10 1016@j Det 2012 06 013

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

U p d a t e o n Va s c u l a r N e o p l a s m s

Omar P. Sangüeza, MD

KEYWORDS
 Wilms tumor  Kaposi sarcoma  Infantile hemangioma  Acquired elastotic hemangioma

KEY POINTS
 Wilms tumor 1 (WT1) is a tumor suppressor gene (map locus 11p13).
 Myc amplification is found in angiosarcomas secondary to radiation and chronic lymphedema but
not in atypical vascular proliferations secondary to radiotherapy.
 ERG is also useful in separating angiosarcomas and epithelioid hemangioendotheliomas from their
histologic mimics, such as nonendothelial tumors with corded, myxohyaline, and hemorrhagic,
highly vascular patterns.

NEW IMMUNOHISTOCHEMICAL MARKERS hemangioendothelioma was negative for WT1.


FOR THE DIAGNOSIS OF VASCULAR However, vascular malformations (2 port-wine
NEOPLASMS stains, 10 venous malformations, and 8 lymphatic
Wilms Tumor 1 malformations) did not show any positive endo-
thelial staining. More recently Trindade and
Wilms tumor 1(WT1) is a tumor suppressor gene
colleagues,9 confirmed these results and ex-
(map locus 11p13). The gene product, WT1/Wilms
panded these findings. They found expression of
tumor protein, is a transcription factor that
WT1 in arteriovenous malformations (AVMs), non-
contains 4 epidermal growth factor (EGR)–family
involuting congenital hemangiomas (NICHs)
C2H2-type zinc fingers at 323–347, 353–377,
(Fig. 1A and B), rapidly involuting congenital
383–405, and 414–438 and a 57 amino acid,
hemangiomas (RICHs), tufted angiomas, and
proline-rich region, which recognizes and binds
spindle cell hemangiomas. They also confirmed
the DNA sequence 50 -CGCCCCCGC-30 . This
the absence of WT1 expression in anomalous
gene plays an essential role in the normal develop-
vessels in capillary, lymphatic, and venous malfor-
ment of the urogenital system but is also involved
mations. All AVMs showed positivity for WT1 in the
in hematopoiesis and angiogenesis.1 The expres-
lesional endothelia; this could be related to the
sion of WT1 is maintained during the differentiation
proliferative stage of the AVM, because all their
of the various bone marrow stem cells into endo-
cases were in stage II, with arteriovenous shunt-
thelial cells.2 WT1 has been detected in acute
ings and clinical enlargement.
leukemias, breast carcinomas, melanomas, non–
small cell lung cancer, and carcinomas of the geni-
Claudins
tourinary tract.3–7
WT1 immunostaining has been used to discrim- Claudins are a family of proteins that are compo-
inate between vascular neoplasms and most nents of the tight junctions and are part of the
vascular malformations. A study by Lawley and cellular barrier that controls the flow of molecules
colleagues8 showed WT1 expression in infantile in the intercellular space between epithelial cells.
hemangiomas (8 of 9), pyogenic granulomas (2 They have 4 transmembrane domains, with the
of 2), angiosarcomas (9 of 9), epithelioid heman- N-terminus and the C-terminus in the cytoplasm.
gioendothelioma (1 of 1), and hobnail hemangi- They are expressed in various endothelia and in
omas (1 of 1). A single case of malignant some, especially juxtaluminal, glandular and
derm.theclinics.com

Departments of Pathology and Dermatology, Wake Forest University School of Medicine, Winston Salem,
NC 27157, USA
E-mail address: osanguez@wakehealth.edu

Dermatol Clin 30 (2012) 657–665


http://dx.doi.org/10.1016/j.det.2012.06.013
0733-8635/12/$ – see front matter Ó 2012 Elsevier Inc. All rights reserved.
658 Sangüeza

Fig. 1. (A) NICH staining strongly positive for WT1. (B) NICH-negative staining for GLUT1 (original magnification 40).

ductal epithelial cells.10 Claudin-5 is also required none phenomenon. ERG is also useful in separating
in cardiovascular development, and monoallelic angiosarcomas and epithelioid hemangioendothe-
loss of a chromosome 22 segment including liomas from their histologic mimics, such as nonen-
claudin-5 locus causes velocardiofacial syn- dothelial tumors with corded, myxohyaline, and
drome, including cardiac malformations.11 hemorrhagic, highly vascular patterns.13
Miettinen and colleagues12 found that claudin-5 ERG is more specific than other endothelial
is a sensitive marker for angiosarcoma and markers, such as CD31, the current gold standard
hemangioendothelioma but is not very specific, in the definition of angiosarcoma. ERG immuno-
considering its widespread expression in carci- reactivity is highly endothelium-restricted and
nomas. Claudin-5 is significantly expressed in straightforward to interpret, whereas interpretation
most angiosarcomas, indicating that this cell junc- of CD31 can be problematic, because it also stains
tion protein is generally conserved in endothelia histiocytes and plasma cells, which can lead to an
with malignant transformation. Claudin-5 is very overdiagnosis of angiosarcoma. CD31 also stains
specific for angiosarcomas. Only synovial sar- platelets.
comas are positive for this antibody. Claudin-5
is useful to differentiate from mimics of angiosar- c-Myc
coma, such as epithelioid sarcoma, and mela-
noma, which are consistently claudin-5–negative. c-Myc is a regulator gene that codes for a tran-
scription factor. A mutated version of Myc is found
in many cancers, which causes Myc to be consti-
Avian V-Ets Erythroblastosis Virus E26
tutively (persistently) expressed. This function
Oncogene Homolog
leads to the unregulated expression of many
Avian v-ets erythroblastosis virus E26 oncogene genes, some of which are involved in cell prolifer-
homolog (ERG) is a transcription factor of the ation and results in the formation of cancer.
erythroblast transformation–specific family. It is A common translocation involving Myc is t(8;14).
expressed in the nuclei of endothelial cells and in This translocation is found in some malignant
normal lymphatics. It is also expressed in subsets neoplasms, including secondary but not primary
of prostatic carcinoma, acute myeloid leukemia, angiosarcomas. This is the most frequent recur-
and Ewing sarcoma.13 rent alteration on chromosome 8q24.21 (50%), fol-
ERG stain is a highly specific endothelial lowed by 10p12.33 (33%) and 5q35.3 (11%).14
marker for vascular neoplasms. It can be useful Myc amplification is found in angiosarcomas
in differentiating hemangiomas, which have a secondary to radiation and chronic lymphedema
dual cell population (ERG-positive endothelial (Fig. 2A–C), but not in atypical vascular prolifera-
cells and ERG-negative nonendothelial compo- tions secondary to radiotherapy.15
nents, especially pericytes) from angiosarcomas,
which generally have only ERG-positive endothe- INFANTILE HEMANGIOMAS
lial components.
ERG is also expressed in malignant vascular Infantile hemangioma (IH) is the most common
endothelial neoplasms (hemangioendotheliomas, benign vascular proliferation, and traditionally has
angiosarcomas, and Kaposi sarcomas). The stain- been considered a neoplasm. This classification
ing is usually seen in most tumor cells, indicating may be true for a minority of these proliferations,
that the expression of the ERG transcription factor but most IHs are better considered as hyperpla-
in endothelial cells and angiosarcomas is an all-or- sias. Classical IHs, after an initial proliferative
Update on Vascular Neoplasms 659

Fig. 2. Angiosarcoma in radiated skin. (A and B) Irregular dilated spaces lined by prominent endothelial cells
(hematoxylin and eosin stain, original magnification A 20, B 40). (C) Positive staining for c-Myc of the endo-
thelial cells.

phase, undergo complete regression, through a patient or expands slightly over time, and does
process of fibrosis, even in the absence of therapy. not regress.17 RICH is also fully formed at birth
The histopathologic composition of IHs varies but completely regresses within the first 6 months
with the age of the lesion. Early hemangiomas are to 1 year of life. NICH is characterized by the pres-
highly cellular and are characterized by plump ence of lobular collections of small, thin-walled
endothelial cells aligned to vascular spaces with vessels with large, often stellate, central lumina,
small inconspicuous lumina. As the lesions mature, separated by variable amounts of fibrous tissue
blood flow increases, endothelium flattens, and the richly supplied with normal and abnormal veins
lumina of the vessels enlarge and become more and arteries (Fig. 3).
obvious. During this interval, the vessels convey These vascular proliferations differ from IH in
a “cavernous” appearance that can be misinter- their growth patterns and lack of immunoreactivity
preted as a venous malformation. Regression is for the glucose transporter-1 protein (GLUT1),
portrayed as progressive interstitial fibrosis and which is a sensitive and specific marker for IH
adipose metaplasia, a process without known (see Fig. 1B). This protein is not detectable in the
stimulus.16 blood vessels of normal skin or in most other types
The other 2 types of IH include NICH and RICH. of vascular tumors but is highly expressed in endo-
NICH is a rare true vascular tumor that is fully thelia at sites of blood-tissue barriers, which
formed at birth, grows proportionally with the include the brain and placenta.

Fig. 3. (A) NICH. Low-power view shows a large vascular neoplasm showing an admixture of large dilated vessels
and lobular collections of small, thin-walled vessels (hematoxylin and eosin stain, original magnification A 20).
(B) Higher magnification of the lobular areas showing thin-walled vessels lined with uniform endothelial cells
(hematoxylin and eosin stain, original magnification B 40).
660 Sangüeza

MYOPERICYTOMAS for smooth muscle antigen (SMA), h-caldesmon,


and vimentin, and are sometimes weakly positive
Myopericytomas are a group of neoplasms that orig- for desmin.
inate from perivascular myoid cells (myopericytes),
which share features of both smooth muscle cells PERIVASCULAR EPITHELIOID CELL TUMORS
and glomus cells.18
Clinically, myopericytoma arises most com- Perivascular epithelioid cell tumors (PEComas) are
monly in the dermis or subcutaneous tissue, with a group of rare mesenchymal neoplasms that
a predilection for distal extremities; however, it show perivascular epithelioid differentiation. They
can affect other sites. They are more common in include angiomyolipoma, lymphangiomyomatosis,
younger adults, although other age groups also clear cell “sugar” tumor of the lung, and a group of
can be affected.19 The skin lesions may have rare, morphologically, and immunophenotypically
a purplish hue, whereas others present as white similar lesions arising at a variety of visceral and
nodules or fixed masses. Larger skin lesions can soft tissue sites. These tumors all share a distinc-
cause ulceration. The lesions tend to be generally tive cell type, the perivascular epithelioid cell.22
painless and slow growing. They are usually soli- PEComas show a marked female predominance
tary in adults, but can be multiple, especially in and seem to arise most commonly at visceral
children. Recurrence has been reported to occur (especially gastrointestinal and uterine), retroperi-
in 10% to 20% of patients. toneal, and abdominopelvic sites, with a subset
Histopathologically, myopericytomas show a occurring in somatic soft tissue and skin.23
broad range of histologic growth patterns. Malig- PEComas are composed of nests and sheets of
nant transformation may also occur. usually epithelioid but occasionally spindled cells
Myopericytomas are circumscribed but unen- with clear to granular eosinophilic cytoplasm and
capsulated tumors composed of relatively mono- a focal association with blood vessel walls. So
morphic oval- to spindle-shaped myoid-like cells far, most of the skin cases have presented as an
with cytoplasm that may be eosinophilic to ampho- ill-defined dermal neoplasm, with extension into
philic, and which share overlapping morphologic the subcutis composed of perivascularly arranged
features with myofibromas. Classically, 3 growth clear and pale eosinophilic tumor cells sur-
patterns have been described (classic myopericy- rounding numerous blood vessels with a lace-like
toma, myofibroma-like, and glomangiopericyto- pattern and slightly thickened walls (Fig. 5A, B).
ma), although Mentzel and colleagues20 recently Nearly all PEComas show immunoreactivity for
described other histologic patterns, including hy- both melanocytic (HMB-45 or melan-A, tyrosi-
pocellular, fibroma-like, angioleiomyoma-like, and nase, microphthalmia transcription factor, and
hemangiopericytoma-like neoplasms, and imma- NKIC3) (see Fig. 5C) and muscle markers, such
ture and cellular lesions (Fig. 4). Lesions with focal as SMA, muscle actin, muscle myosin, and calpo-
glomoid features and intravascular growth have nin, whereas desmin is expressed less frequently.
been also reported.21 Features of all these patterns In contrast, S100 protein and pancytokeratin are
may be present in a lesion, although one pattern usually absent. Some PEComas may behave in
may predominate. Most cases had low mitotic a malignant fashion.24,25 Although clear criteria
rates of less than 1/10 per high-power fields, and for malignancy are still lacking, the combination
necrosis was not usually seen. Immunohisto- of infiltrative growth, hypercellularity, cellular aty-
chemically, myopericytomas are uniformly positive pia, increased proliferative activity, and tumor

Fig. 4. Myopericytoma. (A) Monomorphic oval- to spindle-shaped myoid-like cells with an eosinophilic cytoplasm
and a centrally placed nuclei (hematoxylin and eosin stain, original magnification 20). (B) Detail of the cells
present in a myopericytoma (hematoxylin and eosin stain, original magnification B 40).
Update on Vascular Neoplasms 661

Fig. 5. PEComa. (A) Ill-defined neoplasm composed of nests of epithelioid and spindle cell with clear cytoplasm
(hematoxylin and eosin stain, original magnification 20). (B) Detail of the clear cells (hematoxylin and eosin
stain, original magnification 40). (C) Positive staining for HMB-45.

necrosis seem to be associated with a malignant round lumina and contain few erythrocytes
clinical course. (Fig. 6). The endothelial cells in many cases have
a hobnail appearance, but cellular atypia of endo-
ACQUIRED ELASTOTIC HEMANGIOMA thelial cells is absent and very few mitotic figures
are identified. Papillary projections are also present
Acquired elastotic hemangioma is a recently
in some cases. No hemosiderin deposits or ex-
described variant of cutaneous hemangioma.
travasated erythrocytes are seen. The connective
These lesions develop during adulthood on
tissue surrounding or intermingled with the newly
chronic sun-damaged skin on the extensor
formed capillaries show intense solar elastosis,
surface of the forearms or the lateral aspects of
which is seen as basophilic degeneration or amor-
the neck. It mainly affects middle-aged and elderly
phous basophilic granular material replacing the
women.26
normal superficial dermis.27,28
Acquired elastotic hemangioma presents as
Immunohistochemically, the neoplastic endo-
a slightly elevated, irregularly shaped, solitary
thelial cells of acquired elastotic hemangioma
lesion with violaceous coloration. The lesions are
strongly express CD31, CD34, and D2-40. A
usually well-demarcated plaques that range from
continuous rim of a-SMA–positive pericytes
2 to 5 cm in diameter. The lesions blanch under
surrounds most of the neoplastic vascular chan-
diascope pressure. Clinically, these lesions are
nels. Proliferating markers Ki-67 and MPM2 stain
confused with basal cell carcinoma, hemangioma,
only a few of the nuclei of the endothelial cells of
or Bowen disease. In most cases no evidence of
the newly formed blood vessels.
previous trauma is seen at the site of the lesion.
Histopathologically, acquired elastotic heman- VASCULAR PROLIFERATIONS IN RADIATED
gioma consists of a proliferation of capillaries SKIN
involving the superficial dermis in a band-like
arrangement parallel to the epidermis. A narrow Vascular proliferations are well recognized in
band of noninvolved papillary dermis separates previously irradiated areas of the skin. The nomen-
the newly formed capillaries from the normal or flat- clature in the literature is complex, compounded
tened epidermis. The capillary proliferation involves by terms such as lymphangiectases, benign lym-
the papillary and upper reticular dermis, but the phangiomatous papules, lymphangiomas, atypical
deep reticular dermis is characteristically spared. vascular lesions, and benign lymphangioendothe-
The neoformed capillaries show small cleft-like or lioma. The vascular lesions appear within the field
662 Sangüeza

Fig. 6. Acquired elastotic hemangioma. (A) Low-power view showing a proliferation of capillaries involving the
superficial dermis (hematoxylin and eosin stain, original magnification 20). (B) Detail of the capillaries, which
are lined by uniform endothelial cells. The stroma shows solar elastosis (hematoxylin and eosin stain, original
magnification 40).

of radiation. The interval between the application the lymphatic counterpart of telangiectases. They
of radiotherapy and the appearance of the cuta- result from acquired permanent dilatation of
neous lesions spans several years.29–31 lymphatic capillaries having appeared in areas of
Assessment of these lesions is problematic, the skin affected by obstruction or destruction of
because sometimes they can be difficult to the lymphatics. It is conjectured that they result
differentiate from well-differentiated angiosarco- from interference in the drainage of lymphatic
mas. In a few cases, lesions initially diagnosed vessels secondary to radiotherapy or surgery.
as benign vascular proliferations have evolved Benign lymphangiomatous papules and plaques,
into angiosarcomas. however, may also appear in the skin of the elderly
Clinically, benign vascular proliferations present without any evidence of primary lymphatic injury.31
as papules, small vesicles, and erythematous pla- Histologically, at low magnification, the lesions
ques (Fig. 7A). Lymphangiomatous papules and appear as well-circumscribed vascular prolifera-
plaques are the most common. These lesions are tions centered in the dermis, without extension

Fig. 7. Vascular proliferation in radiated skin. (A) Small translucent papules on the breast. (B) Low-power view
showing several irregularly dilated lymphatics present in the upper part of the dermis (hematoxylin and eosin
stain, original magnification 20). (C) Higher magnification showing prominent uniform endothelial cells (hema-
toxylin and eosin stain, original magnification 40).
Update on Vascular Neoplasms 663

into the subcutaneous fat. The epidermis is usually atypia. The nuclei of the endothelial cells are
spared. Most lesions show irregularly dilated monomorphous, have inconspicuous nucleoli,
lymphatic spaces that branch and anastomose and lack mitotic figures. Contrastingly, the endo-
within the superficial dermis. The vascular spaces thelial cells of an angiosarcoma may form stratified
most often devoid of content; however, in some layers that irregularly line anastomotic channels in
cases lymph is present (see Fig. 7B). They a manner and to a degree not seen in the atypical
are lined by a discontinuous single layer of endo- vascular proliferations of irradiated skin (see
thelial cells with flattened nuclei (see Fig. 7C). Fig. 2A, B). Recently, the use of the marker
Commonly, adjacent vascular channels lie “back- c-Myc has been advocated to differentiate these
to-back,” separated only by a thin layer of endo- processes. Angiosarcomas usually express the
thelial cells. Multiple papillary projections, covered marker c-Myc, which is absent in benign vascular
by a single layer of endothelium, project into the proliferations (see Fig. 2C).14,15
lumina of the dilated lymphatic. The stroma
consists of fibrillary collagen rich with spindle or NEW HISTOPATHOLOGIC VARIANTS OF AIDS-
stellate fibroblasts. Nodular infiltrates of lympho- RELATED KAPOSI SARCOMA
cytes with germinal centers are occasionally
present in the vicinity of the dilated vascular Kaposi sarcoma is an endothelial lymphatic prolif-
channels. Rarely, vascular proliferations are poorly eration induced by HHV8. Cutaneous lesions of
circumscribed and focally intermingled with irreg- Kaposi sarcoma evolve through different stages;
ular jagged vascular spaces that may permeate patch, plaque, and tumor. Histologically, lesions
the entire dermis. Endothelial cells line the latter of Kaposi sarcoma may show a marked variation
inconspicuously. Irregular slit-like vascular spaces and simulate other vascular lesions. Immunohisto-
may be between collagen bundles of the dermis chemically, lesions of Kaposi sarcoma stain posi-
together with tufts of endothelial cells that tive for the latent nuclear antigen-1 (LNA-1),
protrude into the lumens of the newly formed which identifies HHV8 infection in lesions of Kapo-
vessels. si sarcoma. This stain has proven valuable in iden-
The endothelial cells that line the vascular tifying different variants of Kaposi sarcoma.
spaces express immunoreactivity for CD31, but The spectrum of Kaposi sarcoma includes
they do so only focally or not at all for CD34. The anaplastic (pleomorphic) Kaposi sarcoma, lym-
lymphatic marker podoplanin (D2-40) is also phedematous variants (lymphangioma-like,
expressed by the neoplastic cells. Although a lymphangiectatic, and bullous Kaposi sarcoma),
minority of newly formed vessels show an and hyperkeratotic (verrucous), keloidal, micro-
attenuated muscle layer, external to the nodular, pyogenic granuloma–like, and intravas-
endothelial cells, that occasionally is immunoreac- cular Kaposi sarcoma. More recently, O’Donnell
tivity for a-SMA antibody, this marker is usually and colleagues32 described 5 new histologic vari-
nonreactive. The immunohistochemical profile ants of cutaneous Kaposi sarcoma, including glo-
substantiates the lymphatic nature of these newly meruloid, telangiectatic, ecchymotic, pigmented,
formed vessels. and Kaposi sarcoma with myoid nodules.
Some dermal vascular proliferations in irradi-
ated skin, such as the benign lymphangioendothe- Glomeruloid Kaposi Sarcoma
lioma or an atypical angiomatous proliferation,
Clinically, these lesions present as plaques of
may mimic the histopathologic appearance of
Kaposi sarcoma, and microscopically they show
the patch stage of Kaposi sarcoma or even
areas of plaque-stage Kaposi sarcoma with
a well-differentiated angiosarcoma. Contrastingly
a prominent vascular proliferation involving the
with the patch stage of Kaposi sarcoma, atypical
full thickness of the dermis accompanied by
benign vascular proliferations, as induced by radi-
conspicuous spindle-shaped cells, vascular clefts,
ation, do not contain erythrocytes, hemosiderin
and a mild lymphoplasmacytic inflammatory infil-
deposits, or stromal plasma cells. The striking tufts
trate. The deep dermis at the periphery of the
of endothelial cells and the intravascular papillary
lesion is punctuated by circumscribed, congested,
projections, evidenced in the vascular prolifera-
microscopic glomeruloid vascular structures
tions of irradiated skin, are absent in the lesions
resembling the glomeruli of the kidney.
of Kaposi sarcoma. Benign vascular proliferations
are negative for human herpesvirus 8 (HHV8).
Telangiectatic Kaposi Sarcoma
In contrast with well-differentiated angiosarco-
ma, atypical dermal vascular proliferations in irra- These lesions consist of dark, hemorrhagic
diated skin do not involve the subcutaneous lesions, and histologically they show large, con-
tissues. Distinctively, they each have cytologic gested, ectatic vascular spaces admixed with
664 Sangüeza

areas that show characteristic nodular-stage 7. Ohno S, Dohi S, Ohno Y, et al. Immunohistochemical
Kaposi sarcoma. detection of WT1 protein in endometrial cancer. Anti-
cancer Res 2009;29:1691–5.
8. Lawley LP, Cerimele F, Weiss SW, et al. Expression
Ecchymotic Kaposi Sarcoma
of Wilms tumor 1 gene distinguishes vascular mal-
This variant presents with widespread violaceous formations from proliferative endothelial lesions.
macules, patches, and plaques. Microscopically, Arch Dermatol 2005;141:1297–300.
the lesions exhibit an intradermal vascular prolifer- 9. Trindade F, Tellechea O, Torrelo A, et al. Wilms tumor 1
ation accompanied by extensive extravasation of expression in vascular neoplasms and vascular mal-
red blood cells. formations. Am J Dermatopathol 2011;33:569–72.
10. Heiskala M, Peterson PA, Yang Y. The roles of clau-
Kaposi Sarcoma with Myoid Nodules din superfamily proteins in paracellular transport.
Traffic 2001;2:92–8.
This variant can present with ulcerated nodules, 11. Moll R, Sievers E, Hammerling B, et al. Endothelial
which microscopically consist of nodules of and virgultar cell formations in the mammalian lymph
Kaposi sarcoma involving the deep dermis and node sinus: endothelial differentiation morphotypes
subcutaneous tissue. Within the nodules are characterized by a special kind of junction (com-
aggregates of bland spindle cells with abundant plexus adherens). Cell Tissue Res 2009;335:109–41.
eosinophilic cytoplasm and tapered nuclei remi- 12. Miettinen M, Sarlomo-Rikala M, Wang ZF. Claudin-5
niscent of smooth muscle. as an immunohistochemical marker for angiosarco-
ma and hemangioendotheliomas. Am J Surg Pathol
Pigmented Kaposi Sarcoma 2011;35:1848–56.
13. Miettinen M, Wang ZF, Paetau A, et al. ERG tran-
These lesions microscopically consist of nodules of scription factor as an immunohistochemical marker
Kaposi sarcoma containing spindle and epithelioid for vascular endothelial tumors and prostatic carci-
cells with marked cytologic atypia, increased noma. Am J Surg Pathol 2011;35:432–41.
mitotic figures, and prominent brown pigment 14. Manner J, Radlwimmer B, Hohenberger P, et al.
deposition limited to admixed dendritic-appearing MYC high level amplification is a distinctive feature
cells. This pigment was shown to be melanin (Fon- of angiosarcomas after irradiation or chronic lym-
tana-Masson–positive), and not hemosiderin (iron; phedema. Am J Pathol 2010;176:34–9.
Prussian blue–negative). 15. Guo T, Zhang L, Chang NE, et al. Consistent MYC
ad FLT4 gene amplification in radiation-induced an-
REFERENCES giosarcoma but not in other radiation-associated
atypical vascular lesions. Genes Chromosomes
1. Moore MA. Putting the neo into neoangiogenesis. Cancer 2011;50:25–33.
J Clin Invest 2002;109:313–5. 16. Requena I, Sangueza OP. Cutaneous vascular prolif-
2. Ellisen LW, Carlesso N, Cheng T, et al. The Wilms erations. II. Hyperplasia and benign neoplasms.
tumor suppressor WT1 directs stage-specific quies- J Am Acad Dermatol 1997;37:887–919.
cence and differentiation of human hematopoietic 17. Enroljas O, Mulliken JB, Boon LM, et al. Noninvolut-
progenitor cells. EMBO J 2001;20:1897–909. ing congenital hemangioma: a rare cutaneous
3. Nowakowska-Kopera A, Sacha T, Florek I, et al. vascular anomaly. Plast Reconstr Surg 2001;107:
Wilms’: tumor gene 1 expression analysis by real- 1647–54.
time quantitative polymerase chain reaction for 18. Dray MS, McCarthy SW, Palmer AA, et al. Myoperi-
monitoring of minimal residual disease in acute cytoma: a unifying term for a spectrum of tumours
leukaemia. Leuk Lymphoma 2009;50:1326–32. that show overlapping features with myofibroma. A
4. Gupta S, Joshi K, Wig JD, et al. High frequency of review of 14 cases. J Clin Pathol 2006;59(1):67–73.
loss of allelic integrity at Wilms’: tumor suppressor 19. Gengler C, Guillou L. Solitary fibrous tumour and
gene-1 locus in advanced breast tumors associated haemangiopericytoma: evolution of a concept.
with aggressiveness of the tumor. Indian J Cancer Histopathology 2006;48(1):63–74.
2009;46:303–10. 20. Mentzel T, Tos AP, Sapi Z, et al. Myopericytoma of
5. Perry BN, Cohen C, Govindarajan B, et al. Wilms skin and soft tissues: clinicopathologic and immuno-
tumor 1 expression present in most melanomas histochemical study of 54 cases. Am J Surg Pathol
but nearly absent in nevi. Arch Dermatol 2006;142: 2006 Jan;30(1):104–13.
1031–4. 21. McMenamin ME, Calonje E. Intravascular myoperi-
6. Oji Y, Kitamura Y, Kamino E, et al. WT1 IgG antibody cytoma. J Cutan Pathol 2002 Oct;29(9):557–61.
for early detection of nonsmall cell lung cancer and 22. Bonetti F, Pea M, Martignoni G, et al. Clear cell (‘sugar’)
as its prognostic factor. Int J Cancer 2009;125:381–7. tumor of the lung is a lesion strictly related to
Update on Vascular Neoplasms 665

angiomyolipoma—the concept of a family of lesions characteristics of 14 patients. J Cutan Pathol 2010;


characterized by the presence of the perivascular 37(4):460–4.
epithelioid cells (PEC). Pathology 1994;26:230–6. 28. Brenn T, Fletcher CD. Postradiation vascular prolifer-
23. Folpe AL, McKenney JK, Li Z, et al. Clear cell myo- ations: an increasing problem. Histopathology 2006;
melanocytic tumor of the thigh: report of a unique 48:106–14.
case. Am J Surg Pathol 2002;26:809–12. 29. Sener SF, Milos S, Feldman JL, et al. The spec-
24. Pea M, Bonetti F, Zamboni G, et al. Clear cell tumor and trum of vascular lesions in the mammary skin,
angiomyolipoma. Am J Surg Pathol 1991;15:199–202. including angiosarcoma, after breast conservation
25. Mentzel T, Reisshauer S, Rutten A, et al. Cutaneous treatment for breast cancer. J Am Coll Surg
clear cell myomelanocytic tumour: a new member of 2001;193:22–8.
the growing family of perivascular epithelioid cell 30. Diaz-Cascajo C, Borghi S, Weyers W, et al. Benign
tumours (PEComas). Clinicopathological and immu- lymphangiomatous papules of the skin following
nohistochemical analysis of seven cases. Histopa- radiotherapy: a report of five new cases and review
thology 2005;46:498–504. of the literature. Histopathology 1999;35:319–27.
26. Requena L, Kutzner H, Mentzel T. Acquired elastotic 31. Requena L, Kutzner H, Mentzel T, et al. Benign
hemangioma: a clinicopathologic variant of heman- vascular proliferations in irradiated skin. Am J Surg
gioma. J Am Acad Dermatol 2002;47(3):371–6. Pathol 2002;26:328–37.
27. Martorell-Calatayud A, Balmer N, Sanmartı́n O, et al. 32. O’Donnell PJ, Pantanowitz L, Grayson W. Unique
Definition of the features of acquired elastotic hem- histologic variants of cutaneous Kaposi sarcoma.
angioma reporting the clinical and histopathological Am J Dermatopathol 2010;32:244–50.

You might also like