (12,13). Morpho- growth invading dermal and underlying tissue is observed
logically similar to squamous cell carcinoma (SCC), a better (38–40).
biological understanding of KA as defined as a self-healing benign 3. Subungual KA is a rare KA variant, appearing under nails,
tumor is needed to develop reliable diagnostic tools. Since recent show rarely spontaneous regression and may destroy the
findings showed that altered TGFb signalling is the main cause of terminal phalanx (10,41,42).
familial KA, a detailed view on this pathway should allow new 4. Mucosal KA another extremely rare KA variant, has also no
insights. tendency to regress (43) and is observed mostly intraorally
Etiology (44). Since intraoral mucous membranes bear no follicles it
Sun exposure and thus UV light was known for a long time to be was suggested, that these KA may arise from ectopic seba-
necessary for KA development (14) as suggested by preferential ceous glands (45).
localization on sun exposed skin of elderly patients with light 5. Keratoacanthoma centrifugum is a rare exoendophytic type,
complexions, an increased appearance in summer and autumn, characterized by multifollicular origin, progressive periph-
and their frequent emergence in Xeroderma pigmentosum eral expansion (2–40 cm) and atrophic central healing (46–
(10,11,15–19). Solitary and multiple KA have also been observed 48). Resolving of the tumor occurs spontaneously and invo-
in PUVA-treated patients (20,21). lution is mostly complete within week 12 (5,49).
Similarly, KA emergence was reported after cutaneous X-ray or Multiple KA
Megavolt radiation therapy (13,22,23). Further etiological factors 1. Ferguson Smith type, known as multiple self-healing squa-
are immune-compromised or immune-suppressed patients (24), mous epithelioma (MSSE), is the most common form of
skin trauma when pretreated with chemicals (18), carbondioxide multiple KA. This autosomal dominant genodermatosis is
ablation therapy, likely by induction of a wound healing response characterized by sudden appearance and rapid growth, slow
(13,22,23), and most importantly, genetic predisposition in multi- regression and periodical recurrence (25,50). The number
ple KA of Ferguson Smith type (25). Drugs as Imiquimod and of tumors varies from a few to hundreds, mostly leaving
BRAF-inhibitors induce KA as a side effect (26–31). Contradictory deep scars after involution. With a mean onset in young
results were published on the implication of human papilloma adolescence, appearance of KA is much earlier than the typ-
viruses (HPV) (5,32–34). ically reported in sporadic cases (10). In 2011, Goudie et al.

ª 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
Experimental Dermatology, 2016, 25, 85–91 85
Gleich et al.

(51) identified 11 monoallelic mutations in transforming Proliferation


growth factor beta receptor 1 (Tgfbr1) revealing the cause The initial, rapidly proliferating immature KA appears as a skin-
of the MSSE phenotype. collared papule with an acanthotic hyperproliferating epidermis
2. Generalized eruptive keratoacanthomas of Grzybowski characterized by hypergranulosis with large keratohyaline granules
appear spontaneously in 6–8 months represented by hun- and hyperkeratosis with premature keratinization. Enlarged ker-
dreds to thousands follicular papules (52,53). Skin involve- atinocytes are noticed within the stratum basale and spinosum
ment may be severe with masked facies and ectropion. (4). Eosin staining highlights keratinized areas with a pale cyto-
There is neither preference in sex nor in a familial pattern plasm giving the impression of a ‘glassy’ appearance (13,66). Later
(52,54). Fusing tumor nodules can form larger lesions in proliferation, a central depression and inflammatory infiltrates
rather involving the mucosa than palms and soles. The consisting primarily of neutrophils and less eosinophils are present
tumors may be indistinguishable from solitary KA (52). (4,10). Perineural invasion is rarely observed. Invasion beyond the
3. Multiple KA of Witten and Zak type arise in a range of level of eccrine glands is an ominous sign since KA usually not
1–30 papules which stay either small or increase their size extend deeper than sweat glands (10,67).
enormously. The lesions are showing central healing and Maturity
the life cycle with resolution is completed after several The typical prominent dome shaped symmetric nodule with a ker-
months. A familial predisposition is assumed as well as sun- atin plug in its centre is the prerequisite for correct diagnosis. Epi-
light as an etiological factor (55). dermal lips rise around both sides and cover partially the top of
4. KA in Muir-Torre syndrome appear solitary or multiple the crater (Fig. 1). Thereby, both elongated thin tumor lips are
(10). Muir-Torre syndrome is an autosomal dominant geno- covered on the surface and towards the crater by two epidermal
dermatosis combining visceral malignancy, notably colon sheets which are in parallel with the skin surface. The keratin filled
cancer with sebaceous neoplasms (56). The syndrome is crater plonges into multilocular spaces of epithelial tumor lobules
caused by germline mutations of hMSH2 and hMLH1 in the periphery in a semi-star shaped fashion (68,69). These are
genes, involved in the mismatch repair system. KA appear- composed of well-differentiated squamous cells at the dermal
ing within this syndrome is explained by the common interface (9). Thereby the tumor does not appear to grow verti-
pilosebaceous gland origin of sebaceous neoplasms and KA cally into the dermis like SCC but into all directions below the
(10). skin surface. Atypical basal keratinocytes are decreased in number
5. KA have been observed in patients suffering xeroderma pig- as are keratinocytes with pale and glassy cytoplasm (13). However,
mentosum, an autosomal recessive disorder with 100% pen- apoptotic keratinocytes and intraepithelial abscesses primarily
etrance, caused by defects in genes coding for DNA repair composed of neutrophils and less eosinophils become prominent
enzymes (57). Compared to normal cells, cells of XP (9,10). Horn pearls arranged in concentric keratinocyte layers with
patients fail to repair UV-induced DNA lesions, thereby a central eosinophilic keratin plug, also observed in SCC, are com-
linking sun exposure to a hypermutable genotype (58). This mon (4,10,13). Increased dermal inflammatory infiltrates com-
failure of the repair system causes development of mela- posed of eosinophils, neutrophils, plasma cells and histiocytes are
noma, basal cell carcinoma, SCC and KA in early childhood observed (4,9).
on sun exposed areas of the body (15).
6. Non-familial multiple persistent KA have been observed on
conjunctivae, palms and soles. In one special case, the
tumor continued to grow over a time period of 35 years (a) (b)
(10). Affleck (59) described this distinct variant as sporadic,
idiopathic without spontaneous resolution.
7. Reactive KA is induced by treatment of melanoma patients
with the BRAF kinase inhibitors Vemurafenib or Sorafenib
(29,30). These patients often develop multiple disseminated
solitary KA on sun-exposed and non-sun-exposed areas
(60). The iatrogenic tumor formation usually occurs
8 weeks after inhibitor therapy initiation (29), by paradoxi-
cal activation of the ERK MAPkinase pathway (61). (c)
Histopathology
Three different stages are determined by histological examination
during evolution of KA: (1) a proliferative phase, (2) the resting
fully developed mature stage and (3) regression (10). The majority
of tumors arises over the skin surface (62) embedded in a well- Figure 1. Keratoacanthoma in a proliferative to mature stage (a), in a regressing
differentiated squamous cell epithelium (63,64). A keratin-filled stage (b) and a horn pearl (c). (a) A multilocular crater caused by the follicular
origin of the lesion and epithelial lips arising on both sites of the lesion are well
epidermal invagination, eventually reaching into the dermis, recognizable as well as inflammatory infiltrates of the dermis accumulating at the
emerges from an adjacent hair follicle (65) (13,66). Atypical squa- borders to the epidermis. (b) Regressing keratoacanthoma (KA) maintaining the
typical architecture like multilocular crater and epithelial lips but the tumor
mous cells with multiple mitotic figures have been observed in decreased in size and height. Dermal inflammation is reduced but still present in
dermal extensions (13). this stage. (c) Horn pearl with keratotic plug, including eosinophilic cells.

ª 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
86 Experimental Dermatology, 2016, 25, 85–91
Keratoacanthoma

Regression the conclusion that KA is a benign neoplasm (8,51,85). Subungual


The third stage occurs relatively slowly. Most of the lesions invo- and mucosal KA appear under the global name of KA and arise
lute completely by week 12 (5). Involution is initiated by disap- on non-follicle bearing parts of the body. These borderline tumors
pearance of mitotic activity at the border of the lesion (4) and are histologically different from KA and SCC and show regression
loss of glassy, enlarged squamous cells. KA retain the crateriform only in a few cases (Table 3). Even if subungual KA show rather
architecture but decrease in height, followed by a reduction in progressive growth than spontaneous involution, cellular atypia
horizontal size (70) (Fig. 1). Around the tumor on the dermoepi- and the mitotic index are reduced compared to SCC (88,89) and
dermal interface a lichenoid inflammation reaction may be seen. several authors consider it to be benign (41,90,91).
The regression is likely based on immunological mechanisms
exhibited among others by activated CD4+ T lymphocytes (71).
Mitotic fibroblasts beneath the lesion forcing fibrosis and a dermal
Table 2. Table of differences of keratoacanthoma (KA) and squamous cell
infiltrate with multinucleated histiocytes may be observed (10). carcinoma (SCC)
Recent finding suggested that increased differentiation and loss of
KA SCC
proliferating epithelial cells together with inhibition of the Wnt-b-
catenin pathway are key steps in KA regression (72). Similarly, Biological differences
Rapid growth till 1–2 cm Slowly growing
SOX2 deletion in SCC led to decreased proliferation and increased Involution No involution
apoptosis and consequently to tumor regression in mice (73). Exoendophytic Predominantly endophytic
Histologpathologic differences
After complete resolution, the tumor leaves a fibrous scar without Epithelial lips –
adnexal structures (74). – Stromal desmoplasia
Discrimination: KA versus SCC Intraepithelial abscesses in the lesion Rarely observed
Rare ulceration Common
Classification controversy Distinct edge between Indistinct
Clinical and morphological distinction between KA and SCC may tumor and stroma
– Anaplasia
cause difficulties in certain cases (Tables 1, 2 and 4). Also the Intraepithelial abscesses with No association between
question whether KA is a distinct entity (75–77) or a variant of acantholytic cells eosinophils and acantholytic cells
Flask shaped –
SCC (43,78) was discussed controversially until molecular identifi- Epithelial collarette Rarely observed
cation of TGFbR1 mutations in Ferguson Smith syndrome and Symmetric Asymmetric
– Melanocytes present
genetic studies clarified that at least familial KA have bona fide an Absence of stromal desmoplasia Present
own pathogenetic cause (51,77,79,80). Indeed, the mutation rate
of TGFBR1 for supposed MSSE is about 85–90% (B. Lane, per-
sonal communication). A recent study analysing a large number
Table 3. Table of histopathological differences between solitary
of SCC by exome and targeted sequencing reported a comparable keratoacanthoma (KA) and its variants
frequency of ‘gatekeeper’ mutations in NOTCH1/2 (82%) while
Differences to
TGFBR1 mutations were much less frequent (81). Further studies corresponding Similarities to
are needed to confirm that this is a consistent fundamental dis- Variants Differences from squamous cell corresponding
of KA solitary KA carcinoma (SCC) SCC
tinction between all KA and SCC. Furthermore, the topic if KA is
essentially benign is lively debated and was forced by occasionally Ferguson • Deep
Smith extension
malignant behaviour of KA (82). If KA are truly able to form • Not
metastasis is still a matter of debate, in contrast with SCC which crateriform
metastasize in up to 5% (83,84). As reported, these metastasizing • No glassy
keratinocytes
KA could have been misdiagnosed as KA but are true SCC
Grzybowski • No differences
(5,8,69,76,78,85–87). In fact 10% as KA diagnosed tumors are
malignant and progressing SCC (9,69). Hallmarks of malignant Giant • Dimension
tumors such as, prominent mitoses or cytological atypia, are not Mucosal • No involution • Rare ulceration • No regression
observed in KA. Spontaneous regression and missing features • Rarely • Mitotic activity
crateriform • Low metastatic
characterizing a malignant phenotype, prompted us and others to • No fibrosis potential

KA • Morphology
centrifugum • Size
Table 1. Table of common features of keratoacanthoma (KA) and squamous marginatum
cell carcinoma (SCC) Subungual • No involution • Age of • Painful nodular
• Dyskeratotic appearance lesion on distal
Common biological features cells • KA evolves phalanx
Lateral growth predominant • No fibrosis rapidly, • Endo-exophytic
Metastatic potential • Little or no gathering • Radiometrically
Local tissue destruction (if untreated) atypia a tumor-like very similar
Similar etiologic factors (UV exposure) configuration
Common histopatological features • KA can regress
Intraepithelial polymorphonuclear abscesses • No increase
Parakeratosis of Ki-67 or
Dyskeratosis p53 expression
Cup shaped
Atypical squamous cells
Glassy keratinocytes Comparison of the rare subungual and mucosal KA with its SCC counterparts.

ª 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
Experimental Dermatology, 2016, 25, 85–91 87
Gleich et al.

In sum, the recently found TGFbR1 driver mutations in familial Proteins functioning in cellular adhesion processes
KA, Comparative Genomic Hybridization studies, micro array Differences in VCAM1 and ICAM1 expression in KA compared to
experiments and missing clinical features of a malignant pheno- SCC were not sufficient for a reliable marker (98). Significantly
type in KA are in line with our view that KA and SCC are two increased expression of stromelysin (ST3) was found in SCC and
distinct lesions (51,77,79,80). Unfortunately, there is still a notable even more in metastatic SCC allowing to predict their biological
lack of a reliable molecular marker, allowing for the unequivocal behaviour (99).
distinction of KA from SCC in the laboratory. Cell surface receptors
Histological criteria EGFR is a receptor tyrosine kinase affecting epithelial and ker-
The requirement to distinguish KA from SCC is complete excision atinocyte differentiation, proliferation and tumorgenesis (100).
of the lesion reaching to the subcutis including the crater, both Canonical EGFR signalling occurs through mitogen activated
lips and the surrounding of uninvolved epidermis (10). KA are kinase pathway (MAPK), and thus through Ras and Raf proteins.
exoendophytic whereas the majority of cutaneous SCC show inte- Comparative genetic hybridization identified different chromoso-
rior proliferation (13). For reliable diagnosis, dermatolopatholo- mal aberration patterns in KA compared with SCC providing evi-
gists have to include several criteria. Architectural attributes are dence that KA and SCC are two distinct entities (80).
essential since both tumors possess nearly identical cytological fea- Consequently, EGFR copy numbers were altered more often in
tures (Tables 1–3) (10). SCC but amplification was not significant, and no increased EGFR
Cribier et al. examined 14 criteria on 262 previously diagnosed protein expression could be observed (101–103). However, this is
samples and defined five significant criteria: (1) epithelial lips, controversial, as increased expression of EGFR was detected in the
(2) sharp outline between tumor and stroma favouring KA- (3), majority of primary and metastatic SCC but not in KA (100,104).
and ulceration, (4) abundant mitotic cells, (5) pleomorphism/ This may indicate that EGFR expression in skin carcinogenesis is
anaplasia SCC-favouring. These features allowed a consensual rather facilitating progression (104).
diagnosis in 88% of cases, although they did not increase the As in many tumors, alteration of Ras and Raf proteins are also
sensitivity in difficult cases (92). Other studies claimed inflamma- important for SCC and KA formation. Both can develop as sec-
tion or pattern of keratinization as reliable distinctive features ondary tumors upon melanoma treatment with BRAF inhibitors
(Table 4) (10,75). (29,105).
Cytological markers As shown recently, TGFb signalling is implicated in KA forma-
Cytokines tion. Eleven monoallelic mutations in the Tgfbr1 gene were speci-
PCR screening showed elevated levels of interleukin 10 (IL10) fied in Ferguson Smith disease (51). The role of TGFb signalling
and a decrease of granulocyte macrophage colony-stimulating in KA and SCC formation will be discussed in detail in this
factor (GM-CSF) in KA compared with SCC. IL10 usually report.
exhibits anti-tumoral but also immunosuppressive functions Cell cycle and apoptosis regulators
(93,94) thereby, inhibiting eosinophile GM-CSF and Th1 Ki-67 expression varies in KA since proliferative KA display higher
cytokine production (95). Low GM-CSF serum concentration immunostaining frequency than mature KA. Staining of p53 was
leading to an immunosuppressive state probably enables rapid strong and mainly localized to expanding tumor islands in KA
tumor growth on one hand and allows involution on the other and a diffuse nuclear staining throughout the whole lesion in
hand (96). SCC. Expression of p53 in KA and SCC is not sufficiently discrim-
Some studies reported diffuse distribution of transforming inatory to use it as a diagnostic tool but may be supporting the
growth factor-a (TGFa) in the majority of KA. Forty per cent of differential diagnosis of subungual KA versus subungual SCC
SCC exhibit a focal enrichment of TGFa whereas 90% of KA exhi- (88,89,106,107). Cyclin A and B exhibited predominantly basal
bit no staining in this area (97). and parabasal staining in KA, whereas it was diffuse in SCC (108).
In sum, these and many other targets have been tested in the
last decades, but no reliable marker was identified exhibiting suffi-
Table 4. Table of features to reliable distinguish keratoacanthoma (KA) and cient sensitivity and specificity.
squamous cell carcinoma (SCC) described in publications
Genetics
KA SCC Array CGH
Histopathology Array comparative genomic hybridization (array CGH) allows the
Cribier et al.: Cribier et al.: discrimination of KA and SCC in 85% of the cases. Recurrent
• Epithelial lips • Ulceration
• Sharp outline between • Numerous mitosis
aberrations on chromosome 7, 8 and 10 have been the best pre-
tumor and stroma • Pleomorphism/anaplasia dictors for SCC, suggesting a role in prevention of apoptosis or
Schwartz et al.: Schwartz et al.: activation of proliferation and infiltration for genes located in
• Intratumoral abscesses with • Intratumoral abscesses contain these areas. In contrast chromosomes 17, 19, 20 and X seemed to
granulocytes and acantholytic cells few or no inflammatory cells
be aberrant mainly in KA, indicating crucial genes for KA devel-
Weedon et al.: Weedon et al.: opment. These and other differences in the aberration pattern
• Pattern of keratinization • Pattern of keratinization
between the two tumors were proven to be significant, leading to
Genetics the assumption that KA and SCC are two distinct entities (79). A
Li et al.: Li et al.:
• Aberrations on chromosome: • Aberrations on chromosome: recent study investigating the relationship between KA and SCC
17, 19, 20, X 7, 8, 10 by DNA microarray technique concludes that KA and SCC are

ª 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
88 Experimental Dermatology, 2016, 25, 85–91
Keratoacanthoma

distinct, further defining KA as a distinct regressing neoplasm Haploinsufficient TGFb1+/ mice developed fewer benign
(77). tumors with reduced incidence and size compared with TGFb1+/+
Molecular pathways contributing to SCC and KA mice. Even if TGFb+/+ mice had a higher onset of benign tumors,
formation no increased numbers of SCC were observed (121).
TGFb signalling in KA and SCC The fact that KA and SCC appearance is linked to TGFb alter-
The implication of TGFb signaling in KA formation was demon- ation is not sufficient to conclude that they are one entity. Since
strated recently by Goudie et al. They ascertain a clear correlation the molecular background of SCC and KA differs strongly it is
between loss of function mutations in the Tgbr1 gene and a MSSE very probable that the outcome of TGFb signalling is also altered
phenotype. With high-throughput genomic sequencing they iden- (77). Consequently, stem cells within the same SCC tumor, exhibit
tified the disease locus on chromosome 9 and by exome sequenc- opposite phenotypes depending on if they respond to TGFb stim-
ing they localized in three unrelated families several distinct uli or not (122). Furthermore, rather than Tgfbr1 mutations,
mutations in Tgfbr1. In total, 11 monoallelic mutations in 18 fam- recently identified NOTCH1/2 receptor mutations are considered
ilies have been described, occurring in the cytoplasmic kinase as early gatekeeper mutations in SCC development (81).
domain or in the extracellular ligand-binding domain. In the cyto- Par3 signalling in KA formation
plasmatic region of the receptor nonsense or frameshift mutations The Par3 pathway is implicated in KA formation and affects
occurred, resulting in a null mutation. In the extracellular ligand- downstream targets of TGFbR1 such as PI3K/Akt and MAPK.
binding domain frameshift, nonsense or missense mutations have Specifically, Par3 is involved in apico-basal cell polarity and asym-
been demonstrated, affecting the binding of TGFb or the interac- metric cell division. Loss of cell polarity appears to be a prerequi-
tion with TGFbR2. These mutations were shown to be null sup- site in tumor formation and progression. In DMBA/TPA tumor
ported by the fact that the bases of three of four of the missense models, Par3 overexpression facilitates papilloma formation but
mutations are highly conserved across vertebrates. These heterozy- reduces KA development. Opposite effects were observed in the
gous MSSE mutations exhibit a loss of function, which is covered same tumor model with Par3 ablation, what may be explained by
by the functional protein transcribed by the wildtype allele (51). its different intracellular localizations (123). Whether staining of
The paradoxical role of TGFb signalling could explain the rapid Par3 may be helpful in histopathology needs to be investigated.
evolution and involution of KA after ablation of TGFb mediated RAF inhibitors favours KA and SCC development
signal transduction. Additionally, a discontinuous membranous A specific category of KA appears in melanoma patients treated
expression was observed in spontaneous KA but rarely in SCC. If with BRAF inhibitors (29,61,105). Ninety per cent of all BRAF
this was due to mutation in the receptor could not be answered mutations are V600E changes, causing a higher BRAF activity
definitively (109). thereby activating MAPK signalling pathway independently of
Transforming growth factor beta receptor 1 and 2 (TGFbR1 RAS (124,125). A side effect of the mutation-specific BRAF V600E
and TGFbR2) are transmembrane serine/threonine kinases, trans- drug Vemurafenib in melanoma patients is the appearance of SCC
ducing signals of the TGFb protein superfamily from the cell sur- or KA in 15–20% of the patients. Sixty per cent of these epidermal
face to the cytoplasm. Downstream signalling occurs mostly lesions have a mutated RAS leading to BRAF inhibitor mediated
through SMAD-proteins but also affects Ras, PI3K/Akt- or paradoxical MAPK pathway activation in BRAF wild-type cells
MAPK-pathways (110,111). TGFb signalling is found in a large (29). Melanoma patients treated with Sorafenib also developed
number of tissues where it controls differentiation and growth SCC or KA lesions in up to 7% of the cases (30,31).
(112) and alterations are implicated in malignant transformation Interferon treatment
of keratinocytes and tumorgenesis (113,114). Imiquimod, a nucleoside analogue of the imidazoquinolone fam-
Transgenic mice overexpressing TGFb, were protected against ily, is usually applied against actinic keratoses and basal cell carci-
an early development of tumors but promoted tumor metastasis noma. It leads to upregulation of pro-inflammatory cytokines due
later in tumorgenesis (111). A widely accepted hypothesis is that to activation of the NFkB pathway. Recently, there is a growing
an effect favouring tumor development is mediated by paracrine evidence of Imiquimod induced KA as a secondary effect of the
signalling, stimulating angiogenesis and inflammation at late stages treatment (26,28).
whereas autocrine signalling seems to function as tumor inhibitor Treatment of KA and SCC
at least in early stages (115). Secondary effects as onset of inflam- Treatment of patients with diagnosed KA include excision (stan-
mation or angiogenesis upon TGFb overexpression seem to be dard or Mohs), radiotherapy, systemic retinoids and intralesional
responsible for keratinocyte hyperproliferation of head and neck methotrexate (19,126). Mohs surgery should be used in the appli-
epithelium in vivo (116). cable case (19). Application of a Imiquimod 5% cream led to fast
TGFbR1 deletion leads in a small number of animals to sponta- regression of KA (127). Regarding the patients interest, conserva-
neous formation of HNSCC, thus indicating that loss of the recep- tive handling or Mohs surgery are the treatments of choice.
tor is not an initiation event in tumor formation (117). Conclusion
Concomitant loss of TGFbR1 and of PTEN provoked the forma- The etiology of KA involves many different factors, of which some
tion of spontaneous tumors (118). Combining TGFbR2 ablation are seemingly more evident than others giving rise to a broad
with either overexpression of oncogenic K-ras or DMBA treatment spectrum of KA variants (10,11,13–30,32,33,69,128). Genetic pre-
led rapidly to the formation of aggressive growing SCC (119,120). disposition or spontaneous formation adds another level to the
Glick et al. (113) reported significantly augmented tumor forma- complex classification system of KA. Today we conclude that SCC
tion after skin graft experiments of TGFb1 negative keratinocytes and the benign neoplasm KA are two distinct entities, prototypi-
expressing virally transduced oncogenic v-ras. cally evidenced by alterations in the TGFb pathway in Ferguson

ª 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
Experimental Dermatology, 2016, 25, 85–91 89
Gleich et al.

Smith Disease, chromosomal aberration differences and pro- remains challenging. Consequently, it is essential to find morpho-
nounced alterations in gene expression (51,77,79,80). Since logical, biological and/or molecular markers which are showing
TGFbR1 mutations were identified as driver mutations in Fergu- reliable differences between these two lesions to prove that a given
son Smith Disease, it would be important to identify mutations KA is not a SCC. Taken together, KA is a rapidly growing, sponta-
leading to spontaneous KA and other KA variants e.g. by identify- neously regressing tumor. Recently published data lead us to
ing more disease loci followed by high-throughput genomic assume that KA is distinct from SCC and caused by early gate-
sequencing. Additionally, the identification of pathways implicated keeper alterations in the TGFb pathway.
in the spontaneous involution of KA would be of serious interest. Acknowledgements
With regard to the development of new therapeutics it would be This work has been supported by grants to M.H. and D.H. by the Swiss
interesting to investigate the immune response in KA during dif- National Science Foundation and the Dind Cottier Foundation.
ferent stages, leading to new insights concerning presented tumor Author contribution
antigens or identification of specific reactive T-cells. Up to now, T.G. wrote and conceptualized this review. E.C. and M.H. revised the
dermatopathologists have a difficult task to distinguish KA and paper. D.H. conceptualized, revised and coordinated the work.
SCC on histopathological grounds. For this purpose a fully excised Conflict of interests
lesion is helpful, but to discriminate both tumors in an early stage The authors have declared no conflicting interests.

References
1 Hutchinson J. Trans Pathol Soc London 1889: 30 Arnault J P, Mateus C, Escudier B et al. Clin 61 Holderfield M, Deuker M M, McCormick F
40: 275–281. Cancer Res 2012: 18: 263–272. et al. Nat Rev Cancer 2014: 14: 455–467.
2 Maccormac H, Scarff R. Br J Dermatol 1936: 31 Di Marco V, De Vita F, Koskinas J et al. Ann 62 Levy E J, Cahn M M, Shaffer B et al. J Am
48: 624–626. Oncol 2013: 24 (Suppl 2): ii30–ii37. Med Assoc 1954: 155: 562–564.
3 Dunn J, Smith J F. Br J Dermatol 1934: 46: 32 Zelickson A. Acta Derm Venereol 1962: 42: 23. 63 Brenn T, McKee P, Mihm M. Keratoacanthoma.
519. 33 Payne D, Newman C, Tyring S. J Am Acad Der- In: McKee P et al., ed. Pathology of Skin.
4 Kwittken J. Mt Sinai J Med 1975: 42: 127– matol 1995: 33: 1047. Philadelphia: Elsevier Mosby, 2005: 1221–
135. 34 Ghadially R, Ghadially F N. Keratoacanthoma. 1227.
5 Rook A, Whimster I. Br J Dermatol 1979: 100: In: Fitzpatrick T B, Eisen A Z, Wolff K, eds. Der- 64 Fisher E R, McCoy M M, Wechsler H L. Cancer
41–47. matology in General Medicine. New York: 1972: 29: 1387–1397.
6 Miedzinski F. Postepy Dermatol (Poznan) 1990: McGraw-Hill, 1993: 845–855. 65 Ghadially F N. Cancer 1961: 14: 801–816.
7: 61–70. 35 Leonard A L, Hanke C W. J Drugs Dermatol 66 Wade T, Ackerman A. J Dermatol Surg Oncol
7 Musso L, Proc R. Soc Med 1950: 43: 838. 2006: 5: 454. 1978: 4: 498.
8 Mandrell J C, Santa Cruz D. Semin Diagn 36 De Visscher J, van der Wal J, Starink T M et al. 67 Cooper P H, Wolfe J T 3rd. Arch Dermatol
Pathol 2009: 26: 150–163. Oral Surg Oral Med Oral Pathol Oral Radiol 1988: 124: 1397–1401.
9 Beham A, Regauer S, Soyer H P et al. Adv Anat Endod 1996: 81: 193–196. 68 Ghadially F. J Pathol Bacteriol 1958: 75: 441–
Pathol 1998: 5: 269–280. 37 Webb A, Ghadially F. J Pathol Bacteriol 1966: 453.
10 Schwartz R A. J Am Acad Dermatol 1994: 30: 91: 505–509. 69 Yus E S, Simo n P, Requena L et al. Am J Der-
1–19; quiz 20–12. 38 Lejman K, Starzycki Z. Br J Vener Dis 1977: 53: matopathol 2000: 22: 305–310.
11 Schwartz R A. Keratoacanthoma. In: Chu A C, 65–67. 70 Ko C J, McNiff J M, Bosenberg M et al. J Am
Edelson R L, eds. Malignant Tumors of the 39 Rapaport J. Arch Dermatol 1975: 111: 73. Acad Dermatol 2012: 67: 1008–1012.
Skin. London: Chapman & Hall, 1999: 96–118. 40 Saito M, Sasaki Y, Yamazaki N et al. Plast 71 Patel A, Halliday G, Cooke B et al. Br J Derma-
12 Karaa A, Khachemoune A. Int J Dermatol Reconstr Surg 2003: 111: 1561–1562. tol 1994: 131: 789–798.
2007: 46: 671–678. 41 Cramer S, Am J. Clin Pathol 1981: 75: 425. 72 Zito G, Saotome I, Liu Z et al. Nat Commun
13 Schwartz R A. J Surg Oncol 1979: 12: 305– 42 Patel M R, Desai S. J Hand Surg Am 1989: 14: 2014: 5: 3543.
317. 139–142. 73 Boumahdi S, Driessens G, Lapouge G et al.
14 Kingman J, Callen J P. Arch Dermatol 1984: 43 Choonhakarn C, Ackerman A. Dermatopathol Nature 2014: 511: 246–250.
120: 736. Pract Concept 2001: 7: 7–16. 74 Blessing K, Nafussi A, Gordon P. Histopathol-
15 Dogliotti M, Caro I. Int J Dermatol 1976: 15: 44 Janette A, Pecaro B, Lonergan M et al. J Oral ogy 1994: 24: 381–384.
524. Maxillofac Surg 1996: 54: 1026–1030. 75 Weedon D. Curr Diagn Pathol 2003: 9: 259–265.
16 Dufresne R, Marrero G, Robinson-Bostom L. Br 45 Svirsky J A, Freedman P D, Lumerman H. Oral 76 Weedon D, Malo J, Brooks D et al. ANZ J Surg
J Dermatol 1997: 136: 227–229. Surg Oral Med Oral Pathol 1977: 43: 116–122. 2010: 80: 129–130.
17 Chuang T-Y, Reizner G T, Elpern D J et al. Arch 46 Belisario J C. Aust J Dermatol 1965: 8: 65–72. 77 Ra S H, Su A, Li X et al. Mod Pathol 2015: 28:
Dermatol 1993: 129: 317. 47 V’Lckova-Laskoska M T, Laskoski D S. Clin Exp 799–806.
18 Ghadially F, Barton B, Kerridge D. Cancer Dermatol 2008: 33: 259–261. 78 Hodak E, Jones R E, Ackerman A B. Am J Der-
1963: 16: 603–611. 48 Sung C S, Chuang F C, Ho J C et al. Dermato- matopathol 1993: 15: 332–342.
19 Schwartz R A. Dermatol Surg 2004: 30: 326– logica Sinica 2014: 32: 25–28. 79 Li J, Wang K, Gao F et al. J Invest Dermatol
333; discussion 333. 49 Aydin F, Senturk N, Sabanciler E et al. Clin Exp 2012: 132: 2060–2066.
20 Sina B, Adrian R M. J Am Acad Dermatol Dermatol 2007: 32: 683–686. 80 Clausen O P F, Aass H C D, Beigi M et al. J
1983: 9: 686–688. 50 Smith J F. Br J Dermatol 1934: 46: 267–272. Invest Dermatol 2006: 126: 2308–2315.
21 Maddin W, Wood W S. J Cutan Pathol 1979: 51 Goudie D R, D’Alessandro M, Merriman B 81 South A P, Purdie K J, Watt S A et al. NOTCH1
6: 96–100. et al. Nat Genet 2011: 43: 365–369. mutations occur early during cutaneous squa-
22 Gewirtzman A, Meirson D H, Rabinovitz H. 52 Consigli J, Gonzalez M, Morsino R et al. Br J mous cell carcinogenesis. J Invest Dermatol
Dermatol Surg 1999: 25: 666–668. Dermatol 2000: 142: 800–803. 2014: 134: 2630–2638.
23 Shaw J C, Storrs F J, Everts E. J Am Acad Der- 53 Grzybowski M. Br J Dermatol 1950: 62: 310– 82 Schnur P L, Bozzo P. Plast Reconstr Surg 1978:
matol 1990: 23: 1009–1011. 313. 62: 258–262.
24 Wagdy Dessoukey M, Omar M F, Abdel- 54 Schwartz R A, Blaszczyk M, Jablonska S. Der- 83 Joseph M G, Zulueta W P, Kennedy P J. ANZ J
Dayem H. J Am Acad Dermatol 1997: 37: matology 2002: 205: 348–352. Surg 1992: 62: 697–701.
478–480. 55 Witten V H, Zak F G. Cancer 1952: 5: 539– 84 Brantsch K D, Meisner C, Schonfisch B et al.
25 Ferguson-Smith M, Wallace D, James Z et al. 550. Lancet Oncol 2008: 9: 713–720.
Birth Defects Orig Artic Ser 1971: 7: 157. 56 Ponti G, Losi L, Di Gregorio C et al. Cancer 85 Ko C J. Clin Dermatol 2010: 28: 254–261.
26 Foxton G, Delaney T. Australas J Dermatol 2005: 103: 1018–1025. 86 Jackson I T, Brown G E D. Br J Plast Surg 1970:
2011: 52: 66–69. 57 Ponti G. Ponz de Leon M. Lancet Oncol 2005: 23: 373–379.
27 Pini A M, Koch S, Scharer L et al. J Am Acad 6: 980–987. 87 Ledo E. Int J Dermatol 1992: 31: 777–778.
Dermatol 2008: 59: S116–S117. 58 DiGiovanna J J, Kraemer K H. J Invest Dermatol 88 Honma M, Kato N, Hashimoto M et al. Clin
28 D’Addario S, Carrington P R. Acta Derm Vener- 2012: 132: 785–796. Exp Dermatol 2011: 36: 57–62.
eol 2006: 86: 366–367. 59 Affleck A G. Int J Dermatol 2007: 46: 1105. 89 Gonzalez-Rodriguez A J, Gutierrez-Paredes E
29 Sharma A, Shah S R, Illum H et al. Drugs 60 Harvey N T, Millward M, Wood B A. Am J Der- M, Montesinos-Villaescusa E et al. Actas Der-
2012: 72: 2207–2222. matopathol 2012: 34: 822–826. mosifiliogr 2012: 103: 549–551.

ª 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
90 Experimental Dermatology, 2016, 25, 85–91
Keratoacanthoma

90 Levy D W, Bonakdarpour A, Putong P B et al. 104 Shimizu T, Izumi H, Oga A et al. Dermatology 117 Bian Y, Terse A, Du J et al. Cancer Res 2009:
Skeletal Radiol 1985: 13: 287–290. 2001: 202: 203–206. 69: 5918–5926.
91 Oliwiecki S, Peachey R D, Bradfield J W et al. 105 LaPresto L, Cranmer L, Morrison L et al. JAMA 118 Bian Y, Hall B, Sun Z J et al. Oncogene 2012:
Clin Exp Dermatol 1994: 19: 230–235. Dermatol 2013: 149: 279–281. 31: 3322–3332.
92 Cribier B, Asch P-H, Grosshans E. Dermatology 106 Wrone-Smith T, Bergstrom J, Quevedo M E 119 Lu S L, Herrington H, Reh D et al. Genes Dev
1999: 199: 208–212. et al. J Dermatol Sci 1999: 19: 53–67. 2006: 20: 1331–1342.
93 Kundu N, Beaty T L, Jackson M J et al. J Natl 107 Connolly M, Narayan S, Oxley J et al. Clin Exp 120 Malkoski S P, Haeger S M, Cleaver T G et al.
Cancer Inst 1996: 88: 536–541. Dermatol 2008: 33: 625–628. Clin Cancer Res 2012: 18: 2173–2183.
94 Lippitz B E. Lancet Oncol 2013: 14: e218–e228. 108 Tran T A, Ross J S, Boehm J R et al. J Cutan 121 Perez-Lorenzo R, Markell L M, Hogan K A
95 Borish L. J Allergy Clin Immunol 1998: 101: Pathol 1999: 26: 391–397. et al. Carcinogenesis 2010: 31: 1116–1123.
293–297. 109 Berger F, Geddert H, Faller G et al. Pathol Res 122 Oshimori N, Oristian D, Fuchs E. Cell 2015:
96 Lowes M, Bishop G, Cooke B et al. Br J Cancer Pract 2014: 210: 596–602. 160: 963–976.
1999: 80: 1501. 110 Massague J. Nat Rev Mol Cell Biol 2012: 13: 123 Iden S, van Riel W E, Schafer R et al. Cancer
97 Ho T, Horn T, Finzi E. Arch Dermatol 1991: 616–630. Cell 2012: 22: 389–403.
127: 1167. 111 Bierie B, Moses H L. Nat Rev Cancer 2006: 6: 124 Pratilas C A, Xing F, Solit D B. Curr Top Micro-
98 Melendez N D, Smoller B R, Morgan M. Mod 506–520. biol Immunol 2012: 355: 83–98.
Pathol 2003: 16: 8–13. 112 Massague J, Blain S W, Lo R S. Cell 2000: 103: 125 Joseph E W, Pratilas C A, Poulikakos P I et al.
99 Asch P-H, Basset P, Roos M et al. Am J Der- 295–309. Proc Natl Acad Sci U S A 2010: 107: 14903–
matopathol 1999: 21: 146–150. 113 Glick A, Popescu N, Alexander V et al. Proc 14908.
100 Groves R W, Allen M H, MacDonald D M. J Natl Acad Sci U S A 1999: 96: 14949–14954. 126 Annest N M, VanBeek M J, Arpey C J et al. J
Cutan Pathol 1992: 19: 66–72. 114 Levy L, Hill C S. Cytokine Growth Factor Rev Am Acad Dermatol 2007: 56: 989–993.
101 Jacobs M S, Persons D L, Fraga G R. J Cutan 2006: 17: 41–58. 127 Di Lernia V, Ricci C, Albertini G. J Eur Acad
Pathol 2013: 40: 447–454. 115 De Wever O, Mareel M. J Pathol 2003: 200: Dermatol Venereol 2004: 18: 626–629.
102 Reuter C, Morgan M, Eckardt A. Br J Cancer 429–447. 128 Barrett J C. Environ Health Perspect 1993: 100:
2007: 96: 408–416. 116 Lu S L, Reh D, Li A G et al. Cancer Res 2004: 9–20.
103 Schlauder S M, Calder K B, Moody P et al. Am 64: 4405–4410.
J Dermatopathol 2007: 29: 559–563.

ª 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
Experimental Dermatology, 2016, 25, 85–91 91

You might also like