You are on page 1of 21

Oncology (Research)

Veterinary Pathology
55(5) 806-826
The Author(s) 2013
A Comparative Review of Canine and Reprints and permission:
sagepub.com/journalsPermissions.nav
Human Rhabdomyosarcoma With Emphasis DOI: 10.1177/0300985813476069
vet.sagepub.com
on Classification and Pathogenesis

B. G. Caserto, DVM, Dipl ACVP1

Abstract
Rhabdomyosarcomas are a diverse group of malignant mesenchymal neoplasms exhibiting variable levels of differentiation toward
skeletal myocytes. Neoplastic cells may resemble relatively undifferentiated myoblasts, satellite cells, or more differentiated
elongated spindle cells and multicellular myotubes. In veterinary medicine, classification into subtypes and variants is based on an
outdated system derived from human pathology and is solely based on histologic characteristics. In contrast, classification of
human rhabdomyosarcoma is based on histologic, immunohistochemical, and molecular diagnostic techniques, and sub-
classification has clinical and prognostic relevance. Relevance of tumor subtyping has not been established in veterinary medicine.
Recent discoveries of components of the molecular pathogenesis and genomes of human rhabdomyosarcomas have led to new
diagnostic techniques and revisions of the human classification system. The current classification system in veterinary medicine is
reviewed in light of these changes. Diagnosis of rhabdomyosarcoma using histopathology, electron microscopy, and the clinical
aspects of human and canine rhabdomyosarcomas is compared. The clinical features and biologic behavior of canine rhabdo-
myosarcomas are compared with canine soft tissue sarcomas.

Keywords
dog, musculoskeletal, oncology, ultrastructure, immunohistochemistry

Rhabdomyosarcomas (RMS) are relatively rare neoplasms of on the phenotype of the variant.33,48,146,158 Diagnostic features
domestic animals that have a variety of gross morphologies, his- of skeletal muscle differentiation may not be evident at the light
tologic variations, and cellular phenotypic variations. Common microscopic level and may only be recognized by immunohis-
histologic subclasses include embryonal RMS, alveolar RMS, tochemistry or electron microscopy. This article reviews the
and botryoid RMS (also called botryoid embryonal RMS). Less diagnostic features, pathogenesis, and classification of rhabdo-
common subclasses include pleomorphic RMS. The relative rar- myosarcomas in dogs and compares them with their human
ity of these tumors makes diagnosis, clinical prognosis, and counterparts to facilitate the recognition of these tumors by
treatment difficult. In some cases, these neoplasms are lumped veterinary diagnostic pathologists.
into the category of high-grade soft tissue sarcomas89,158 but are
explicitly excluded from the common grading system of canine
soft tissue sarcomas.72 The low prevalence of these neoplasms Developmental Biology
in the literature may be due to a failure to diagnose them, since In the developing embryo, uncommitted mesodermal cells dif-
their extreme variation in phenotype, age of onset, and cellular ferentiate into myogenic progenitor cells under the control of
morphology makes diagnosis and classification difficult. The transcription factors such as PAX3, PAX7, and the Fox family
most aggressive of these neoplasms arise in juvenile dogs of transcription factors.2,136 Myogenic precursors then proliferate
younger than 2 years and include alveolar RMS and embryonal by asymmetric replication, forming immature myoblasts under
RMS. Histopathological findings of these primitive neoplasms the direction of myoblast determination protein 1 (MyoD1) and
typically consist of sheets or packets of small round cells with
a lymphoid appearance, or sheets of round cells with abundant
1
cytoplasm, and may be misdiagnosed as lymphomas or other Cornell University College of Veterinary Medicine, Department of Biomedical
primitive neoplasms. Other phenotypes arising in adult animals Sciences, Ithaca, NY, USA
consist of anaplastic cells with large nuclei, abundant cyto-
Corresponding Author:
plasm, and bizarre mitotic figures. These neoplasms may be B. G. Caserto, Cornell University College of Veterinary Medicine, Department
misdiagnosed as undifferentiated sarcoma, anaplastic sar- of Biomedical Sciences, Ithaca, NY, USA.
coma, or even a poorly differentiated sarcoma depending Email: bgc7@cornell.edu
Caserto 807

morphologies that resemble various developmental intermediates


and often have a myxomatous stroma as seen in developing mus-
cle.31,109,127 In the case of RMS, these cells range in morphology
from round, immature myoblast cells with variable amounts of
eosinophilic cytoplasm to multinucleated myotubular cells that
resemble the cellular phenotypes of developing skeletal muscle.
Embryonal muscle tumors consist of embryonal RMS, alveolar
RMS, and botryoid RMS; however, alveolar RMS is given its
own category due to its less favorable prognosis in humans.19,127
In humans, alveolar RMS and embryonal RMS occur predomi-
nantly in children younger than 15 years and is dubbed juvenile
or pediatric RMS.127 In dogs, this same association between age
of onset and histologic type has been recognized by several
Figure 1. Skeletal muscle development and myoblast fusion. Under authors61,65 but has not been universally accepted, as many cases
the direction of myogenin as well as PAX, myoblasts leave the cell of embryonal RMS occur in dogs older than 2 years.31 Pleo-
cycle and begin to fuse with each other or with preexisting myotubes. morphic RMS occurs mostly in adult people,19,109 and the 2 cases
Myotube fusion begins with 2 cells lining up, with their plasma mem- in dogs have been in a 3-year-old and a 14-year-old dog.8,31
branes contacting each other (a). Next, paired vesicular structures Attempts have been made to segregate neoplasms into
form along the apposed membrane surfaces (prefusion complexes) different categories for the purpose of making predictions of
(b). These prefusion complexes resolve into fusion plaques (c) that
biologic behavior based on histomorphologic, ultrastructural,
appear similar to adherens junctions and contain M-cadherin. The
fusion process requires calcium, which is further evidence that these or immunohistochemical features. The most recent, and widely
fusion plaques are members of the adherens junction family of cell- used, human classification system is the international classifica-
cell attachments. Once fusion plaques form, the plasma membrane tion system developed by Newton et al;104 however, similar yet
adjacent to them dissolves and the cytosols of the 2 cells are in direct differing systems exist and may be encountered periodically in
contact with each other (d). By some yet unexplained mechanism, the literature searches.109 According to the international classifica-
fusion plaques then disappear, and the walls of each adjoining cell are tion system, human RMS is subdivided into embryonal,
removed, fusing the 2 cells (e). The fused cells then elongate, the nuclei
botryoid, alveolar, and pleomorphic (anaplastic) subcate-
line up in rows to form myotubes, and the cells begin producing con-
tractile filaments and myoglobin (f). Additional myoblasts may fuse gories.31,65,109 With regard to human childhood RMS, there are
with myotubes in a similar fashion. Adapted from Doberstein et al40 3 main categories: embryonal, alveolar, and undifferentiated,
and Paululat et al.112 and these are distinguished from each other by histologic pattern
and cytogenetic studies.109,145 The undifferentiated juvenile
other transcription factors of the Myf family that includes myo- RMS has not been recognized in veterinary medicine. Classifi-
genin (myf4).165 During development and after injury, these cation of canine RMS closely parallels classification schemes in
cells proliferate, elongate, and fuse together, forming myotubes. human medicine (Table 1). Furthermore, embryonal and alveo-
Under the direction of myogenin, the myotubes begin forming lar RMS can be further subdivided into variants based on the
actin and myosin filaments organized into sarcomeres character- predominant cell morphology. Variants of embryonal RMS
istic of striated muscle.58 In both neoplastic and nonneoplastic include the rhabdomyoblastic, myotubular, and spindyloid var-
myogenic cells, MyoD1 is associated with cellular differentiation iants. Variants of alveolar RMS include the classic alveolar pat-
into myoblast cells with a high proliferative capacity, whereas tern and the solid alveolar pattern, which lacks fibrous septa.31
myogenin is associated with cell cycle arrest, cell fusion into mul- Such a precise classification system would potentially allow for
tinucleated myotubular cells, and formation of muscle fibers.58 subsequent study of clinical course of disease and response to
Mature skeletal muscle expresses very little of these transcription treatment. Detailed descriptions of histologic characteristics of
factors,19,60,67,121 and as normal skeletal muscle matures, it loses each category are below.
its replicative capacity but retains undifferentiated satellite cells at
the periphery of the myotube as stem cells, which are activated Embryonal Rhabdomyosarcoma
when the muscle is damaged. Figure 1 illustrates the major events
in skeletal muscle myoblast fusion. More extensive reviews of the Embryonal RMS includes 3 variants that are named because of their
subject have been published.2224,32,111 predominant cellular morphology.31,35,36,109 The myotubular var-
iant of embryonal RMS is dominated by multinucleated strap
cells forming myotubes with frequent cross-striations (often only
Diagnosis and Classification identified by histochemical staining with phosphotungstic acid
hematoxylin [PTAH] stain, which highlights striations as dark
Histologic Patterns blue/purple lines among paler blue/purple myofibers), whereas the
RMS in humans and dogs can resemble undifferentiated myo- rhabdomyoblastic variant is dominated by round to polygonal cells
blasts or early embryonic myotubes. The term embryonal is a with abundant eosinophilic cytoplasm (Fig. 2), with only rare
descriptive term for neoplasms exhibiting a range of cellular PTAH positive cross-striations.36,109 The rhabdomyoblastic variant
808 Veterinary Pathology 55(5)

Table 1. Classification of Rhabdomyosarcomas, With Variants and Relevant Clinical Aspects.

Subclass Variant Distinguishing Histologic Features Age Locations

Embryonal Cells exhibit different stages of development from Juvenile/adult Face, skull, masticatory muscle,
myoblast to myotubular, in a mucinous stroma oropharynx, trachea, axilla,
scapula, perirenal, tongue, flank,
leg, mammary gland, hard palate
Myotubular Myotube forms predominate
Rhabdomyoblastic Large myoblast cells with abundant cytoplasm
Spindle cell Streams of plump spindle cells Skull
Botryoid Characteristic submucosal location and gross Juvenile Urinary bladder, uterus
appearance; mixed round and myotubular cells
in mucinous stroma
Alveolar Juvenile Hip, maxilla, greater omentum,
uterus
Classic Fibrous bands divide small round cells into
clusters, loose aggregates
Solid Closely packed cells, thin fibrous septa
Pleomorphic Haphazardly arranged plump spindle cells with Adult Skeletal muscle
marked anisocytosis and anisokaryosis and
bizarre mitotic figures
The 3 major subclasses of rhabdomyosarcoma (RMS) are embryonal, alveolar, and pleomorphic. Botryoid RMS is often considered a separate subclass but is
technically a subset of embryonal RMS. In addition, embryonal RMS has 3 variants that are differentiated by the predominant cell morphology. Myotubular
embryonal RMS is dominated by multinucleated and elongated tubular cells. Rhabdomyoblastic embryonal RMS is dominated by large round cells with
abundant cytoplasm. Spindle cell embryonal RMS is dominated by fusiform, elongated cells arranged in streams. Alveolar RMS is divided into the classic
alveolar pattern with distinct fibrous septa lined by neoplastic round cells with a central open space. The solid variant of alveolar RMS lacks the clear space
and often lacks obvious fibrous septa. The pleomorphic RMS most often occurs in adult animals and is composed of haphazard spindle cells with large
myoblast cells often containing large pleomorphic nuclei and bizarre mitotic figures. Adapted from Parham.109

is included along with alveolar RMS and undifferentiated RMS in month-old Boxer dog. This mass was composed of interlacing
the general category of small round cell neoplasms of childhood fascicles of plump spindle cells.35 The spindyloid embryonal
sarcomas.5,149 These 3 categories are more commonly diagnosed RMS, as the name implies, is composed of thin spindyloid myo-
with the aid of molecular diagnostic techniques that are unavailable blast cells forming bundles within a myxoid stroma. Although
in veterinary medicine.145 There is no evidence that classification limited case reports exist, these neoplasms in humans typically
into the myotubular or rhabdomyoblastic variants has any bearing have a better prognosis than any other variant of RMS.94
on prognosis or clinical course in human or canine medicine.36,109

Botryoid Embryonal RMS Alveolar RMS


Botryoid rhabdomyosarcomas are tumors that have a character- Alveolar RMS is the second RMS in the small round cell cate-
istic gross appearance and anatomic location. The term botryoid gory of childhood RMS. It is characterized by small, poorly dif-
refers to the grape-like masses protruding from the mucosa of ferentiated, round cells with scant cytoplasm, arranged into
the urinary bladder, which is the most common location. Like packets of cells separated from each other by fibrous connec-
embryonal RMS, these tumors also recapitulate skeletal muscle tive tissue, forming alveolar-like patterns (Figs. 4, 5).149
embryogenesis and therefore contain undifferentiated myoblast Alveolar RMS is further subdivided into the classic alveolar
cells as well as multinucleated myotube cells within a myxoma- pattern, which is characterized by extensive sloughing of neo-
tous stroma (Fig. 3).31,127 Histologically, botryoid RMS of the plastic cells into a central open space with additional neoplas-
urinary bladder is located in the submucosa, and neoplastic cells tic cells lining the fibrous septa. The second subtype of
are separated from the mucosal epithelium by a cambium alveolar RMS is the solid variant, which is characterized by
(ie, transitional) layer of connective tissue. Because of their sheets of small round cells closely packed together (Fig. 6).5,149
distinct anatomic location and histologic characteristics, these This variant in dogs may have thin fibrous septa dividing nests
neoplasms have been placed into a distinct subcategory of of round cells similar to the classic neuroendocrine pattern31
embryonal RMS in human and veterinary medicine.109 but is most often lacking this histologic distinction, making
differentiation between solid alveolar RMS and rhabdomyo-
blastic embryonal RMS very difficult.36,109 Differentiation
Spindyloid Embryonal RMS between alveolar RMS and embryonal RMS is important in
The spindyloid variant of embryonal RMS is a relatively new human medicine because alveolar RMS of any variant is more
category. This type of tumor has been reported only once in locally aggressive and has a higher metastatic rate, carrying a
veterinary medicine as a mass arising from the skull of an 11- poorer prognosis than embryonal RMS.36,60,116,127 Currently,
Caserto 809

Figure 2. Dog, embryonal rhabdomyosarcoma (RMS), rhabdomyoblastic variant: neoplastic cells are uniform in size with abundant eosino-
philic cytoplasm with variable amounts of cytoplasmic glycogen or lipid vacuoles. Hematoxylin and eosin (HE). Figure 3. Dog, botryoid RMS:
this is the prototypical embryonal tumor, characterized by both uninucleate myoblastic cells similar to the rhabdomyoblastic embryonal
RMS, but also has a large proportion of myotubular cells or strap cells, which have single or multiple nucleoli. This is the typical histologic
appearance of RMS of the urinary bladder or urogenital tracts. HE. Figure 4. Dog, classic alveolar RMS: neoplastic cells are subdivided into
packets separated by fibrous septa in an alveolar pattern. Neoplastic cells vary in size and amount of cytoplasm, occasionally slough into the
lumen, and occasionally have multiple nuclei. HE. Figure 5. Dog, classic alveolar RMS: an additional variant of the classic alveolar pattern with
810 Veterinary Pathology 55(5)

there are insufficient data to draw the same prognostic conclu-


sion in veterinary medicine.

Pleomorphic Rhabdomyosarcoma
Pleomorphic RMS occurs less frequently than alveolar and
embryonal RMS in humans. It is found more often in adult
humans and animals and arises almost exclusively within skeletal
muscle. Histologically, it is composed exclusively of haphazardly
arranged spindle cells with rare multinucleated cells present and
is recognized by the appearance of bizarre or multipolar mitotic
figures and a high degree of cellular and nuclear pleomorphism
(Fig. 7).35,109 This diagnosis should be used only when there are
no areas of embryonal or alveolar morphology within the
tumor.31 This tumor undoubtedly causes confusion for patholo-
gists, as histologic evidence of skeletal muscle differentiation can
be rare. Without the aid of immunohistochemistry, some of these
neoplasms may be diagnosed as anaplastic sarcomas.

Ultrastructural Diagnosis of
Rhabdomyosarcoma
Transmission electron microscopy is the gold-standard diagnos-
tic test for rhabdomyosarcomas in humans and dogs, but
attempts to use ultrastructural characteristics to aid in classifica-
tion have been relatively unrewarding in human medicine, with
both alveolar and embryonal RMS exhibiting a variety of sub- Figure 8. Dog, skeletal muscle, rhabdomyosarcoma (RMS). Transmis-
cellular structures.133 Common to all human RMS are Z-lines, sion electron micrograph showing a small crescent of the nucleus on
large numbers of mitochondria, myofilament tangles, and the right, a mitochondrion (arrowhead), and a developing muscle fiber
myosin-ribosome complexes (Fig. 8). Several studies have indi- consisting of numerous polyribosomes with myosin thick filaments
(short arrows) and an M-line (block arrow). Z-lines were poorly
cated the presence of basement membranes in both alveolar and
formed and scattered haphazardly (30 k; scale bar = 500 nm). Inset
embryonal tumors in human RMS.95,142 Desmosome-like struc- (upper left corner) shows a closeup of the polyribosomes and myosin
tures may be a unique feature of alveolar RMS,27 but since pub- filaments (40 k).
lication of this study, there has not been further investigation
into these unique structures. identified actin and myosin filaments, Z-lines, and A- and
I-bands in both alveolar and embryonal tumors.129,133,139 In
Cytoskeletal Proteins some cases of embryonal RMS, myofilament tangles are
observed, indicating poor formation and disorganization.133
In the absence of myotubes, strap cells, and cross-striations,
small round cell neoplasms must be differentiated from lympho-
mas and primitive neuroectodermal tumors or other embryonal
Adhesion Structures in Human RMS
neoplasms. Early studies of human RMS identified many ultra- Some alveolar RMS contains desmosome-like structures
structural features unique to skeletal muscle that can aid diagno- between adjacent neoplastic cells lining the fibrous septa.27
sis of rhabdomyosarcomas. Identification of sarcomeric Churg and Ringus27 described these structures and hypothe-
structures such as unambiguous Z-lines within I-bands, sized that they are remnants from the embryological ectoderm
A-bands with H- and M-bands, and other structures such as free before the epithelial to mesenchymal transition. To complicate
ribosomes closely associated with myofilaments, abundant gly- matters, these structures resemble skeletal muscle fusion pla-
cogen granules, external lamina, prominent nucleoli, and pleo- ques found in Drosophila and mouse myoblast cultures.22,23,40
morphic mitochondria can all be used to support a diagnosis Fusion plaques appear similar to desmosomes and adherens
of rhabdomyosarcoma.17 Electron microscopy studies of both junctions by electron microscopy. They are transient structures,
alveolar and embryonal RMS in humans have consistently like adherens junctions, and contain M-cadherin rather than

Figure 5. (continued) larger cells but clearly defined fibrous septa between groups of cells. HE. Figure 6. Dog, solid alveolar RMS:
this variant consists of dense sheets of small round cells with small amounts of cytoplasm and hyperchromatic nuclei. Fibrous septa are
not present. HE. Figure 7. Dog, pleomorphic RMS: neoplastic cells vary in size and shape, as well as nuclear size and shape, with frequent
karyomegalic cells and bizarre mitotic figures. HE.
Caserto 811

desmosomal cadherins or desmoglein.10,22,46,53 Myoblasts of RMS in both humans and dogs95,129,166,167 (Figs. 9, 10), but
Drosophila spp that lack the gene Rolling stone (Rost) fail to staining can be patchy and sparse, and other neoplastic and nor-
complete myoblast fusion and accumulate fusion plaques mal tissue can express desmin.71,121 Despite the variability in
resembling the junctional complexes described in human alveo- staining pattern and intensity, it is a useful marker of myogenic
lar RMS.27,32,40 It is unknown whether these junctional com- differentiation in poorly differentiated RMS such as alveolar
plexes in human RMS are arrested fusion plaques since they and embryonal RMS. Because of desmins low specificity and
have not been studied extensively. It is unknown whether high degree of variability of staining, newer methods of identi-
human embryonal RMS has similar desmosome-like struc- fying undifferentiated myoblast cells have been developed,
tures, and it is unclear whether these junctional complexes are including using MyoD1 and myogenin, 2 nuclear transcription
related to the pathogenesis of RMS in humans. RMS cells of factors.
C2C12 mouse myoblast cultures have reduced M-cadherin
expression and elevated R-cadherin expression, both of which
can activate Rac1 GTPase,22,70 which has been implicated in
Actin, Myosin, Sarcomeric Actin, and Myoglobin
promoting myoblast proliferation and myoblast fusion, and yet Muscle actin (ms actin or HHF35) is an antibody that can iden-
later acts as an inhibitor of myogenesis induction.22 Although tify -actin isoforms in both human and canine skeletal, cardiac
not proven, it is possible that adhesion structures are involved and smooth muscle, and myofibroblasts, myointimal cells, and
in the failure of many RMS types to fuse into myotubes and reactive mesothelial cells49,150 with an equivalent specificity
therefore fail to histologically resemble skeletal muscle. to desmin.126 Identification of desmin or ms actin in myogenic
sarcomas cannot confirm a diagnosis of RMS by themselves,
Canine RMS but neoplasms with negative immunostaining for smooth mus-
cle actin can help rule out leiomyosarcomas and malignant myo-
There are few electron microscopic studies of canine RMS and no fibroblastic tumors and make a reasonably certain diagnosis of
descriptions of differences between histologic variants. Actin and RMS.51,133
myosin filaments, polyribosomes, Z-bands, and mitochondria More specific to skeletal muscle is expression of proteins
(Fig. 8) are prominent in botryoid RMS, laryngeal muscle tumors involved in sarcomere construction such as the -actin isoform
(both rhabdomyomas and RMS), and RMS that is not further found in sarcomeres, identified by the antibody sarcomeric actin
classified (RMS not otherwise specified [NOS]).3,76,93,113 (src actin).31 As skeletal myocytes differentiate further, they begin
to accumulate sarcomeric actin, followed by myosin or fast
myosin, and finally myoglobin, which is first seen in large
Immunohistochemical Diagnosis of RMS amounts following myoblast fusion to form myotubes or strap
Immunohistochemistry (IHC) is useful in differentiating rhab- cells.9,31,36,60,83,101,121,166,167 Mature skeletal muscle typically has
domyosarcomas from other mesenchymal tumors or embryonal little vimentin expression, whereas immature skeletal muscle and
tumors. A potential diagnostic pitfall is the fact that all undifferentiated rhabdomyosarcomas typically have rare expres-
myogenic markers used to diagnose RMS will also be present sion of myoglobin and myosin.36 See Table 2 for a brief summary
in developing and regenerating skeletal muscle,19,168 so it is the of common IHC markers used in canine rhabdomyosarcomas.
job of the diagnostic pathologist to first differentiate satellite cell In humans and dogs, rhabdomyosarcomas are consistently
proliferation and differentiation from a neoplastic proliferation. immunohistochemically positive using antibodies for vimentin,
Diagnosis of RMS in humans and dogs is based on detection of desmin (Fig. 10), muscle actin, and src actin (Fig. 11),133 but
1 or more muscle-specific markers and absence of smooth mus- because of their lack of cellular differentiation, myoglobin stain-
cle markers. The presence of these markers depends to some ing is variable.3,31,61,65,109,167 In dogs, embryonal and botryoid
extent on the degree of differentiation of the neoplastic cells but RMS are frequently positive for myoglobin compared with
is also subject to variation due to altered expression of myogenic alveolar RMS (Table 2).67
regulating factors.26,34,97,133
Myo D1 and Myogenin
Vimentin and Desmin MyoD1 and myogenin are early embryological transcription fac-
Identification of skeletal muscle fibers is largely based on tors involved in the differentiation of mesoderm cells into myo-
specific expression of cytoskeletal proteins such as intermediate blast cells, myoblast cell proliferation, and myoblast
filaments or contractile proteins. Skeletal muscle cells are char- differentiation into multinucleated myotubes. MyoD1 expression
acterized by expression of different cytoskeletal proteins at pro- is associated with immature rhabdomyoblasts that have a higher
gressive stages of development.31 Like all mesenchymal cells, proliferative capacity, whereas myogenin is expressed during
myoblast cells first express the intermediate filament vimentin. myoblast fusion and is associated with exit from the cell cycle.
Later, during differentiation, they express another intermediate Rhabdomyosarcomas that are composed of relatively undifferen-
filamentdesmin, which is common to skeletal muscle, cardiac tiated cells should be expected to express less desmin, actin, myo-
muscle, smooth muscle, and myofibroblasts.9,97 Desmin has sin, and myoglobin and more MyoD1 and myogenin. Several
been proven a useful marker even in relatively undifferentiated studies have confirmed the specificity of myogenin and MyoD1
812 Veterinary Pathology 55(5)

Figure 9. Dog, embryonal rhabdomyosarcoma (RMS). This is a biphasic embryonal RMS with round myoblast cells in the upper left corner and a
more spindyloid population in the lower right with a very myxomatous stroma. Hematoxylin and eosin (HE). Figure 10. Dog, embryonal RMS,
desmin immunohistochemistry. Neoplastic rhabdomyoblasts (a) and spindle cells (b) have diffuse positive cytoplasmic immunoreactivity for anti-
desmin antibodies (3,3-diaminobenzidine [DAB]/hematoxylin). Figure 11. Dog, embryonal RMS. Sarcomeric actin immunohistochemistry.
Scattered neoplastic cells have positive cytoplasmic immunoreactivity to antisarcomeric actin antibodies (DAB/hematoxylin) Figure 12. Dog,
embryonal RMS. Myogenin immunohistochemistry. Neoplastic cells have frequent positive nuclear immunoreactivity for antimyogenin antibo-
dies (DAB/hematoxylin). Figure 13. Dog, alveolar RMS. MyoD1 immunohistochemistry. Neoplastic cells have moderate multifocal nuclear
immunoreactivity (DAB/hematoxylin).

as specific and sensitive markers of undifferentiated RMS in RMS.127,146 Although immunohistochemical identification of
humans.19,60,101,121,127,165 Furthermore, several studies have con- myogenin and MyoD1 is a useful tool in human medicine, more
cluded that differential expression of myogenin can be used to specific tools have been developed that are in the process of sup-
subclassify human RMS into alveolar or embryonal RMS (Table planting these methods.
3).19,60,109,165 Human alveolar RMS has been found by several Precise correlations cannot yet be made between human and
investigators to diffusely express myogenin in the majority of canine RMS using myogenin or MyoD1 staining. One study of
nuclei of neoplastic cells by immunohistochemistry, whereas canine botryoid RMS found myogenin IHC stained elongated
nuclear MyoD1 expression, although consistently present, is pat- strap cells preferentially, whereas immunohistochemical detec-
chy and less intense. Conversely, human embryonal RMS has tion of MyoD1 was limited to the nuclei of small, round myo-
few cells expressing either myogenin or MyoD1.19,39,60,109 The blast cells.67 In published case reports, both myogenin and
different IHC staining patterns between myogenin and MyoD1 MyoD1 have been used to diagnose canine RMS, but there have
were used to differentiate human alveolar RMS from embryonal been no differences in the staining pattern or intensity
Caserto 813

Table 2. Common Immunohistochemistry Antibodies in Veterinary Medicine.

Canine, All Ages Vimentin Desmin Ms Actin Src Actin Myoglobin Myogenin MyoD1 Smooth Muscle Actin

Alveolar 5/5 6/6 1/1 2/2 1/2 1/1 1/1 0/3


Embryonal 10/10 15/15 5/5 7/7 9/10 1/1 2/2 1/5
Botryoid 9/9 13/13 3/3 5/5 5/8 2/2 2/2 0/4
Pleomorphic 2/2 2/2 1/1 1/1 1/1 NA NA 0/2
RMS NOS 5/5 4/5 2/2 NA 2/2 NA 1/1 0/2

Numbers indicate the number of cases with positive staining (numerator) over the number of cases stained (denominator). Vimentin, desmin, muscle actin (ms
actin), and sarcomeric actin (src actin) are consistently positive in canine rhabdomyosarcoma (RMS). Myoglobin staining is inconsistent but positive in many cases
of embryonal and botryoid RMS and is rarely used in alveolar RMS. Positive staining with desmin, ms actin, src actin, or myoglobin plus negative smooth muscle
actin staining is diagnostic for rhabdomyosarcoma. Rarely is canine RMS positive for smooth muscle actin, making myogenin and MyoD1 necessary for diagnosis.
NA, not applicable; NOS, not otherwise specified.31,109

Table 3. Comparison of Myogenin and MyoD1 Staining in Different Categories of RMS Between Human and Canine Neoplasms.

Human Canine

Myogenin MyoD1 Myogenin MyoD1

Alveolar + Uniform + Sparse + +


Embryonal + Multifocal + Sparse + +
Botryoid NA NA Myotube cells Round cells
Alveolar rhabdomyosarcoma (RMS) tends to have uniform and more abundant myogenin staining compared with embryonal RMS. MyoD1 staining is present is both
but sparse. In dogs, both alveolar and embryonal RMS stains with myogenin and MyoD1, but staining can be sparse, and there is no proven difference between the two.
In canine botryoid RMS, myogenin stained the myotube cells preferentially, and MyoD1 stained the round cells preferentially.60,68,121,127 NA, not applicable.

described.21,25,35,62,67,100,140 Myogenin staining can also be embryonal RMS develops from muscle progenitor cells, or ske-
seen in large round rhabdomyoblastic cells of embryonal RMS letal muscle stem cells present within the pool of satellite cells,
in dogs (Fig. 12). These cells often contain large amounts of because embryonal RMS in mice contains a high number of
eosinophilic cytoplasm but may be mixed with smaller cells that muscle stem cells.58 It has been suggested that the pleomorphic
resemble undifferentiated myoblast cells. In dogs, MyoD1 RMS subclass is derived from adult skeletal muscle, but
immunostaining of embryonal RMS is rare and sometimes non- whether they are derived from satellite cells or other cell popu-
existent (B. G. Caserto, unpublished data), but MyoD1 staining lations in the adult muscle was not mentioned.125
can be observed in alveolar RMS more frequently (Fig. 13). At
present, there is insufficient information to conclude whether
myogenin or MyoD1 is useful in subclassifying RMS into Cytogenetics of RMS in Domestic Animals
alveolar or embryonal categories. Very little is known about the cytogenetics of RMS in dogs and
other domestic animals when compared with knowledge of
human RMS. Although a tumor cell line was established from
Histogenesis and Cytogenetics a pleomorphic RMS from a 14-year-old mixed breed dog, no
Many different hypotheses have been proposed regarding the cytogenetic abnormalities were reported.8 One report described
histogenesis of RMS in all species. RMS of any species may single or multiple RMS developing in 25 of 100 piglets from
develop from pluripotent stem cells from the primitive urogen- multiple litters over a 6-month period in the Netherlands,
ital ridge remnants (in the case of botryoid RMS) or from suggesting an inherited component.157 van der Loop and
mesenchymal progenitor cells invading the Mllerian and Wolf- colleagues established a cell culture from a pleomorphic RMS
fian ducts, either locally or through the circulation originating in a 3.5-week-old female piglet and described a deletion of the
from the bone marrow stroma.58,91 The presence of mesenchy- long arm of the X chromosome (Xq24-qter) in these cells.154
mal stem cells capable of myogenic differentiation in the bone
marrow and elsewhere in the body may explain why RMS can
be found in tissue with no skeletal muscle.58 Alternatively, RMS
Cytogenetics of Human RMS
can develop from early myogenic progenitor cells remaining Human embryonal RMS has been associated with abnormalities
from embryonic development or from myoblast cells within the of chromosome 11p15, leading to loss of maternal information
pool of satellite cells. Using a mouse model, Nitzki et al105 sug- and duplication of paternal genetic information (loss of hetero-
gest that early mesenchymal stem cells (embryonic cells uncom- zygosity or LOH). The IGF-II gene is normally expressed by
mitted to myogenesis) are the major source of embryonal RMS. the paternal allele only, so the loss of the maternal allele in
This view is contradicted by others who have suggested that these tumors with a duplication of the paternal allele leads to
814 Veterinary Pathology 55(5)

a 2-fold increase of IGF-II, leading to uncontrolled cell cycle Studies of the mRNA transcripts and in vitro studies of trans-
progression and cell proliferation.36 Embryonal RMS can also fected cultured myoblasts have discovered increased activity of
have DNA contents ranging between diploid and hyperdiploid, MDM2, CDK4, and cyclin D.163,169 A recent study of in vitro
with diploid tumors having a worse prognosis than hyperdiploid mouse mesenchymal stem cells demonstrated the ability of the
tumors.36 PAX-FKHR translocation to produce alveolar RMS when
Human alveolar RMS has a characteristic translocation coupled with loss of RB and p53 tumor suppressor gene func-
between the long arm of chromosome 2 and the long arm of tion and constitutive H-RAS activation.120 In normal cells,
chromosome 13: [t(2;13)(q35;q14)]. This mutation fuses the CDK4 and cyclin D form a complex that begins a cells pro-
PAX3 gene (Paired box family) with the FKHR gene (Forkhead gression through the G1 phase of the cell cycle. Uncontrolled
box family). A variant of the mutation, t(1;13)(p36;q14), fuses accumulation of CDK4 and cyclin D lead to uncontrolled cell
the PAX7 gene from chromosome 1 with the FKHR gene and cycle progression through phosphorylation of RB. RB is a
leads to more aggressive neoplasms in younger children. These tumor suppressor protein normally present in the nucleus
mutations are detectable in 70% to 90% of histologically bound to E2F, thereby preventing transcription of mRNA
diagnosed alveolar RMS.2,36,60,127,136 needed for cyclin production and cell cycle advancement
Polymerase chain reaction (PCR) identification of mRNA beyond the G1 phase. When RB is phosphorylated, E2F is
transcripts of fused genes (PAX3 or PAX7-FKHR) can identify freed, leading to transcription of cyclins needed for cell cycle
and distinguish human alveolar RMS from embryonal progression. If H-RAS is constitutively active as in this mouse
RMS.136,160 This is a rapid diagnostic test that can detect the model,120 RAS continually sends positive signals for cell cycle
mutation in cases identified by histology as alveolar RMS. In progression. These continuous growth signals, coupled with
addition, studies of alveolar RMS lacking this mutation the inability to halt the cell cycle or undergo apoptosis, leads
(fusion-negative alveolar RMS) found that the biologic beha- to neoplastic growth.169,170
vior was similar to embryonal RMS and, like embryonal RMS, P53 is an important regulator of cell senescence and apopto-
showed high levels of expression of genes from chromosome 8. sis. It senses DNA damage and can halt cell cycle progression,
The biological behavior of fusion-negative alveolar RMS is dis- leading to either senescence or apoptosis. MDM2 is responsible
tinct from fusion-positive alveolar RMS, indicating that histolo- for inactivating p53, thereby possibly preventing apoptosis in
gic pattern is not as specific a predictor of biologic behavior as these cells.163,170 Inactivation of P53 coupled with uncontrolled
cytogenetic studies.160 It was concluded that the presence of the cell cycle advancement results in neoplastic growth. There are
PAX-FKHR mutation is a significant factor in the aggressive more alterations in cell senescence and growth found in human
phenotype of human alveolar RMS. Using this method to help and mouse models of RMS. Mirk expression is elevated in
classify human RMS is especially helpful in cases where there human RMS. Normal functions of Mirk range from cell senes-
is lack of distinct histologic features.160 It is becoming more cence in the G0 phase to responses to cellular injury and reduc-
common in human medicine to test for these mutations using tion in apoptosis. Increased expression of Mirk in RMS is
reverse transcription PCR (RT-PCR) and making the diagnosis thought to result in reduced apoptosis and increased survival
in conjunction with a biopsy.145,160 of all RMS variants.90 Alterations in other tumor suppressors
have also been found. Micro-RNAs act as tumor suppressors
and are downregulated in human RMS.124
Furthermore, other studies of transfected cultured pediatric
Proposed Molecular Pathogenesis of RMS RMS and mouse mesenchymal stem cells have demonstrated
Human alveolar RMS and embryonal RMS are thought to arise that the PAX-FKHR mutation is necessary for myogenic differ-
by a multistep process leading to loss of tumor suppressor genes entiation in alveolar RMS but in itself is not sufficient to pro-
and genes affecting the regulation of apoptosis and cellular duce a neoplasm without additional mutations. Consequently,
senescence.79 Recent molecular studies have suggested that multiple mutations over several generations are needed to
alveolar RMS represents an arrested stage of development in produce a sarcoma with the phenotype of alveolar RMS.58,165
undifferentiated myoblast cells.165 The cause of the arrested Further study is needed to better clarify the origin of RMS in
stage of development could be related to the PAX-FKHR muta- human and veterinary medicine.
tion; under certain circumstances, increased expression of PAX Currently, there are so few cases of canine RMS that there is
inhibits myogenic differentiation in mesoderm cells.43 The no information available regarding molecular pathogenesis of
fusion of the PAX and FKHR genes leads to uncontrolled cell this neoplasm. Although genetic mutations may prove useful
growth signals, cell cycle progression, failure of tumor suppres- in future diagnosis of canine RMS, the relative rarity of this neo-
sor gene function, and myogenic differentiation. Fusion of PAX- plasm and the difficulty in making a diagnosis make genomic
FKHR causes increased levels of MyoD1, resulting in cell pro- analysis less useful as a diagnostic tool. At present, immunohis-
liferation and also increased levels of myogenin, leading to tochemistry is the most feasible adjunct to standard histopathol-
myogenic differentiation. Increased levels of IGF-II also result ogy for diagnostic purposes. The diagnostic utility of myogenin
in cell proliferation.163 Human alveolar RMS has been associ- and MyoD1 immunohistochemistry can be investigated from
ated with increased expression of c-Met receptor and CXCR4, archived cases and be readily applied to diagnostic laboratories
both of which enhance cell growth and cell cycle progression.87 throughout the country.
Caserto 815

Pathogenesis of Metastatic Disease in medicine, embryonal RMS and alveolar RMS are by far the most
Human RMS common tumors in children.78
Twelve laryngeal rhabdomyomas/rhabdomyosarcomas have
Mutations in the oncogene FGFR4 have been found in 7.5% of been reported in dogs, all but one in dogs older than 2 years. It
human alveolar and embryonal RMS.80 This mutation is is generally believed that laryngeal rhabdomyomas/rhabdomyo-
associated with advanced stage of disease in alveolar RMS in partic- sarcomas are a distinct clinical entity of dogs, being locally inva-
ular, whose uncontrolled expression leads to cell cycle progression sive yet rarely metastatic.13,15,30,31,56,77,84,92,107,164,166 Complete
and has a role in metastasis that is not well defined.143 Interleukin-4 excision is often difficult due to local invasion, and recurrence
(IL-4) helps recruit myoblasts to form myotubes in normal mamma- of these tumors often leads to euthanasia. Histologically, these
lian myogenesis and has been associated with childhood and mouse neoplasms contain large cells with granular eosinophilic cyto-
RMS differentiation, mitogenesis, and metastasis.59 plasm containing large amounts of periodic acid-Schiff (PAS)
Immunohistochemical identification of receptors down- positive glycogen and mitochondria. Although some have been
stream of Hedgehog signaling such as Smoothened (SMO) and diagnosed as oncocytomas based on ultrastructural findings of
glioblastoma transcription factor 2 (GLI2) are common in large numbers of mitochondria, laryngeal rhabdomyomas can
human RMS and are the result of increased Hedgehog signaling be distinguished from oncocytomas by positive staining for myo-
in these tumors. In fact, inhibition of SMO by cyclopamine globin and desmin or by the ultrastructural presence of myofibers
slows the growth of RMS cell lines in vitro. Inhibition of GLI1 in addition to glycogen and mitochondria.76,92,161
by GANT61 (inhibitor of Hedgehog signaling by interfering in Cutaneous rhabdomyomas are rarely reported. Two cases of
DNA binding with GLI1) also slowed growth of in vitro RMS cutaneous rhabdomyoma were reported by Copper and Valen-
cell lines.63,148 Inhibitors of Hedgehog signaling have been sug- tine,31 one occurring in the skin and subcutis of a leg and the
gested as a therapeutic method for human gliomas and may be other in the skin of a foot in 11-year-old and 14-year-old dogs,
useful in the treatment of RMS as well.29 respectively. Cutaneous RMS can occur in the dermis or subcu-
In patients with distant metastasis, there is high expression of tis of humans.86 These occur mostly in adults and are diagnosed
FOXF1 (Forkhead family of transcription factors) and LMO4 as pleomorphic RMS. Those that occur in children are usually
(LIM domain transcription factor), which are thought to aid in embryonal and alveolar RMS. In 1 report, 6 of 12 cases metas-
tumor cell migration.4 Overexpression of MET by some tumors tasized.86 Primary cutaneous RMS can occur in dogs, and some
may provide RMS cells with the same property as embryonal cutaneous RMS that occurs in the dermis is likely misdiagnosed
myoblasts to migrate into surrounding connective tissues.36 as anaplastic or poorly differentiated soft tissue sarcoma if
Snail1 is highly expressed in alveolar RMS, E-cadherin is immunohistochemical studies are not pursued. Despite being
downregulated, and expression of MMP2 and MMP9 is upregu- excluded from the category of soft tissue sarcomas by some
lated in alveolar RMS.117 Because of Snail1s known roles in investigators,37,72,130 others have included them in their soft tis-
the metastasis of other tumors, it is presumed, yet unproven, sue sarcoma category on the basis of similar behavior.156 Some
to aid in metastasis of RMS. The role of E-cadherin and MMP2 subcutaneous masses undoubtedly arise from muscles of the
in the pathogenesis of RMS and metastasis is less well known. appendicular skeleton but invade superficially, making determi-
Loss of E-cadherin is thought to reduce sensitivity of cell nation of origin difficult from a biopsy.
growth to external factors such as contact with other cells or the Cardiac muscle tumors are rare in dogs, but the majority of those
extracellular matrix. Increased amounts of matrix metallopro- reported and available through PubMed are RMS (6/12),1,6,52,69,114
teases presumably aid in cell migration, local invasion, and 3 of which had metastasized. Four (4/12) cardiac rhabdomyomas
metastasis.59,117 are reported,50,66,85,119 as well as 1 myocardial hamartoma of the
right atrium.81 One malignant mixed mesenchymal tumor
(mesenchymoma) with rhabdomyosarcomatous differentiation
Case Reports in Veterinary Literature was reported in the heart of an 8-year-old female Pug.82
One rhabdomyoma of the tongue was reported in a 9-year-
Frequency of Published Cases old, castrated male, mixed breed dog.122 One mesenchymoma
The frequency of (nonlaryngeal/noncardiac) canine rhabdomyo- with rhabdomyosarcomatous differentiation was reported in the
sarcoma in veterinary medicine is low, with 65 total case reports spleen of a dog.159 There are 15 reports of cats with
published (Table 4) and available by a PubMed search and in the RMS,20,31,57,88,96,98,110,132,137 8 in sheep,41,106,142 and 15 in the
textbook Tumors of Domestic Animals.31 Pleomorphic RMS is the horse.18,28,44,45,55,147,151 Feldman recounts 1 rhabdomyoma in
least common (n = 2; 3%),8,31 and botryoid RMS is the most com- the shoulder of a 13-year-old horse.44
mon (n = 28; 43%),3,11,31,47,54,64,65,68,73,83,108,115,131,138,140,141,144,155 In addition to the skeletal muscle RMS mentioned in pigs,
followed by embryonal RMS (n = 15; 23%).7,31,35,61,62,65,153,166,167 cardiac rhabdomyomas are found in pigs more often than in
Unclassified RMS (RMS NOS) is prevalent in the literature (n = other species. A peculiar tumor of cattle appears histologically
13; 20%),14,16,21,49,74,75,99,102,103,113,162 and probably reflects the as cuboidal epithelioid cells arranged in glandular acini in a col-
lack of knowledge of RMS classification in veterinary medicine. lagenous stroma. These have been termed myocardial epithelial
Alveolar RMS (n = 7; 11%) is less frequently diagnosed in canines inclusions or myocardial adenomatoid tumors.12,135,152 They
second only to pleomorphic RMS.11,31,65,100,125,128,134 In human are hamartomas composed of pericardial mesothelial inclusions.
Table 4. Veterinary Case Reports and Literature Review of Canine Rhabdomyosarcomas (Noncardiac).

816
Reference Species/Breed Sex Age Clinical Signs Location Metastasis

Alveolar RMS
Seibold, 1974128 Mixed breed FS 2y Subcutaneous mass in the left maxillary Maxilla Pancreas, adrenals, left
area popliteal, and right
sublumbar lymph node;
local infiltration into bone
Sarnelli et al, 1994125 Dog FS 2y Palpable abdominal mass Greater omentum None
Kim et al, 199665 Rottweiler F 11 mo Maxillary swelling Gingiva/ maxilla None
Cooper and Valentine, 200231 Dog M 7y Hip Not reported
Bae et al, 200711 Golden F 2y Uterus Lung, liver, kidney LN
Murakami et al, 2010100 Mixed breed MC 15 mo Epistaxis and sneezing Cauliflower-shaped gingival mass Lung, brain
Snyder and Michael, 2011134 Golden MC 7 mo Maxillary Gingiva None
Embryonal RMS
Kim et al, 199665 Basset M 1.5 y Unilateral periorbital swelling, Oropharynx and the temporal Regional lymph node and
exophthalmia, and pain upon opening muscles maxillary gingiva lungs
the mouth
Yanoff et al, 1996166 Akita M 4 mo Noisy respiration, excessive panting, Trachea/neck/recurrent None
and exercise intolerance; veterinarian laryngeal nerve
showed a lateral deviation in the
cervical trachea
Cooper and Valentine, 200231 Dog F 14 y NA Axilla Not reported
Dog M 6y NA Perirenal Not reported
Dog F 12 y NA Tongue Not reported
Dog M 4y NA Muscle of flank Not reported
Dog F 11 y NA Tongue Not reported
Dog F Adult NA Mammary Not reported
Dog M 1.5 y NA Hard palate Not reported
Illanes, 200261 Labrador Retriever M 23 mo Unilateral atrophy of masticatory Right side masticatory muscle Liver
muscles invasive and trigemical nerve
Ueno et al, 2002153 Labrador Retriever MC 11 y Mass on perineum Perineum None confirmed
Yhee et al, 2008167 Golden M 8 mo Left forelimb lameness Mass in the left axillary region; Lung
osteolytic lesion in the scapula
Avallone et al, 20107 English Pointer FI 11 y Subcutaneous mass Left forearm None
da Roza et al, 201135 Boxer M 11 mo Incoordination and presence of a little Skull None
mass in the frontal region of the skull
Kato et al, 201262 Welsh Corgi M 2y Left upper eyelid enlargement Lacrimal gland, orbit Subcutis, multifocally, body
Botryoid
Osborne et al, 1968108 St Bernard M 5y Bladder None
Stamps and Harris, 1968138 Basset Hound F 1y dysuria Bladder, ureters Retroperitoneal lymph node,
great and lesser omentum,
small intestine, mesentery,
peritoneal surface, kidneys,
adrenal glands, liver, spleen,
lungs
Teunissen and Misdorp, 1968144 Great Dane FI 1.5 y Dysuria Bladder None
(continued)
Table 4. (continued)

Reference Species/Breed Sex Age Clinical Signs Location Metastasis

Kelly, 197364 St Bernard F 1y Dysuria, hematuria Urinary bladder neck None


Miniature Poodle F 1.5 y Dysuria, hematuria Urinary bladder neck None
St Bernard F 1.5 y Dysuria, hematuria Urinary bladder neck None
St Bernard F 14 mo Dysuria, hematuria Urinary bladder neck None
Great Dane F 1.5 y Dysuria, hematuria Urinary bladder neck None
St Bernard M 5y Dysuria, hematuria Urinary bladder neck None
Basset Hound F 1y Dysuria, hematuria Urinary bladder neck Local lymph nodes,
mesentery, kidney, adrenal,
liver, spleen, and lungs
Halliwell and Ackerman, 197454 Doberman Pinscher M 1.5 y Cystitis, enlarged tarsal and carpal joints Bladder None
Pletcher and Dalton, 1981115 Irish Setter M 20 mo Dysuria and hematuria Bladder Not reported
Andreasen et al, 19883 Doberman Pinscher M 2y Dysuria, hematuria Bladder Not reported
Labrador FS 18 mo Hematuria, pollakiuria Bladder Not reported
Great Dane MC 1y Stranguria, dysuria Bladder Not reported
van Vechten et al, 1990155 Doberman Pinscher MC 13 mo Urinary incontinence Bladder Colonic lymph node
Senior et al, 1993131 Labrador Retriever M 10 mo Stranguria, pollakiuria, hematuria Bladder None
Kim et al, 199665 Rottweiler F 11 mo Cystitis Urinary bladder None
Kuwamura et al, 199873 Newfoundland F 13 mo Hematuria and stranguria Urinary bladder None
Cooper and Valentine, 200231 Dog F 6 mo Bladder Not reported
Dog F 1y Bladder Not reported
Takiguchi et al, 2002141 Maltese F 2y Hematuria, pollakiuria Urinary bladder Liver
Madarame et al, 200383 Labrador Retriever F 9 mo Pollakiuria, anorexia Urinary bladder None
Kobayashi et al, 200467 Dog F 18 mo Hematuria Urinary bladder None
Dog F 12 mo hematuria Urinary bladder Liver, lung, heart, kidney,
ovary, and skin
Suzuki et al, 2006140 Dog 10 y Vagina None
Bae et al, 200711 Golden Retriever F 2y Pain, vaginal bleeding, dysuria Urinary bladder None
Gerber and Rees, 200947 Labrador Retriever MI 8 mo Lower urinary tract obstruction, Urinary bladder Lymph nodes and the liver,
hydronephrosis, hyrodureter lungs, mesenteric lymph
nodes, mediastinum, heart,
subcutaneous tissue, and
several muscle groups
Pleomorphic RMS
Cooper and Valentine, 200231 Dog M 3y Neck Not reported
Azakami et al, 20118 Mixed MI 14 y Prostate Not reported
Rhabdomoysarcoma NOS
Mulligan, 194999 Dog Unknown Tongue Not reported
Worley and Gorham, 1954162 English Setter M 8y Weight loss, lameness left rear leg Thigh Lung, liver, spleen, kidney,
lymph node, adrenal glands
Peter and Kluge, 1970113 German Shepherd M 2y Dysphagia Mandible Parotid, mandibular,
retropharyngeal,
mediastinal, and bronchial
lymph nodes; lungs; heart
(continued)

817
818
Table 4. (continued)

Reference Species/Breed Sex Age Clinical Signs Location Metastasis

Ladds and Webster, 197174 Welsh Corgi FI 6y Dysphagia Pharynx, soft palate Locally invasive to lymph
node
Bastianello, 198314 Dog Ocular muscle Not reported
Lascelles et al, 199875 Staffordshire Bull Terrier 6y Dysphagia Tongue None
Ginel et al, 200249 Boxer FI 7y Left forelimb lameness Fifth metacarpal Another limb
Necas et al, 2003103 Hovawart F 3y Right eye hyperemia, epiphora, Retrobulbar None
exophthalmos
Brockus and Myers, 200416 Dog 10 y Tongue Multicentric
Chapman et al, 200821 Rat Terrier FS 3.5 y Dysphagia, gagging Tongue None
Nakaichi et al, 2007102 Dog 10 y Left maxillofacial area Lung
Dog 10 y Left maxillofacial area Lymph node
Dog 12 y Right upper molar Lung
Laryngeal rhabdomoyoma or RMS
Liggett et al, 198577 Schnauzer cross F 4y Dyspnea Larynx None
Meuten et al, 198592 Scottish Terrier F 2y Dyspnea Larynx None
Doberman Pinscher MC 7y Dyspnea larynx None
Henderson, et al, 199156 Spitz FS 6y Exercise intolerance, stridor Larynx Liver
Block et al, 199515 Mixed breed dog FS 9y Larynx None
Clercx et al, 199830 Golden Retriever M 3y Dyspnea, exercise intolerance, stridor larynx None
Madewell et al, 198884 Doberman Pinscher F 11 y Larynx None
Barnhart and Lewis, 200013 Dachshund FS 3y Dyspnea Larynx None
OHara et al, 2001107 Golden FS 4y Dyspnea Larynx None
Cooper and Valentine, 200231 Dog FS 10 y Larynx None
Dog M 8y Larynx None
Yamate et al, 2011164 Mixed breed dog FS 6y Dyspnea, stridor Larynx, epiglottis None

F, female; FS, FI, female intact; female spayed; LN, lymph node; M, male; MC, male castrated; MI, male intact; NA, ; NOS, not otherwise specified; RMS, rhabdomyosarcoma.
Caserto 819

Table 5. Comparison of Signalment and Biologic Behavior of Canine Rhabdomyosarcoma (RMS) and Canine Soft Tissue Sarcomas.

Rhabdomyosarcoma
Soft Tissue Sarcoma Alveolar and Embryonal Botryoid

Metastasis 17% overall/grade 3 up to 50% 50% 27%


Survival 60% Poor Poor
Growth rate Slow Rapid Rapid
Degree of differentiation Grade 1 = well differentiated/grade 3 = poor Poor Moderate
Cell morphology Spindyloid-polygonal Round/mixed Mixed round/spindle
Age 10+ y <2 y (57%); <10 y (86%)
Canine soft tissue sarcoma (STS) has a range of metastatic rates. Grade 3 STS compares with the metastasis of alveolar and embryonal RMS, and grade 2 STS
compares with metastatic rates of botryoid RMS. Survival is generally poor in canine RMS, but survival depends on grade of STS. Growth rate of RMS tends to be more
rapid than the average growth rate of STS. STS has a range of cellular morphologies, and RMS tends to have poor differentiation. Age of onset of STS differs greatly
with RMS, being about 10 years on average for STS and most RMS occurring <10 years old, with 53% (n = 39/73) occurring in dogs <2 years old.38,42,47,72,130

Poorly differentiated sarcomas are occasionally reported in prognostic implications. Children 1 to 4 years old have a 5-year
the literature. These microscopically resemble embryonal survival of 77%, and for those with localized disease, it is even
tumors, with small cells and scanty cytoplasm, and have higher at 83%.116 Orbital and genitourinary locations have a 5-
hyperchromatic nuclei. Immunohistochemical studies often year survival of 86% and 80%, respectively.116 Poor prognosis
do not include myogenin or MyoD1 but are negative for des- is associated with RMS diagnosed at infancy or adolescence
min.33 Young age, or genitourinary location, should indicate (47%-48%, respectively) and locations including the limbs
a possible RMS despite negative desmin staining. Increased (50%), trunk (52%) and retroperitoneum (52%).116
availability of myogenin, MyoD1, or other primitive myogenic In humans, there is slightly better survival with embryonal
markers may help diagnose alveolar and embryonal RMS in histology (67%) compared with alveolar histology (49%). Low-
these circumstances. est 5-year survival is correlated to metastasis at the time of pre-
In a single case report,16 a dog developed multiple rhabdo- sentation (31%) and alveolar histology (49%).116 Prognosis of
myosarcomas in multiple locations that were categorized as patients with regional lymph node metastasis is similar to distant
embryonal RMS at one location and alveolar RMS at another. metastasis in alveolar RMS but not embryonal RMS.123
It is unclear whether these neoplasms arose separately or are the Botryoid rhabdomyosarcomas in humans are locally infiltrative
product of a metastatic process and subsequent divergence in and are second only to the less common spindyloid variant
phenotype. Kim et al65 diagnosed an alveolar RMS in the gin- embryonal RMS in favorable prognosis.94 Most human embry-
giva of an 11-month-old Rottweiler, and upon necropsy, onal rhabdomyosarcomas fall into the intermediate risk category
although no metastasis was reported, a botryoid RMS was diag- (55%), with a 3-year survival of 59% to 85%94 and a 5-year sur-
nosed in the urinary bladder. These tumors were considered to vival of 67%.116 In contrast, even though spindyloid RMS com-
arise independent of each other. A report of orbital embryonal prises 3% of all RMS, it has a 95% survival at 5 years. Survival
RMS in a dog described metastasis to the cervical region with of people with alveolar RMS ranges from 49% to 66% at 5
a histologic pattern of alveolar RMS.62 Clearly, these incidences years.94,116 Embryonal and botryoid RMS are frequently
warrant further investigation into the diagnosis, pathogenesis, reported to have lower metastatic rates than alveolar RMS, but
and the identification of prognostic markers of RMS in dogs. precise figures are not often reported.118 Only about 15% to
In the meantime, comparison to human RMS pathogenesis and 20% of patients have clinically detectable distant metastasis at
prognosis should be made with caution. the time of diagnosis, but all are considered to have micrometa-
static disease.94

Clinical Features of Rhabdomyosarcomas Canine Rhabdomyosarcoma and Soft Tissue Sarcomas. Rhabdo-
myosarcomas are occasionally grouped together within the broad
Human Rhabdomyosarcoma category of canine soft tissue sarcomas,89 yet they often have a
Childhood rhabdomyosarcoma primarily affects children different clinical signalment and location. Soft tissue sarcomas
younger than 15 years19,36,47,58,60,101,109,116 and comprises graded by the Kuntz et al72 system are restricted to subcutaneous
about two-thirds of soft tissue sarcomas in children.146 Because neoplasms and exclude hemangiosarcomas and rhabdomyosarco-
of the significant difference in survival times in humans mas. Table 5 compares the biological behavior of canine RMS
between alveolar and embryonal rhabdomyosarcomas, it is nec- and soft tissue sarcomas. The most significant difference between
essary for human pathologists to reliably differentiate the two, the two is the age of onset, location, and survival.
even when both appear as small round cell neoplasms. With cur- RMS is often reported as appearing most often in younger
rent chemotherapy treatments, the 5-year survival of embryonal dogs, 2 years old or younger. A survey of the reported cases
RMS in children is about 67% to 70%, whereas for alveolar of nonlaryngeal RMS indicates that 63% (n = 39/62) occurs
RMS, it is about 40% to 49%.116,158 Age and location also have in young dogs (2 years old) (Fig. 14). In dogs, 89% (n = 25/
820 Veterinary Pathology 55(5)

Figure 14. Age of diagnosis of canine rhabdomyosarcoma (RMS). A literature search shows 63% (n = 39/62; in 2 cases, the age was not
reported) of canine RMS (excluding laryngeal rhabdomyomas/rhabdomyosarcoma) occurs in dogs younger than 2 years. The remaining 37%
is divided between animals 3 to 6 years old (n = 9/62; 15%), 7 to 10 years old (n = 7/62; 11%), and >10 years old (n = 7/62; 11%). Eighty-
nine percent (n = 55/62) of cases occur in dogs younger than 10 years. Figure 15. Subclasses of canine RMS. (a) Botryoid RMS was the most
frequent canine RMS (n = 28/65; 43% of cases excluding laryngeal tumors). Embryonal RMS was diagnosed in 23% of cases of nonlaryngeal ske-
letal muscle tumors (n = 15/65). RMS not otherwise specified (NOS) was diagnosed in 20% of cases (n = 13/65), and alveolar RMS was diagnosed
in 11% (n = 7/65) of cases. Pleomorphic RMS was diagnosed least frequently (n = 2/65; 3%). Most of the alveolar (86%; n = 6/7) and botryoid RMS
Caserto 821

28) of botryoid RMS occurs in dogs 2 years old or younger. may also resemble pleomorphic RMS with numerous cells hav-
Similarly, in alveolar RMS, 86% (n = 6/7) occurs in young dogs. ing bizarre mitotic figures and having cells with a high degree of
By contrast, only 47% of embryonal RMS occurs in dogs 2 anisocytosis and anisokaryosis.42,72 Drawing conclusions from
years old or younger. Only 8% (n = 1/13) of RMS NOS occurs these data is difficult because of the paucity of clinical cases,
in young dogs, and no reported cases of pleomorphic RMS have lack of follow-up and survival data, and in many cases unknown
occurred in young dogs. Although this seems to correspond to frequency of metastasis. Improved diagnostic techniques for
the high prevalence of this subclass in human adults, the fre- RMS may lead to more diagnosed cases and better data on sur-
quency of this diagnosis is very low (n = 2) (Fig. 15). vival and prognosis.
Soft tissue sarcomas arise mostly in middle-aged to older
dogs and occur in the skin and subcutis of the trunk and extre-
mities and oral cavity.38,42 RMS occurs often in the urogenital
Summary
tract (n = 32/65; 49%), and these are most frequently botryoid Rhabdomyosarcomas are a diverse and histologically variable
RMS of the urinary bladder. The second most common location set of highly malignant neoplasms occurring predominately in
is the head, neck, and face (n = 24/65; 37%), and a small per- younger individuals. Subclassification has been attempted
centage occurs in the hip/pelvis/spinal column (n = 2/65; 3%), based on histologic morphology, but these classification
appendicular skeleton (n = 5/65; 8%), and skin (including mam- schemes in human and veterinary medicine fall short when deal-
mary gland) (n = 2/65; 3%) (Fig. 16). ing with very homogeneous cell populations with no particu-
It is difficult to compare metastatic rates between canine larly identifying characteristics. Identification of particular
RMS subgroups because in some cases, these data were not gene mutations is useful in human medicine and may become
reported or euthanasia was elected at the time of diagnosis. In the standard for distinguishing between alveolar and embryonal
veterinary medicine, the prognosis depends on the severity and tumors. The use of IHC to identify desmin, -actins, myogenin,
extent of invasiveness, as well as the presence of metastatic dis- and MyoD1 and electron microscopic identification of sarco-
ease at the time of diagnosis. In cases of extensive local infiltra- meric structures can be used to identify RMS in relatively undif-
tion or radiographic evidence of metastatic disease, euthanasia ferentiated neoplasms. In veterinary medicine, much work
may be sought instead of radical surgery or adjunct therapy.31,65 needs to be done to evaluate the prognostic significance of sub-
Metastasis reported at the time of RMS diagnosis or at necropsy classifications and to identify new methods to characterize these
shows that metastatic rates of RMS in dogs is highest in RMS neoplasms to make classification effective and useful for veter-
NOS (n = 6/11; 55%), followed closely by embryonal RMS inary oncologists and pathologists. Further investigation is
(n = 4/8; 50%) and alveolar RMS (n = 3/6; 50%). Botryoid needed to better understand the biology of these neoplasms that
RMS has the lowest metastatic rate (n = 6/22; 27%) (Fig. 17). may aid in diagnosis and prognosis. Unless veterinary medicine
In reported literature, botryoid RMS is reported to rarely metas- can identify prognostically significant markers or mutations in
tasize.65,149 There are no data on metastasis of pleomorphic dogs, the classification system currently used may continue to
RMS (n = 2). It is interesting that of the metastatic cases of yield poorly prognostic diagnoses.
canine botryoid, embryonal, and alveolar RMS, all are 2 years
old, whereas the metastatic cases of RMS NOS are mostly older Declaration of Conflicting Interests
than 2 years (Fig. 17). The author(s) declared no potential conflicts of interest with respect to
High-grade soft tissue sarcomas have similar rates of metas- the research, authorship, and/or publication of this article.
tasis and recurrence to reported incidence of canine RMS and
therefore may carry the same prognosis. Kuntz et al72 reported Funding
the metastatic rate of grade 3 soft tissue sarcomas without adju- The author(s) received no financial support for the research, authorship,
vant therapy as 46% (ranging from 41%50%), which is and/or publication of this article.
roughly equivalent to embryonal RMS, alveolar RMS, and
RMS NOS but is greater than botryoid RMS.130 Grade 1 and References
2 soft tissue sarcomas fare better at 11% and 19%, respectively. 1. Akko A, Ozyigit MO, Yilmaz R, et al. Cardiac metastasising
In addition, high-grade soft tissue sarcomas may be poorly dif- rhabdomyosarcoma in a Great Dane. Vet Rec. 2006;158(23):
ferentiated, resembling alveolar and embryonal RMS, and yet 803804.

Figure 15. (continued) (89%; n = 25/28) occurred in dogs younger than 2 years. Figure 16. Locations of canine RMS. The urogenital tract was
the most common location (n = 32/65; 49%), including botryoid RMS and rarely alveolar RMS (n = 2) and embryonal RMS (n = 1). The head, neck,
and face were common locations (n = 24/65; 37%). In this location, tumors were diagnosed as RMS NOS (n = 11) and occasionally as embryonal
RMS (n = 8) and alveolar RMS (n = 4). Less common locations included the limbs (n = 5/65; 8%), including 3 embryonal RMS and 2 RMS NOS. In the
hip and spine (n = 2/65; 3%) 1 alveolar RMS and 1 embryonal RMS were reported. In the skin and mammary glands (n = 2/65; 3%), 2 embryonal RMS
had been diagnosed. Figure 17. Metastasis of canine RMS (a). Alveolar (n = 3/6) and embryonal RMS (n = 4/8) had equal rates of metastasis (50%).
RMS NOS metastasized in 55% (6/11) of cases. Botryoid RMS had a lower metastatic rate (n = 6/22; 27%). Of the 2 cases of pleomorphic RMS,
metastasis was not mentioned. Comparison of only metastatic tumors (b) shows that all of the metastatic tumors occurred in dogs 2 years old in
alveolar, embryonal, and botryoid subtypes. In contrast, almost all RMS NOS occurred in dogs older than 2 years. Metastatic rates are hard to
interpret due to inadequate follow-up, euthanasia at the time of diagnosis, and time interval between diagnosis and necropsy.
822 Veterinary Pathology 55(5)

2. Anderson J, Gordon T, McManus A, et al. Detection of the PAX3- with emphasis on spindle cell mimics. Am J Surg Pathol. 2001;25
FKHR fusion gene in paediatric rhabdomyosarcoma: a reproduci- (9):11501157.
ble predictor of outcome? Br J Cancer. 2001;85(6):831835. 20. Chang H-W, Ho S-Y, Lo H-F, et al. Vaccine-associated rhabdo-
3. Andreasen CB, White MR, Swayne DE, et al. Desmin as a marker myosarcoma with spinal epidural invasion and pulmonary metas-
for canine botryoid rhabdomyosarcomas. J Comp Pathol. 1988;98 tasis in a cat. Vet Pathol. 2006;43(1):5558.
(1):2329. 21. Chapman S, Nabity M, Calise D. What is your diagnosis? Lingual
4. Armeanu-Ebinger S, Bonin M, Hbig K, et al. Differential expres- mass in a dog. Vet Clin Pathol. 2008;37(1):133139.
sion of invasion promoting genes in childhood rhabdomyosar- 22. Charrasse S, Comunale F, Fortier M, et al. M-cadherin activates
coma. Int J Oncol. 2011;38(4):9931000. Rac1 GTPase through the Rho-GEF trio during myoblast fusion.
5. Asmar L, Gehan EA, Newton WA, et al. Agreement among and Mol Biol Cell. 2007;18(5):17341743.
within groups of pathologists in the classification of rhabdomyo- 23. Chen EH, Olson E. Towards a molecular pathway for myoblast
sarcoma and related childhood sarcomas: report of an international fusion in Drosophila. Trends Cell Biol. 2004;14(8):452460.
study of four pathology classifications. Cancer. 1994;74(9): 24. Chen EH. Unveiling the mechanisms of cell-cell fusion. Science.
25792588. 2005;308(5720):369373.
6. Aupperle H, Marz I, Ellenberger C, et al. Primary and secondary 25. Chi AC, Barnes JD, Budnick S, et al. Rhabdomyosarcoma of the
heart tumours in dogs and cats. J Comp Pathol. 2007;136(1): maxillary gingiva. J Periodontal. 2007;78(9):18391845.
1826. 26. Choi J, Costa ML, Mermelstein CS, et al. MyoD converts primary
7. Avallone G, Pinto da Cunha N, Palmieri C, et al. Subcutaneous dermal fibroblasts, chondroblasts, smooth muscle, and retinal pig-
embryonal rhabdomyosarcoma in a dog: cytologic, immunocyto- mented epithelial cells into striated mononucleated myoblasts and
chemical, histologic, and ultrastructural features. Vet Clin Pathol. multinucleated myotubes. Proc Natl Acad Sci U S A. 1990;87(20):
2010;39(4):499504. 79887992.
8. Azakami D, Shibutani H, Dohi M, et al. Establishment and charac- 27. Churg A, Ringus J. Ultrastructural observations on the histogen-
terization of canine rhabdomyosarcoma cell line CMS-C. J Vet esis of alveolar rhabdomyosarcoma. Cancer. 1978;41(4):
Med Sci. 2011;73(8):11051108. 13551361.
9. Babai F, Musevi-Aghdam J, Schurch W, et al. Coexpression of 28. Clegg PD, Coumbe A. Alveolar rhabdomyosarcoma: an unusual
alpha-sarcomeric actin, alpha-smooth muscle actin and desmin cause of lameness in a pony. Equine Vet J. 1993;25(6):547549.
during myogenesis in rat and mouse embryos, I: skeletal muscle. 29. Clement V, Sanchez P, de Tribolet N, et al. HEDGEHOG-GLI1
Differentiation. 1990;44(2):132142. signaling regulates human glioma growth, cancer stem cell self-
10. Bach A-S, Enjalbert S, Comunale F, et al. ADP-ribosylation factor renewal, and tumorigenicity. Curr Biol. 2007;17(2):165172.
6 regulates mammalian myoblast fusion through phospholipase D1 30. Clercx C, Desmecht D, Michiels L, et al. Laryngeal rhabdomyoma
and phosphatidylinositol 4,5-bisphosphate signaling pathways. in a golden retriever. Vet Rec. 1998;143(7):196198.
Mol Biol Cell. 2010;21(14):24122424. 31. Cooper BJ, Valentine B. Tumors of muscle. In: Meuten DJ, ed.
11. Bae I-H, Kim D-Y, Pakhrin B, et al. Genitourinary rhabdomyosar- Tumors in Domestic Animals. 4th ed. Ames, IA: Blackwell;
coma with systemic metastasis in a young dog. Vet Pathol. 2007;44 2002:319363.
(4):518520. 32. Cooper E. A new role for ion channels in myoblast fusion. J Cell
12. Baker DC, Schmidt SP, Langheinrich KA, et al. Bovine myocar- Biol. 2001;153(4):F9F11.
dial epithelial inclusions. Vet Pathol. 1993;30(1):8288. 33. Cora MC, Neel JA. Poorly differentiated uterine or cervical
13. Barnhart K, Lewis B. Laryngopharyngeal mass in a dog with upper sarcoma in a young dog. Case Rep Vet Med. 2011;2011:15.
airway obstruction. Vet Clin Pathol. 2000;29(2):4750. 34. Costa ML, Escaliera RC, Rodrigues VB, et al. Some distinctive
14. Bastianello SS. A survey on neoplasia in domestic species over a features of zebrafish myogenesis based on unexpected distribu-
40-year period from 1935 to 1974 in the Republic of South Africa, tions of the muscle cytoskeletal proteins actin, myosin, desmin,
VI: tumours occurring in dogs. Onderstepoort J Vet Res. 1983;50 -actinin, troponin and titin. Mech Dev. 2002;116(12):95104.
(3):199220. 35. da Roza MR, de Amorim RFB, Carneiro FP, et al. Aggressive spin-
15. Block G, Clarke K, Salisbury SK, et al. Total laryngectomy and dle cell rhabdomyosarcoma in an 11-month-old boxer dog. J Vet
permanent tracheostomy for treatment of laryngeal rhabdomyosar- Med Sci. 2010;72(10):13631366.
coma in a dog. J Am Anim Hosp Assoc. 1995;31(6):510513. 36. Dagher R, Helman L. Rhabdomyosarcoma: an overview. Oncolo-
16. Brockus CW, Myers RK. Multifocal rhabdomyosarcomas within gist. 1999;4(1):3444.
the tongue and oral cavity of a dog. Vet Pathol. 2004;41(3): 37. Dennis MM, McSporran KD, Bacon NJ, et al. Prognostic factors
273274. for cutaneous and subcutaneous soft tissue sarcomas in dogs. Vet
17. Bundtzen JL, Norback DH. The ultrastructure of poorly differen- Pathol. 2011;48(1):7384.
tiated rhabdomyosarcomas: a case report and literature review. 38. Dennis MM, Reddacliff LA, Whittington RJ. Longitudinal study
Hum Pathol. 1982;13(4):301313. of clinicopathological features of Johnes disease in sheep natu-
18. Castleman WL, Toplon DE, Clark CK, et al. Rhabdomyosarcoma rally exposed to Mycobacterium avium subspecies paratuberculo-
in 8 horses. Vet Pathol. 2011;48(6):11441150. sis. Vet Pathol. 2011;48(3):565575.
19. Cessna M, Zhou H, Perkins S, et al. Are myogenin and MyoD1 39. Dias P, Chen B, Dilday B, et al. Strong immunostaining for
expression specific for rhabdomyosarcoma? A study of 150 cases, myogenin in rhabdomyosarcoma is significantly associated
Caserto 823

with tumors of the alveolar subclass. Am J Pathol. 2000;156 testing in diagnosing the alveolar subtype. Cancer. 2004;101
(2):399408. (12):28172824.
40. Doberstein S, Fetter R, Mehta A, et al. Genetic analysis of myo- 61. Illanes O. Juvenile parameningeal rhabdomyosarcoma in a dog
blast fusion: blown fuse is required for progression beyond the pre- causing unilateral denervation atrophy of masticatory muscles. J
fusion complex. J Cell Biol. 1997;136(6):12491261. Comp Pathol. 2002;126(4):303307.
41. Donnelly WJ, Hamilton AF. Fatal pulmonary rhabdomyosarcoma 62. Kato Y, Notake H, Kimura J, et al. Orbital embryonal rhabdomyo-
in a lamb. Vet Rec. 1972;91(12):280282. sarcoma with metastasis in a young dog. J Comp Pathol. 2012;147
42. Ehrhart N. Soft-tissue sarcomas in dogs: a review. J Am Anim Hosp (23):191194.
Assoc. 2005;41(4):241246. 63. Kawabata N, Ijiri K, Ishidou Y, et al. Pharmacological inhibition of
43. Epstein J, Lam P, Jepeal L, et al. Pax3 inhibits myogenic differen- the Hedgehog pathway prevents human rhabdomyosarcoma cell
tiation of cultured myoblast cells. J Biol Chem. 1995;270(20): growth. Int J Oncol. 2011;39(4):899906.
1171911722. 64. Kelly DF. Rhabdomyosarcoma of the urinary bladder in dogs. Vet
44. Feldman WH. Rhabdomyoblastoma. In: Neoplasms of Domestic Pathol. 1973;10(5):375384.
Animals. Philadephia, PA: WB Saunders Co; 1932:169177. 65. Kim D-Y, Hodgin EC, Cho DY, et al. Juvenile rhabdomyosarco-
45. Finocchio EJ, Coffman JR, Strafuss AC. Rhabdomyosarcoma in a mas in two dogs. Vet Pathol. 1996;33(4):447450.
horse. Vet Med Small Anim Clin. 1969;64(6):494496. 66. Kizawa K, Furubo S, Sanzen T, et al. Cardiac rhabdomyoma in a
46. Franke WW. Discovering the molecular components of intercellu- Beagle dog. J Toxicol Pathol. 2002;15(1):6972.
lar junctionsa historical view. Cold Spring Harb Perspec. 2009; 67. Kobayashi M, Sakai H, Hirata A, et al. Expression of myogenic
1(3):a003061a003061. regulating factors, myogenin and MyoD, in two canine botryoid
47. Gerber K, Rees P. Urinary bladder botryoid rhabdomyosarcoma rhabdomyosarcomas. Vet Pathol. 2004;41(3):275277.
with widespread metastases in an 8-month-old Labrador cross dog. 68. Kobayashi M, Sakai H, Hirata A, et al. Expression of myogenic
J S Afr Vet Assoc. 2009;80(3):199203. regulating factors, myogenin and MyoD, in two canine botryoid
48. Ghibaudo G, Bettini G, Abramo F. Anaplastic and aggressive sub- rhabdomyosarcomas. Vet Pathol. 2004;41(3):275277.
cutaneous sarcoma in a seven-month-old dog. J Small Anim Pract. 69. Krotje LJ, Ware WA, Niyo Y. Intracardiac rhabdomyosarcoma in a
2008;49(6):310313. dog. J Am Vet Med Assoc. 1990;197(3):368371.
49. Ginel PJ, Martin de las Mulas J, Lucena R, et al. Skeletal muscle 70. Kucharczak J, Charrasse S, Comunale F, et al. R-cadherin expres-
rhabdomyosarcoma in a dog. Vet Rec. 2002;151(24):736739. sion inhibits myogenesis and induces myoblast transformation via
50. Girard C, Helie P, Odin M. Intrapericardial neoplasia in dogs. J Vet Rac1 GTPase. Cancer Res. 2008;68(16):65596568.
Diagn Invest. 1999;11(1):7378. 71. Kumar S, Perlman E, Harris CA, et al. Myogenin is a specific mar-
51. Gofflot S, Kischel P, Thielen C, et al. Characterization of an anti- ker for rhabdomyosarcoma: an immunohistochemical study in
body panel for immunohistochemical analysis of canine muscle paraffin-embedded tissues. Mod Pathol. 2000;13(9):988993.
cells. Vet Immunol Immunopathol. 2008;125(34):225233. 72. Kuntz CA, Dernell W, Powers B, et al. Prognostic factors for sur-
52. Gonin-Jmaa D, Paulsen DB, Taboada J. Pericardial effusion in a gical treatment of soft-tissue sarcomas in dogs: 75 cases (1986
dog with rhabdomyosarcoma in the right ventricular wall. J Small 1996). J Am Vet Med Assoc. 1997;211(9):11471151.
Anim Pract. 1996;37(4):193196. 73. Kuwamura M, Yoshida H, Yamate J, et al. Urinary bladder rhabdo-
53. Green KJ, Simpson CL. Desmosomes: new perspectives on a myosarcoma (sarcoma botryoides) in a young Newfoundland dog.
classic. J Investig Dermatol. 2007;127(11):24992515. J Vet Med Sci. 1998;60(5):619621.
54. Halliwell WH, Ackerman N. Botryoid rhabdomyosarcoma of the 74. Ladds PW, Webster DR. Pharyngeal rhabdomyosarcoma in a dog.
urinary bladder and hypertrophic osteoarthropathy in a young dog. Vet Pathol. 1971;8(3):256259.
J Am Vet Med Assoc. 1974;165(10):911913. 75. Lascelles BD, McInnes E, Dobson JM, et al. Rhabdomyosarcoma
55. Hanson PD, Frisbie DD, Dubielzig RR, et al. Rhabdomyosarcoma of the tongue in a dog. J Small Anim Pract. 1998;39(12):587591.
of the tongue in a horse. J Am Vet Med Assoc. 1993;202(8): 76. Liggett AD, Weiss R, Thomas KL. Canine laryngopharyngeal
12811284. rhabdomyoma resembling an oncocytoma: light microscopic,
56. Henderson RA, Powers RD, Perry L. Development of hypopar- ultrastructural and comparative studies. Vet Pathol. 1985;22(6):
athyroidism after excision of laryngeal rhabdomyosarcoma. 526532.
J Am Vet Med Assoc. 1991;198(4):639643. 77. Liggett AD, Weiss R, Thomas KL. Canine laryngopharyngeal
57. Hendrick MJ, Brooks JJ. Postvaccinal sarcomas in the cat: rhabdomyoma resembling an oncocytoma: light microscopic,
histology and immunohistochemistry. Vet Pathol. 1994;31(1): ultrastructural and comparative studies. Vet Pathol. 1985;22(6):
126129. 526532.
58. Hettmer S, Wagers AJ. Uncovering the origins of rhabdomyosar- 78. Lima LL de, Rodrigues CAC, Pereira PMR, et al. Primary
coma. Nat Med. 2010;16(2):171173. cutaneous alveolar rhabdomyosarcoma in a pediatric patient [in
59. Hosoyama T, Aslam MI, Abraham J, et al. IL-4R drives dediffer- English, Portuguese]. An Bras Dermatol. 2011;86(2):363365.
entiation, mitogenesis, and metastasis in rhabdomyosarcoma. Clin 79. Linardic CM, Naini S, Herndon JE, et al. The PAX3-FKHR fusion
Cancer Res. 2011;17(9):27572766. gene of rhabdomyosarcoma cooperates with loss of p16INK4A to
60. Hostein I, Andraud-Fregeville M, Guillou L, et al. Rhabdomyosar- promote bypass of cellular senescence. Cancer Res. 2007;67(14):
coma: value of myogenin expression analysis and molecular 66916699.
824 Veterinary Pathology 55(5)

80. Liu J, Guzman MA, Pezanowski D, et al. FOXO1-FGFR1 fusion 100. Murakami M, Sakai H, Iwatani N, et al. Cytologic, histologic,
and amplification in a solid variant of alveolar rhabdomyosarcoma. and immunohistochemical features of maxillofacial alveolar
Mod Pathol. 2011;24(10):13271335. rhabdomyosarcoma in a juvenile dog. Vet Clin Pathol. 2010;39
81. Machida N, Katsuda S, Yamamura H, et al. Myocardial hamar- (1):113118.
toma of the right atrium in a dog. J Comp Pathol. 2002;127(4): 101. Mysorekar VV, Harish K, Kilara N, et al. Embryonal rhabdomyo-
297300. sarcoma of the chest wall: a case report and review of the litera-
82. Machida N, Kobayashi M, Tanaka R, et al. Primary malignant ture. Indian J Pathol Bacteriol. 2008;51(2):274276.
mixed mesenchymal tumour of the heart in a dog. J Comp Pathol. 102. Nakaichi M, Itamoto K, Hasegawa K, et al. Maxillofacial rhabdo-
2003;128(1):7174. myosarcoma in the canine maxillofacial area. J Vet Med Sci.
83. Madarame H, Ito A, Tanaka R. Urinary bladder rhabdomyosar- 2007;69(1):6567.
coma (botryoid rhabdomyosarcoma) in a Labrador Retriever dog. 103. Neas A, Rauer P, Tich F, et al. Retrobulbar rhabdomyosarcoma
J Toxicol Pathol. 2003;16(4):279281. in a Hovawart. Acta Veterinaria Brno. 2003;72(3):399406.
84. Madewell B, Lund J, Munn R, et al. Canine laryngeal rhabdomyo- 104. Newton WA, Gehan EA, Webber BL, et al. Classification of rhab-
sarcoma: an immunohistochemical and electron microscopic domyosarcomas and related sarcomas: pathologic aspects and
study. Jpn J Vet Sci. 1988;50(5):10771084. http://www.ncbi. proposal for a new classificationan Intergroup Rhabdomyosar-
nlm.nih.gov/pubmed/3199616 coma Study. Cancer. 1995;76(6):10731085.
85. Mansfield CS, Callanan JJ, McAllister H. Intra-atrial rhabdo- 105. Nitzki F, Zibat A, Frommhold A, et al. Uncommitted precursor
myoma causing chylopericardium and right-sided congestive heart cells might contribute to increased incidence of embryonal rhab-
failure in a dog. Vet Rec. 2000;147(10):264267. domyosarcoma in heterozygous Patched1-mutant mice. Onco-
86. Marburger TB, Gardner JM, Prieto VG, et al. Primary cutaneous gene. 2011;30(43):44284436.
rhabdomyosarcoma: a clinicopathologic review of 11 cases. 106. ODonahoo R, Seawright AA. Rhabdomyosarcoma of the lungs
J Cutan Pathol. 2012;39(11):987995. of a sheep. Aust Vet J. 1971;47(11):572573.
87. Marshall AD, van der Ent MA, Grosveld GC. PAX3-FOXO1 and 107. OHara A, McConnell M, Wyatt K. Laryngeal rhabdomyoma in a
FGFR4 in alveolar rhabdomyosarcoma. Mol Carcinog. 2012;51 dog. Aust Vet J. 2001;79(12):817821.
(10):807815. 108. Osborne CA, Low DG, Perman V, et al. Neoplasms of the canine
88. Martin de las Mulas J, Vos JH, Van Mil FN. Desmin and vimentin and feline urinary bladder: incidence, etiologic factors, occurrence
immunocharacterization of feline muscle tumors. Vet Pathol. 1992; and pathologic features. Am J Vet Res. 1968;29(10):20412055.
29(3):260262. 109. Parham DM. Pathologic classification of rhabdomyosarcomas
89. McKnight J, Mauldin G, McEntee M, et al. Radiation treatment for and correlations with molecular studies. Mod Pathol. 2001;14
incompletely resected soft-tissue sarcomas in dogs. J Am Vet Med (5):506514.
Assoc. 2000;217(2):205210. 110. Patnaik AK, Schwartz PD, Greene RW. A histopathologic study
90. Mercer SE. Mirk/Dyrk1b mediates cell survival in rhabdomyosar- of twenty urinary bladder neoplasms in the cat. J Small Anim
comas. Cancer Res. 2006;66(10):51435150. Pract. 1986;27(7):433445.
91. Merlino G, Helman L. Rhabdomyosarcomaworking out the 111. Paululat A, Holz A, Renkawitz-Pohl R. Essential genes for
pathways. Oncogene. 1999;18(38):53405348. myoblast fusion in Drosophila embryogenesis. Mech Dev.
92. Meuten DJ, Calderwood Mays MB, Dillman RC, et al. Canine 1999;83(12):1726.
laryngeal rhabdomyoma. Vet Pathol. 1985;22(6):533539. 112. Paululat A, Holz A, Renkawitz-Pohl R. Essential genes for
93. Meuten DJ, Mays MBC, Dillman RC, et al. Canine laryngeal rhab- myoblast fusion in Drosophila embryogenesis. Mech Dev.
domyoma. Vet Pathol. 1985;22(6):533539. 1999;83(12):1726.
94. Meyer W, Spunt S. Soft tissue sarcomas of childhood. Cancer 113. Peter C, Kluge J. An ultrastructural study of a canine rhabdomyo-
Treat Rev. 2004;30(3):269280. sarcoma. Cancer. 1970;26(6):12801288.
95. Miettinen M, Lehto V, Badley R, Virtanen I. Alveolar rhabdomyo- 114. Prez J, Perez-Rivero A, Montoya A, et al. Right-sided heart fail-
sarcoma: demonstration of the muscle type of intermediate fila- ure in a dog with primary cardiac rhabdomyosarcoma. J Am Anim
ment protein, desmin, as a diagnostic aid. Am J Pathol. 1982; Hosp Assoc. 1998;34(3):208211.
108(2):246251. 115. Pletcher JM, Dalton L. Botryoid rhabdomyosarcoma in the
96. Miller A, Steffey M, Alcaraz A, et al. Embryonal rhabdomyosar- urinary bladder of a dog. Vet Pathol. 1981;18(5):695697.
coma in a young Maine Coon cat. J Am Anim Hosp Assoc. 116. Punyko JA, Mertens AC, Baker KS, et al. Long-term survival
2009;45(1):4347. probabilities for childhood rhabdomyosarcoma. Cancer. 2005;
97. Milner DJ, Weitzer G, Tran D, et al. Disruption of muscle 103(7):14751483.
architecture and myocardial degeneration in mice lacking desmin. 117. Psklloglu M, Lukasiewicz E, Miekus K, et al. Differential
J Cell Biol. 1996;134(5):12551270. expression of Snail1 transcription factor and Snail1-related genes
98. Minkus G, Hillemanns M. Botryoid-type embryonal rhabdo- in alveolar and embryonal rhabdomyosarcoma subtypes. Folia
myosarcoma of liver in a young cat. Vet Pathol. 1997;34(6): Histochem Cytobiol. 2010;48(4):671677.
618621. 118. Qualman S, Coffin C, Newton W, et al. Intergroup Rhabdomyo-
99. Mulligan RM. Other neoplasms. In: Neoplasms of the Dog. sarcoma Study: update for pathologists. Pediatr Dev Pathol.
Philadelphia, PA: The Williams & Wilkins Co.; 1949:110114. 1998;1:550561.
Caserto 825

119. Radi ZA, Metz A. Canine cardiac rhabdomyoma. Toxicologic 137. Spugnini EP, Filipponi M, Romani L, et al. Electrochemotherapy
Pathol. 2009;37(3):348350. treatment for bilateral pleomorphic rhabdomyosarcoma in a cat.
120. Ren Y, Finckenstein F, Abdueva D. Mouse mesenchymal stem J Small Anim Pract. 2010;51(6):330332.
cells expressing PAX-FKHR form alveolar rhabdomyosarcomas 138. Stamps P, Harris DL. Botryoid rhabdomyosarcoma of the
by cooperating with secondary mutations. Cancer Res. 2008;68 urinary bladder of a dog. J Am Vet Med Assoc. 1968;153(8):
(16):65876597. 10641068.
121. Riedlinger WFJ, Kozakewich HPW, Vargas SO. Myogenic mar- 139. Stiller D, Holzhausen HJ. Ultrastructural organization of cell
kers in the evaluation of embryonal botryoid rhabdomyosarcoma characteristics relevant to diagnosis in rhabdomyosarcomas [in
of the female genital tract. Pediatr Dev Pathol. 2005;8(4): German]. Zentralbl Allg Pathol. 1988;134(45):449466.
427434. 140. Suzuki K, Nakatani K, Shibuya H, et al. Vaginal rhabdomyosar-
122. Rivera RY, Carlton WW. Lingual rhabdomyoma in a dog. J Comp coma in a dog. Vet Pathol. 2006;43(2):186188.
Pathol. 1992;106(1):8387. 141. Takiguchi M, Watanabe T, Okada H, et al. Rhabdomyosarcoma
123. Rodeberg DA, Garcia-Henriquez N, Lyden ER, et al. Prognostic (botryoid sarcoma) of the urinary bladder in a Maltese. J Small
significance and tumor biology of regional lymph node disease in Anim Pract. 2002;43(6):269271.
patients with rhabdomyosarcoma: a report from the Childrens 142. Tanaka K, Stromberg PC. Embryonal rhabdomyosarcoma in a
Oncology Group. J Clin Oncol. 2011;29(10):13041311. sheep. Vet Pathol. 1993;30(4):396399.
124. Rota R, Ciarapica R, Giordano A, et al. MicroRNAs in rhabdo- 143. Taylor JG VI, Cheuk AT, Tsang PS, et al. Identification of
myosarcoma: pathogenetic implications and translational potenti- FGFR4-activating mutations in human rhabdomyosarcomas that
ality. Mol Cancer. 2011;10:120134. promote metastasis in xenotransplanted models. J Clin Invest.
125. Sarnelli R, Grassi F, Romagnoli S. Alveolar rhabdomyosarcoma of 2009;119(11):33953407.
the greater omentum in a dog. Vet Pathol. 1994;31(4):473475. 144. Teunissen GHB, Misdorp W. Rhabdomyosarcoma of the urinary
126. Schmidt RA, Cone R, Haas JE, et al. Diagnosis of rhabdomyosar- bladder and fibromatosis of the extremities in a young dog.
comas with HHF35, a monoclonal antibody directed against mus- Zentralbl Veterinarmed A. 1968;15(1):8188.
cle actins. Am J Pathol. 1988;131(1):19. 145. Tobar A, Avigad S, Zoldan M, et al. Clinical relevance of mole-
127. Sebire N, Malone M. Myogenin and MyoD1 expression in pae- cular diagnosis in childhood rhabdomyosarcoma. Diagn Mol Pol.
diatric rhabdomyosarcomas. J Clin Pathol. 2003;56(6):412416. 2000;9(1):913.
128. Seibold H. Juvenile alveolar rhabdomyosarcoma in a dog. Vet 146. Tonin P, Scrable H, Shimada H, et al. Muscle-specific gene
Pathol. 1974;11(6):558560. expression in rhabdomyosarcomas and stages of human fetal ske-
129. Seidal T, Kindblom LG, Angervall L. Alveolar and poorly letal muscle development. Cancer Res. 1991;51(19):51005106.
differentiated rhabdomyosarcoma: a clinicopathologic, light- 147. Torbeck RL, Kittleson SL, Leathers CW. Botryoid rhabdomyo-
microscopic, ultrastructural and immunohistochemical analysis. sarcoma of the uterus of a filly. J Am Vet Med Assoc. 1980;176
APMIS. 1988;96(9):825838. (9):914916.
130. Selting KA, Powers BE, Thompson LJ, et al. Outcome of dogs 148. Tostar U, Toftgrd R, Zaphiropoulos PG, et al. Reduction of
with high-grade soft tissue sarcomas treated with and without human embryonal rhabdomyosarcoma tumor growth by inhibi-
adjuvant doxorubicin chemotherapy: 39 cases (19962004). tion of the hedgehog signaling pathway. Genes Cancer. 2010;1
J Am Vet Med Assoc. 2005;227(9):14421448. (9):941951.
131. Senior DF, Lawrence DT, Gunson C, et al. Successful treatment 149. Tsokos M, Webber BL, Parham DM, et al. Rhabdomyosarcoma: a
of botryoid rhabdomyosarcoma in the bladder of a dog. J Am new classification scheme related to prognosis. Arch Pathol Lab
Anim Hosp Assoc. 1993;29(5):386390. Med. 1992;116(8):847855.
132. Simon D, Gruber AD, Hewicker-Trautwein M, et al. Pathological 150. Tsukada T, McNutt MA, Ross R, et al. HHF35, a muscle actin-
femoral fracture due to rhabdomyosarcoma in a cat. J Small Anim specific monoclonal antibody, II: reactivity in normal, reactive,
Pract. 2000;41(12):566570. and neoplastic human tissues. Am J Pathol. 1987;127(2):
133. Skalli O, Gabbiani G, Babai F, et al. Intermediate filament pro- 389402.
teins and actin isoforms as markers for soft tissue tumor differen- 151. Turnquist S, Pace L, Keegan K, et al. Botryoid rhabdomyosar-
tiation and origin, II: rhabdomyosarcomas. Am J Pathol. 1988; coma of the urinary bladder in a filly. J Vet Diagn Invest. 1993;
130(3):515531. 5(3):451453.
134. Snyder LA, Michael H. Alveolar rhabdomyosarcoma in a juve- 152. Tursi M, Martinetti M, Gili S, et al. Myocardial adenomatoid
nile Labrador Retriever: case report and literature review. J Am tumor in eight cattle: evidence for mesothelial origin of bovine
Anim Hosp Assoc. 2011;47(6):443446. myocardial epithelial inclusions. Vet Pathol. 2009;46(5):
135. Soilleux EJ, Davies DR. Epithelial cyst of the cardiac papillary 897903.
muscle: case report and review of the literature. J Clin Pathol. 153. Ueno H, Kadosawa T, Isomura H, et al. Perianal rhabdomyosar-
2006;59(11):12031205. coma in a dog. J Small Anim Pract. 2002;43(5):217220.
136. Sorensen PHB, Lynch JC, Qualman SJ, et al. PAX3-FKHR and 154. van der Loop FT, Bosma AA, Vos JH, et al. Cultured pig rhabdo-
PAX7-FKHR gene fusions are prognostic indicators in alveolar myosarcoma cells with a deletion of the Xq24-qter chromosome
rhabdomyosarcoma: a report from the childrens oncology group. region: an immunochemical and cytogenetic characterization. Am
J Clin Oncol. 2002;20(11):26722679. J Vet Res. 1995;56(8):10621069.
826 Veterinary Pathology 55(5)

155. van Vechten M, Goldschmidt MH, Wortman JA. Embryonal 162. Worley G, Jr, Gorham JR. The comparative pathology of rhabdo-
rhabdomyosarcoma of the urinary bladder in dogs. Comp Cont myosarcoma with a report of a case in a dog. Am J Pathol. 1954;
Educ Pract. 1990;12(6):783793. 30(4):837843.
156. Villamil JA, Henry CJ, Bryan JN, et al. Identification of the most 163. Xia S, Pressey J, Barr F. Molecular pathogenesis of rhabdomyo-
common cutaneous neoplasms in dogs and evaluation of breed sarcoma. Cancer Biol Ther. 2002;1(2):97104.
and age distributions for selected neoplasms. J Am Vet Med 164. Yamate J, Murai F, Izawa T, et al. A rhabdomyosarcoma arising
Assoc. 2011;239(7):960965. in the larynx of a dog. J Toxicol Pathol. 2011;24(3):179182.
157. Vos JH, Borst GHA, Martin de las Mulas J, et al. Rhabdomyo- 165. Yang Z, Macquarrie KL, Analau E, et al. MyoD and E-protein
sarcomas in young pigs in a swine breeding farm: a morphologic heterodimers switch rhabdomyosarcoma cells from an arrested
and immunohistochemical study. Vet Pathol. 1993;30(3): myoblast phase to a differentiated state. Gene Dev. 2009;23(6):
271279. 694707.
158. Wachtel M, Runge T, Leuschner I, et al. Subtype and prognostic 166. Yanoff SR, Fuentealba C, Boothe HW, et al. Tracheal defect and
classification of rhabdomyosarcoma by immunohistochemistry. embryonal rhabdomyosarcoma in a young dog. Can Vet J. 1996;
J Clin Oncol. 2006;24(5):816822. 37(3):172173.
159. Weinstein MJ, Carpenter JL, Schunk CJ. Nonangiogenic and 167. Yhee J, Kim D, Hwang D, et al. Hematogenous metastasis of
nonlymphomatous sarcomas of the canine spleen: 57 cases embryonal rhabdomyosarcoma originating from skeletal muscle
(19751987). J Am Vet Med Assoc. 1989;195(6):784788. in a young dog. J Vet Diagn Invest. 2008;20(2):243246.
160. Williamson D, Missiaglia E, de Reynis A, et al. Fusion gene- 168. Yusuf F, Brand-Saberi B. Myogenesis and muscle regeneration.
negative alveolar rhabdomyosarcoma is clinically and molecu- Histochem Cell Biol. 2012;138(2):187199.
larly indistinguishable from embryonal rhabdomyosarcoma. 169. Zhang J. Selective usage of D-type cyclins by Ewings tumors
J Clin Oncol. 2010;28(13):21512158. and rhabdomyosarcomas. Cancer Res. 2004;64(17):60266034.
161. Wilson DW, Dungworth DL. Tumors of the respiratory tract. In: 170. Zuckerman V, Wolyniec K, Sionov RV, et al. Tumour suppression
Meuten DJ, ed. Tumors in Domestic Animals. 4th ed. Ames, IA: by p53: the importance of apoptosis and cellular senescence.
Blackwell; 2002:365399. J Pathol. 2009;219(1):315.

You might also like