<<

Pathologic Classification of and Correlations with Molecular Studies David M. Parham, M.D. Department of Pathology, Arkansas Children’s Hospital and University of Arkansas for Medical Sciences, Little Rock, Arkansas

veloping embryo, yielding clues to the biology of , the most common soft-tissue these lesions. For this reason, it is more correct to malignancy of childhood and adolescence, com- define rhabdomyosarcoma as a tumor derived from prises a group of differing pathobiologic entities primitive and exhibiting a profound linked by their common propensity for formation of tendency towards myogenesis than to define it as a neoplastic , a feature that results arising from skeletal muscle. The former from biological forces related to aberrant transcrip- concept was eloquently expressed by Masson (2), tion signals and the resultant production of myo- who referred to rhabdomyosarcomas as “rhabdo- genic proteins. At a molecular level, however, rhab- poietic ,” and it better explains the obser- domyosarcomas form a heterogeneous group that vation that more tumors arise from viscera and can be subdivided into distinct clinicopathologic axial soft tissues than from the extremities (3). His- entities based on morphologic appearance and ge- torically, this is a well-established phenomenon, netic makeup. These differing morphologic features considering Stafford’s report in 1839 (4) of a prob- were recognized in the mid-1900s by Horn and En- able prostatic rhabdomyosarcoma in a 5-year-old terline with their division of rhabdomyosarcomas child, and the first series of these tumors, reported into embryonal, alveolar, botryoid, and pleomor- in 1894 (5), in which the majority of cases involved phic subtypes. More recent histologic and biologic the urogenital tract. studies have resulted in description of additional entities, such as spindle and anaplastic rhabdo- , and refinements in recognition of the Standard Classification original entities, such as solid-alveolar rhabdomyo- Ironically, it was not the striking histologic differ- . Familiarity with newer classifications and entiation of rhabdomyosarcomas that led to Guer- their relationship to molecular aberrations is key to sant’s (6) description of the first reported subcate- stratifying patients on current therapeutic protocols gory of these but the gross appearance and proposed innovative genetic therapies. of a tumor arising from the of a young girl. This polypoid lesion resembled a bunch of grapes, KEY WORDS: Classification, Cytogenetics, Molecu- leading to its designation as a , lar biology, Pathology, Review, Rhabdomyosarcoma. from the Greek term botryos. Botryoid rhabdomyo- Mod Pathol 2001;14(5):506–514 sarcoma, the current name for sarcoma botryoides, now comprises a clinically and pathologically dis- Although relatively rare in adults, rhabdomyosarco- tinctive form of rhabdomyosarcomas. These lesions mas are the most common soft-tissue malignancy should abut an epithelial surface, such as that of the in children and adolescents, in whom they com- bladder, bile duct, vagina, or conjunctiva, and prise approximately 60% of sarcomas reported per project into the lumen as multinodular excres- annum (1). In many ways, the of these cences of variable size. Intriguingly, a similar phe- neoplasms is analogous to myogenesis in the de- nomenon occurs in rhabdomyomatous Wilms tu- mors that involve the renal pelvis (7, 8). The microscopic sine qua non of botryoid rhabdomyo- Copyright © 2001 by The United States and Canadian Academy of Pathology, Inc. sarcoma, prerequisite for the diagnosis by the cur- VOL. 14, NO. 5, P. 506, 2001 Printed in the U.S.A. rent classification (9), is the cambium layer (Fig. 1). Date of acceptance: January 22, 2001. Presented at the Pathology Committee Symposium, Children’s This subepithelial condensation of tumor cells de- Group, Tampa, Florida, April 14, 2000. rives its name from the from the cambium layer of Address reprint requests to: David M. Parham, M.D., Slot 820, Arkansas Children’s Hospital, 800 Marshall Street, Little Rock, AR 72202; e-mail: plants, a compressed subsurface zone of cells in [email protected]; fax: 501-320-3912. stems and roots that gives rise to phloem and xy-

506 FIGURE 1. Botryoid rhabdomyosarcoma. A small-cell abuts FIGURE 5. Anaplastic rhabdomyosarcoma, diffuse. Anaplastic an epithelial surface, with condensation of tumor cells in the rhabdomyosarcomas are characterized by cells with enlarged, immediate subepithelial zone. Reprinted from Parham DM, editor: pleomorphic, hyperchromatic nuclei. In this example, they are arrayed Pediatric Neoplasia: Morphology and Biology. Philadelphia: Lippincott- in a discrete cluster, indicative of the clonal proliferation that defines Raven; 1996, by permission of Lippincott-Williams and Wilkins. diffuse . Note the multipolar , also frequently seen in these neoplasms. Reprinted from Parham DM, editor: Pediatric Neoplasia: Morphology and Biology. Philadelphia: Lippincott-Raven; 1996, by permission of Lippincott-Williams and Wilkins.

FIGURE 2. Embryonal rhabdomyosarcoma. This tumor contains dense condensations of rhabdomyoblasts amid foci with a loose myxoid stroma. Reprinted from Parham DM, editor: Pediatric Neoplasia: Morphology and Biology. Philadelphia: Lippincott-Raven; 1996, by permission of Lippincott-Williams and Wilkins. FIGURE 6. Spindle cell rhabdomyosarcoma (type A lesion). This neoplasm contains relatively differentiated spindle cells having cytologic features reminiscent of tumors. In the Palmer classification, spindle cell rhabdomyosarcomas were known as type A lesions. Reprinted from Parham DM, editor: Pediatric Neoplasia: Morphology and Biology. Philadelphia: Lippincott-Raven; 1996, by permission of Lippincott-Williams and Wilkins.

FIGURE 3. Alveolar rhabdomyosarcoma. Aggregates of primitive round cells appear to “float” within nests outlined by fibrous septa lined tumor cells in a picket row configuration. Reprinted from Parham DM, editor: Pediatric Neoplasia: Morphology and Biology. Philadelphia: Lippincott-Raven; 1996, by permission of Lippincott-Williams and Wilkins.

FIGURE 7. Embryonal sarcoma (International Society of Pediatric Oncology [SIOP] classification). In the SIOP classification, this tumor would qualify as an embryonal sarcoma because of its lack of discrete histologic evidence of myogenesis. However, with the International Classification, these tumors are included with embryonal rhabdomyosarcomas. Reprinted from Parham DM, editor: Pediatric Neoplasia: Morphology and Biology. Philadelphia: Lippincott-Raven; 1996, by permission of Lippincott-Williams and Wilkins.

lem. Whether botryoid rhabdomyosarcomas are a FIGURE 4. Alveolar rhabdomyosarcoma, solid variant biologically distinct entity or a site-specific variant (monomorphous round cell tumor of Palmer). The neoplasm is composed of a densely cellular sheet of primitive rhabdomyoblasts with of embryonal rhabdomyosarcomas remains a sub- monomorphous, round nuclei and no discernible septa. This tumor is ject of debate, and biologic studies to date have not also illustrative of the monomorphous round cell category of the yielded a molecular distinction. Palmer classification. Reprinted from Parham DM, editor: Pediatric Neoplasia: Morphology and Biology. Philadelphia: Lippincott-Raven; Microscopic observations led to description of 1996, by permission of Lippincott-Williams and Wilkins. embryonal rhabdomyosarcoma, the next variant of

Classification of Rhabdomyosarcomas (D. M. Parham) 507 rhabdomyosarcoma to be discovered. This was so inition to deal with histologically indistinguishable, named in 1894 by Berard (10), who termed it a undifferentiated, small round cell tumors. Thus, tumeur embryonnaire du muscle striae. The histo- Riopelle and Theriault focused on a key defining logic features of this tumor, with its zones of loose pattern, the alveolar structure formed by the fi- and dense cellularity, remarkably recapitulate nor- brous septa of these neoplasms, which offered a mal embryonal myogenesis, in which loose primi- practical, reproducible means of diagnosis (Fig. 3). tive mesenchyme condenses to form nascent mus- These fibrous septa are composed of collagenous cle (Fig. 2). The tumor cells composing embryonal fibrovascular tissue that forms a scaffolding sus- rhabdomyosarcomas variably exhibit all cellular pending attached rhabdomyoblasts and circum- phases of myogenesis, from stellate undifferenti- scribing central clusters of tumor cells that appear ated mesenchymal cells to elongated myoblasts, to float in alveolar spaces. Of note is that in these multinucleated myotubes, and fully differentiated neoplasms, Riopelle and Theriault described solid myofibers. The latter phases become more preva- areas lacking fibrosis and resembling , a lent after combination and irradia- phenomenon further noted by Enzinger and Shiraki tion, which induces terminal differentiation (11). As (19) in one half of the AFIP series and more exten- a general rule, embryonal rhabdomyosarcomas sively used by recent classifications that recognize a arise from the genitourinary tract, head and neck, solid pattern in so-called alveolar tumors. and abdomen and affect infants and young chil- Drawing from the key reports described above dren. Thus, both histologically and clinically, they and using their own series of 39 cases, in 1958, Horn share features with other embryonal neoplasms of and Enterline (20) derived the first commonly used childhood, such as Wilms tumors, hepatoblasto- subclassification system for rhabdomyosarcomas mas, pancreatoblastomas, and . (Table 1). This system came into common use in They similarly occur as part of the neoplastic spec- the latter part of the 1900s and appeared to work trum of the Beckwith-Wiedemann syndrome, a fa- well from a clinicopathologic standpoint, as it pre- milial lesion with biologic implications for epige- dictably defined tumor types likely to occur in given netic inheritance and imprinting (12, 13). age ranges and anatomic sites. Using univariate In 1946, Stout, one of the premier soft-tissue pa- analysis of Intergroup Rhabdomyosarcoma Study thologists of the twentieth century, made the initial (IRS) material, Newton et al. (17) demonstrated the description of the next published variant of rhab- potential prognostic value of standard rhabdomyo- domyosarcoma, the pleomorphic subtype (14). Ple- sarcoma classification. However, even luminaries omorphic rhabdomyosarcomas arise almost exclu- such as Stout and Lattes (21) felt that subclassifica- sively in the soft tissues of adults and comprise part tion had little bearing on survival, and in one report of the spectrum of lesions known as “malignant (22), Gonzalez-Crussi and Black-Schaffer lamented fibrous ” (15). They form spindle cell that it was based on poorly defined criteria, resulted lesions with a whorled or storiform pattern and in marked disparities in studies, and disregarded containing cells with enlarged, hyperchromatic nu- morphologic heterogeneity. Thus, the stage was set clei. Because the tumor cells only rarely display for further refinements in rhabdomyosarcoma frank myogenesis, there has been controversy as to classification. the very existence of this neoplasm, but immuno- histochemical studies have confirmed their myoge- netic potential and verified their nosologic status (16). In the rare pleomorphic rhabdomyosarcoma Cytohistological (Palmer) Classification seen in the pediatric age group, there is usually The cytohistological classification promulgated another component (17), so that this category cur- by Palmer et al. began as a retrospective investiga- rently is not used for pediatric subtyping and has tion of IRS-I and -II material maintained at the IRS been subsumed into the anaplastic and spindle cell Pathology Center. Beckwith and Palmer (23) had subtypes (see later discussion). published a landmark histopathologic study that In 1956 (18), Riopelle and Theriault described the described the prognostic features of pediatric renal final category of traditionally recognized rhab- tumors, and Palmer applied similar methods to the domyosarcomas, alveolar rhabdomyosarcoma. Us- study of rhabdomyosarcoma. In this manner, ing only six cases of their own and comparing them Palmer used no preconceptions about rhabdomyo- with other published tumors, they engendered a sarcoma histology, but rather he catalogued a series new species of small-cell tumor that was distinctive of recurring cytologic appearances and tested their from similar lesions tending to arise in adolescents, value using statistical analysis. His resultant classi- such as lymphoma and Ewing sarcoma. In those fication is listed in Table 1. Palmer found his clas- halcyon days preceding the advent of immunohis- sification to be more predictive than the Horn and tochemistry, pattern recognition played a solo role Enterline classifications performed by the IRS pa- in pathologic diagnosis, but it lacked sufficient def- thologists, and he and Foulkes estimated that the

508 Modern Pathology TABLE 1. Classifications of Rhabdomyosarcomas, with Comparisons among Schemae

Horn-Enterline (20) Palmer (24)a SIOP (27) NCI (29) International (9) Embryonal “Mixed” Embryonal Embryonal Embryonal Type A Dense, well differentiated Leiomyomatous Spindle cell Dense, poorly differentiated Not otherwise specified Loose, nonbotryoid Botryoid Loose, botryoid Botryoid Botryoid Embryonal sarcoma Anaplastic Pleomorphic embryonal Alveolar Monomorphous round cell Alveolar Alveolar Alveolar (includes solid variant) Solid variant Anaplastic “Mixed?” Pleomorphic Anaplastic Pleomorphic Pleomorphic Rhabdomyosarcoma, not otherwise specified Sarcoma, not otherwise specified Undifferentiated sarcoma SIOP, International Society for Pediatric Oncology; NCI, National Cancer Institute. a Mixed tumors in the Palmer classification might equally apply to alveolar and embryonal rhabdomyosarcomas, as his category is independent of histology; most, however, are embryonal. Anaplasia might also be seen in both .

presence of unfavorable histology increased the risk embryonal sarcomas becomes necessary when one of death by a factor of 3.77 (24). adds muscle immunostains to the diagnostic arma- Palmer’s cytohistologic classification was based mentarium, as they are usually positive in these on nuclear morphology rather than cytoplasmic tumors, and chemotherapy also induces the tumor differentiation. As such, his work foreshadowed ob- cells to exhibit myogenic differentiation (28). As a servations made in subsequent newer classifica- result, embryonal sarcomas are considered a prim- tions that refined the standard Horn-Enterline sys- itive form of embryonal rhabdomyosarcoma in our tem. Among these findings were that tumors current classification (9). composed exclusively of small round cells (mono- In the initial SIOP review, superior outcomes morphous round cell rhabdomyosarcoma; Fig. 4) were seen in three subtypes of rhabdomyosarcoma: were bad actors, that neoplasms with large anaplas- loose botryoid, embryonal NOS, and dense well- tic cells (anaplastic rhabdomyosarcoma; Fig. 5) con- differentiated embryonal. In general, well- noted a bad prognosis, and that - differentiated tumors did better than alveolar and like lesions (Type A rhabdomyosarcomas; Fig. 6) poorly differentiated ones. To put these results into were relatively low grade. Unfortunately, Palmer et perspective with those of the other classifications, it al.’s work was never published in a refereed journal, should be noted that SIOP alveolar tumors were of only as meeting abstracts (24–26). the standard variety with fibrous septa but that solid alveolar tumors had been included with em- bryonal lesions as per the Horn-Enterline classifi- SIOP Classification cation. One may thus assume that a proportion of Using methods similar to those of Palmer, in the poorly differentiated embryonal rhabdomyo- 1989, Caillaud et al. (27) published a classification sarcomas in the SIOP classification might corre- used to test a pediatric sarcoma registry from a spond to solid alveolar rhabdomyosarcomas of the large multi-institutional study, that of the Interna- National Cancer Institute (NCI) classification, as tional Society of Pediatric Oncology (SIOP). The IRS described below. Of interest is that the primitive and SIOP studies diverged, however, in that the embryonal sarcomas fared better than standard SIOP group concentrated on cytologic differentia- embryonal rhabdomyosarcomas in a subsequent tion and cellular density rather than nuclear fea- SIOP review (28). tures (see Table 1). In this manner, embryonal rh- abdomyosarcomas were subdivided into loose or dense, botryoid or non-botryoid, poorly differenti- National Cancer Institute (NCI) Classification ated or well differentiated, or not otherwise speci- The recognition of solid alveolar rhabdomyosar- fied (NOS). Alveolar and pleomorphic tumors re- coma represented the major revision made by the tained the conventional definitions of Horn and NCI classification (Table 1), published in 1990 by Enterline, and a new category, embryonal sarcoma Tsokos et al. (29). As stated above, in many ways, (Fig. 7), arose; these sarcomas were characterized the inclusion of solid alveolar tumors lacking fi- by their low-power similarity to embryonal rhab- brous septa into the alveolar category was a rein- domyosarcomas but high-power lack of obvious vention of the original intent (Fig. 4). Riopelle and myogenesis by routine staining (28). Redefinition of Theriault (18) had noted that these foci occurred

Classification of Rhabdomyosarcomas (D. M. Parham) 509 often at the periphery of the lesions, and Enzinger categories of rhabdomyosarcoma, not otherwise and Shiraki’s AFIP series (19) had contained these specified, and sarcoma, not otherwise specified, areas in one half of alveolar tumors. The key to were respectively retained for cases in which rhab- recognition of alveolar tumors was the round, domyosarcoma could be diagnosed but not sub- monomorphous, lymphoma-like, nuclear cytology, typed or in which the diagnosis was strongly sus- as had been presciently noted by Palmer and pected but not verifiable because of suboptimal Foulkes (24). In a retrospective study of the NCI biopsy material (9). The IRS Pathologic Review classification using a large cohort of rhabdomyosar- Committee (now part of the Children’s Oncology comas treated at the NCI and St. Jude Children’s Group) has continued to use this system to the Research Hospital, the clinical outcome of solid present time (33). tumors paralleled that of typical alveolar rhab- The basic rule for application of the International domyosarcomas. In addition, NCI classification was Classification is that the key diagnostic features an independent prognostic factor in multivariate used for embryonal and alveolar variants of rhab- analysis, whereas conventional Horn-Enterline his- domyosarcoma, as described above, have been re- tology was predictive of outcome only in univariate tained. The category of alveolar rhabdomyosarco- statistical study. mas been expanded to include solid variants (see Subcategories of embryonal histology also con- earlier in this article) that lack fibrous septa (Fig. 4). stituted part of the NCI classification; these in- Botryoid rhabdomyosarcoma has also been re- cluded pleomorphic embryonal, leiomyomatous tained as a diagnostic category, with features as embryonal, and embryonal with aggressive foci described above, with the caveat that a cambium (29). By statistical analysis, these subclassifications layer (Fig. 1) is essential for diagnosis. Finally, a new of embryonal rhabdomyosarcoma did not appear category, spindle cell rhabdomyosarcoma, has been to affect clinical outcome in the combined NCI/St. added, with features as described below. Because of Jude review. pleomorphic rhabdomyosarcoma’s rarity in chil- Categorization of rhabdomyosarcomas was tre- dren and its overlap with anaplastic rhabdomyosar- mendously aided by a major discovery made in the coma (see below), it is not included in the Interna- early 1980s, namely, that alveolar rhabdomyosar- tional Classification. coma is cytogenetically characterized by a recipro- One feature of particular note is that some rhab- cal translocation, the t(2;13)(q35;q14). Using this domyosarcomas contain histologic features of mul- karotypic aberration, which appeared unique to al- tiple subtypes. In current classification parlance, veolar tumors, it was possible to prove that solid- the presence of any alveolar element translates into alveolar rhabdomyosarcomas possess biologic as a bad prognosis (34). The biologic basis for these well as clinicopathologic identity with typical alve- mixed tumors is currently unknown, although some olar tumors (30). studies suggest that even the embryonal elements of “bad” tumors have genetic features of alveolar rhabdomyosarcoma (35, 36). Interestingly, alveolar International Classification rhabdomyosarcomas frequently show amplifica- As this plethora of subcategorization arose, it be- tion of MYCN, a phenomenon observed only in the came evident that rhabdomyosarcoma classifica- alveolar component of mixed tumors (37). tion was heading down the same muddy road of confusion that had been plaguing lymphoma clas- sification. Thus, using the model developed by the Newer Entities Working Classification Group for Non-Hodgkin’s Inclusion of one new diagnostic subcategory of Lymphoma (31), Newton and colleagues at the IRS rhabdomyosarcoma, spindle cell rhabdomyosar- undertook a similarly promethean task, that of test- coma, was deemed necessary when the Interna- ing each of the systems described above and au- tional Classification was devised. Taking their cue thoring a new international classification. The ulti- from the older studies of Palmer of “type A” supe- mate goal of this group was to stratify rior prognosis tumors (25, 26), Cavazzana and col- rhabdomyosarcoma into prognostically significant leagues (38) in 1992 published a retrospective anal- and diagnostically consistent morphologic sub- ysis of these lesions, which they renamed “spindle groups. To this end, Newton and colleagues assem- cell rhabdomyosarcomas.” Using their own clinical bled a panel of experts representing each classifi- data and morphologic studies, they confirmed the cation, tested the overall agreement reached among excellent clinical outcome of these lesions and these pathologists when using all schemas, and de- carefully described their histologic features. Spindle vised a new system based on the level of agreement cell rhabdomyosarcomas primarily arose in parat- and power of prognostic prediction (9, 32). The esticular locations and exhibited a whorled spindly result, an International Classification for Rhabdo- appearance akin to that of smooth muscle tumors myosarcoma, is shown in Table 1. The diagnostic (Fig. 6). Additional retrospective analyses were then

510 Modern Pathology performed by IRS pathologists, who reconfirmed myogenesis program (35, 42). Using this data, in the uniqueness of these tumors (39). They thus 1995, Downing and colleagues (43) developed a were included as a separate category in the Inter- molecular diagnostic test to detect this aberration national Classification (9). One histologic feature using reverse transcriptase–polymerase chain reac- noted in Cavazzana’s study (38) was strong immu- tion (RT-PCR). nopositivity for titin, a marker of terminal Another discovery was that an another member differentiation. of the PAX family, PAX7, fused with FRHR in an Another new entity that was considered for inclu- alternate translocation present in some alveolar sion in the International Classification but ulti- rhabdomyosarcoma, the t(1;13)(p36;q14). The im- mately not included was the anaplastic rhabdo- portance of this discovery has recently been height- myosarcoma. Anaplastic tumors had constituted a ened by indications that tumors with this translo- poor prognosis group of the Cytohistologic Classi- cation behave in a manner more akin to that of fication of Palmer, but their definition was some- embryonal tumors than alveolar ones (44), a phe- what poorly defined because it was based solely on nomenon perhaps related to differing mechanisms the presence of a single atypical multipolar mitosis. of transcription between the PAX 7/FRHR and PAX3/ A reanalysis of the slides and clinical data published FRHR fusions. A surprisingly large subset of alveolar in 1993 by the IRS Pathology Center (40) indicated rhabdomyosarcomas have appeared to show no ev- that enlarged, hyperchromatic nuclei akin to those idence of a translocation (43, 45); investigation of in anaplastic Wilms tumor (23) were also key to the the molecular nature of these fusion-negative alve- diagnosis. However, the critical factor in determin- olar rhabdomyosarcomas is in progress. ing the poor clinical outcome of these tumors ap- Unlike the case in alveolar tumors, efforts to dis- peared to be whether the anaplastic cells were ag- cover a single, key genetic mutation in embryonal gregated in a clone-like cluster or sprinkled in a rhabdomyosarcomas have been fruitless. It is ap- haphazard, single-cell fashion; the former feature parent that many of these tumors exhibit loss of appeared to be a bad-prognosis factor. Preliminary heterozygosity (LOH) at chromosome 11p, and this data from immunohistochemical studies indicates discovery by Scrable et al. (46) was used to pro- that the anaplastic cells contain abnormal (41). posed a classification based on molecular features Work on additional tumors is ongoing at the IRSG rather than histologic ones. LOH in this region links Pathology Center, and prospective data is accumu- embryonal rhabdomyosarcomas with other embry- lating to determine whether these tumors will re- onal neoplasms, such as Wilms tumors and hepa- tain their bad connotation and deserve special ther- toblastomas (47), and constitutional lesions such as apy. At present, they are not included in the the Beckwith-Wiedemann syndrome (48). A International Classification (Table 1). perhaps-related phenomenon is loss of genetic im- For the practicing pathologist, anaplasia is best printing in genes at this locus (49, 50). Although a recognized on low-power examination as areas of single genetic lesion cannot be identified, aberra- cellular and nuclear enlargement. Focusing on tions affecting the methylation status of key genes these areas should reveal hyperchromasia and ple- such as MyoD (51) and p21 (52) have been identi- omorphism in affected cells (Fig. 5). Although cur- fied. These findings suggest that the causation of rently not a critical feature, the presence of multi- embryonal rhabdomyosarcoma may be epigenetic polar mitoses is frequently noted. The significance rather than genetic, which would explain the asso- of these findings lies in the fact that a limited num- ciation of rhabdomyosarcoma with alterations of ber of reports has associated these cells with poor imprinting. This hypothesis might also explain the outcome, particularly if discrete, clonal populations frequent hyperdiploidy (53, 54) and chromosomal are seen. However, recognition of anaplasia does duplications (55, 56) seen in embryonal rhabdomy- not affect clinical management at present on IRSG osarcomas, as lesions affecting methylation genes protocols, pending accumulation of additional pro- can lead to these phenomena (57–59). spective data.

Modern-Day Diagnosis of Rhabdomyosarcoma Biologic Studies Related to Rhabdomyosarcoma Like classification, modern-day diagnosis of Classification rhabdomyosarcoma is often a difficult proposition. Work continued apace on characterization of the However, some of the tools we have learned to use t(2;13) translocation of alveolar rhabdomyosar- in classifying rhabdomyosarcoma are equally effec- coma, with the discovery that the genetic break- tive for ancillary diagnosis. In the 21st century, im- points resulted in a fusion of the genes PAX3 and munohistochemistry has attained ascendancy as FRHR (formerly ALV). The resultant chimeric pro- the foremost method of diagnostic confirmation. A tein causes aberrant transcription resulting in both variety of muscle markers have been identified; a unrestrained cellular division and initiation of a partial listing is seen in Table 2. Myoglobin, one of

Classification of Rhabdomyosarcomas (D. M. Parham) 511 TABLE 2. Immunohistochemical Stains Useful for (61). Ectomesenchymomas are defined by their Rhabdomyosarcoma Diagnosis mixture of muscle and neural elements (62). Some Type Marker aggressive rhabdomyosarcomas present with no Cytoplasmic nonfilamentous Myoglobin defined primary, extensive marrow involve- Creatine kinase M Cytoplasmic, filamentous ment, and even leukemic spread (63). Thus, it is Muscle-specific sometimes desirable to resort to molecular testing Sarcomeric with fusion markers or cytogenetics. In circum- Nuclear MyoD stances in which histologic features do not match immunohistochemical stains, I find it particularly helpful to use genetic studies and electron . the first markers identified, suffers from its relative Another problematic feature of rhabdomyosarco- lack of sensitivity, as only differentiated cells ex- mas arises with excisions or biopsies performed press it. Desmin and muscle-specific actin have after multimodality therapy. Treatment induces not been used most extensively, gaining much popular- only necrosis and shrinkage but also fibrosis and ity during the late 1980s. These filamentous pro- cytodifferentiation (64). The terminal differentia- teins are relatively sensitive, but they are also ex- tion so induced can be so striking that the neoplas- pressed by a variety of cells other than tic myoblasts recapitulate normal muscle, particu- rhabdomyoblasts, including smooth muscle, myo- larly entrapped, regenerative myofibers. In tumors fibroblasts, pericytes, and myoepithelium. Among involving skeletal muscle, an unresolvable Gordian sarcomas, peripheral neuroectodermal tumors knot may arise that cannot be completely solved (PNET), myofibrosarcomas, , ma- with current techniques. At present, the clinical lignant peripheral nerve sheath tumors, and des- significance of terminally differentiated tumor is moplastic small-cell tumors may express desmin unclear, although some limited, retrospective stud- and/or muscle actin. Perhaps the most sensitive ies would indicate that it is nontumorigenic (65). and specific group of proteins useful in immuno- The exquisite specificity of myoglobin in terminally histochemical diagnosis of rhabdomyosarcoma are differentiated myocytes makes it a useful marker of the muscle transcription factors MyoD and myoge- differentiated tumor in non–skeletal muscle- nin. These proteins, which define the earliest events containing tissues like or prostate. in molecular determination of myoblastic lineage, MyoD and myogenin are less helpful because their are paradoxically expressed at high levels in most expression is usually down-regulated with terminal rhabdomyosarcomas, even though the cells often differentiation. show little differentiation. Immunohistochemical techniques using heat retrieval allows use of these markers in paraffin sections, although this process SUMMARY tends to decrease staining and to introduce an ele- ment of nonspecific cytoplasmic positivity. Thus, It is apparent that rhabdomyosarcoma comprises only nuclear staining should be considered posi- a group of morphologically similar but biologically tive. The pattern of staining appears to predict clas- diverse lesions. The latest classification systems sification, as a heterogeneous pattern is more typ- represent attempts to synthesize clinically relevant ical of embryonal rhabdomyosarcoma, whereas subgroups based on histologic appearance and ver- strong, diffuse staining is a feature of alveolar rhab- ified by molecular genetic techniques. Using these domyosarcoma (60). tools, rhabdomyosarcomas can be subdivided into Other diagnostic approaches are appropriate superior, good, and bad prognosis groups. Work with some tumors, as the differential diagnosis of with some categories, such as anaplastic rhab- rhabdomyosarcoma comprises the entire breadth domyosarcomas, is ongoing. Evolution of these of small, round cell tumors, including neuroblas- ideas will doubtlessly progress as biologic and clin- toma, the (Ewing sarcoma icopathologic studies yield additional data. and PNET), lymphoma, and desmoplastic small cell tumor. Spindle cells also are common in rhab- domyosarcomas, prompting consideration of fibro- REFERENCES sarcoma and myofibrosarcoma, leiomyosarcoma, 1. Miller RW, Young JL Jr, Novakovic B. . malignant fibrous histiocytoma and its congeners, Cancer 1995(1 Suppl);75:395–405. malignant peripheral nerve sheath tumor (particu- 2. Masson P. Human tumors. Histology, diagnosis, and tech- larly Triton tumor), , and syno- nique. 2nd ed. Detroit, MI: Wayne State University Press; 1970. vial sarcoma. Occasional rhabdomyosarcomas dis- 3. Donaldson SS. Rhabdomyosarcoma. Contemporary status play a bewildering phenotype; even monoclonal and future directions: The Lucy Wortham James Clinical light-chain and B-cell marker positivity are possible Research Award. Arch Surg 1989;124:1015–20.

512 Modern Pathology 4. Stafford RA. A case of enlargement from melanoid tumor of childhood rhabdomyosarcoma: a report from the Interna- the prostate gland in a child five years of age. Med Chir Trans tional Society of Pediatric Oncology Pathology Panel. Med 1839;22:218. Pediatr Oncol 1989;17:391–400. 5. Wolfensberger R. Uber ein rhabdomyom der speiserohre. 28. Wijnaendts LCD, van der Linden JC, van Unnik AJM, Beitr Pathol Anat Allg Pathol 1894;15:491. Delemarre JFM, Voute PA, Meijer CJLM. Histologic classifi- 6. Guersant MP. Polypes du vagin chez une petite fille de treize cation of childhood rhabdomyosarcomas: relationship with mois. Monit Hopit 1854;2:187. clinical parameters and prognosis. Hum Pathol 7. Mahoney JP, Saffos RO. Fetal rhabdomyomatous nephro- 1994;25:900–7. blastoma with a renal pelvic mass simulating sarcoma bot- 29. Tsokos M, Webber B, Parham D, Wesley R, Miser A, Miser JS, ryoides. Am J Surg Pathol 1981;5:297–306. et al. Rhabdomyosarcoma: a new classification scheme re- 8. Parham DM. Pediatric neoplasia: morphology and biology. lated to prognosis. Arch Pathol Lab Med 1992;116:847–55. Philadelphia: Lippincott-Raven; 1996. 30. Parham DM, Shapiro DN, Downing JR, Webber BL, Douglass 9. Newton WA Jr, Gehan EA, Webber BL, Marsden HB, van EC. Solid alveolar rhabdomyosarcomas with the t(2;13): re- Unnik AJM, Hamoudi AB, et al. Classification of rhabdomyo- port of two cases with diagnostic implications. Am J Surg sarcoma and related sarcomas: pathologic aspects and pro- Pathol 1994;18:474–8. posal for a new classification—an Intergroup Rhabdomyo- 31. The Non-Hodgkin’s Lymphoma Pathologic Classification sarcoma Study. Cancer 1995;76:1073–85. Project. National Cancer Institute sponsored study of classi- 10. Berard M. Tumeur embryonnaire du muscle striae. Lyon fications of non-Hodgkin’s : summary and de- Med 1894;77:52. scription of a working formulation for clinical usage. Cancer 11. Parham DM. The molecular biology of childhood rhabdo- 1982;49:2112–35. myosarcoma. Semin Diag Pathol 1994;11:39–46. 32. Asmar L, Gehan EA, Newton WA, Webber BL, Marsden HB, 12. Reid LH, Davies C, Cooper PR, Crider-Miller SJ, Sait SN, van Unnik AJM, et al. Agreement among and within groups Nowak NJ, et al. A 1-Mb physical map and PAC contig of the of pathologists in the classification of rhabdomyosarcoma imprinted domain in 11p15.5 that contains TAPA1 and the and related childhood sarcomas: report of an international BWSCR1/WT2 region. Genomics 1997;43:366–75. study of four pathology classifications. Cancer 1994;74:2579– 13. Besnard-Guerin C, Newsham I, Winqvist R, Cavenee WK. A 88. common region of loss of heterozygosity in Wilms’ tumor 33. Qualman SJ, Coffin CM, Newton WA, Hojo H, Triche TJ, and embryonal rhabdomyosarcoma distal to the D11S988 Parham DM, et al. Intergroup Rhabdomyosarcoma Study. locus on chromosome 11p15.5. Hum Genet 1996;97:163–70. Update for pathologists. Pediatr Dev Pathol 1998;1:550–61. 14. Stout AP. Rhabdomyosarcoma of the skeletal muscles. Ann 34. Pappo AS, Shapiro DN, Crist WM, Maurer HM. Biology and Surg 1946;123:447–72. therapy of pediatric rhabdomyosarcoma. J Clin Oncol 1995; 15. Fletcher CDM. Pleomorphic malignant fibrous histiocytoma: 13:2123–39. Fact or fiction? A critical reappraisal based on 159 tumors 35. Tobar A, Avigad S, Zoldan M, Mor C, Goshen Y, Zaizov R. diagnosed as pleomorphic sarcoma. Am J Surg Pathol 1992; Clinical relevance of molecular diagnosis in childhood rhab- 16:213–28. domyosarcoma. Diagn Mol Pathol 2000;9:9–13. 16. Wesche WA, Fletcher CDM, Dias P, Houghton PJ, Parham 36. Anderson J, Renshaw J, McManus A, Carter R, Mitchell C, DM. Immunohistochemistry of MyoD1 in adult pleomorphic Adams S, et al. Amplification of the t(2;13) and t(1;13) trans- sarcomas. Am J Surg Pathol 1995;19:261–9. locations of alveolar rhabdomyosarcoma in small formalin- 17. Newton WA Jr, Soule EH, Hamoudi AB, Reiman HM, Shi- fixed biopsies using a modified reverse transcriptase poly- mada H, Beltangady M, et al. Histopathology of childhood merase chain reaction. Am J Pathol 1997;150:477–82. sarcomas, Intergroup Rhabdomyosarcoma Studies I and II: 37. Hachitanda Y, Toyoshima S, Akazawa K, Tsuneyoshi M. clinicopathologic correlation. J Clin Oncol 1988;6:67–75. N-myc gene amplification in rhabdomyosarcoma detected 18. Riopelle JL, Thériault JP. Sur une forme méconnue de sar- come des parties molles: le rhabdomyosarcome alvéolaire. by fluorescence in situ hybridization: its correlation with Ann Anat Pathol 1956;1:88–111. histologic features. Mod Pathol 1998;11:1222–7. 19. Enzinger FM, Shiraki M. Alveolar rhabdomyosarcoma: an 38. Cavazzana AO, Schmidt D, Ninfo V, Harms D, Tollot M, Carli analysis of 110 cases. Cancer 1969;24:18–31. M, et al. Spindle cell rhabdomyosarcoma: A prognostically 20. Horn RC, Enterline HT. Rhabdomyosarcoma: a clinicopath- favorable variant of rhabdomyosarcoma. Am J Surg Pathol ologic study of 39 cases. Cancer 1958;11:181–99. 1992;16:229–35. 21. Stout AP, Lattes R. Atlas of tumor pathology. 2nd series, 39. Leuschner I, Newton WA Jr, Schmidt D, Sachs N, Asmar L, Fascicle I: tumors of the soft tissues. Washington, DC: Armed Hamoudi A, et al. Spindle cell variants of embryonal rhab- Forces Institute of Pathology; 1967. domyosarcoma in the paratesticular region: A report of the 22. Gonzalez-Crussi F, Black-Schaffer S. Rhabdomyosarcoma of Intergroup Rhabdomyosarcoma Study. Am J Surg Pathol infancy and childhood: problems of morphologic classifica- 1993;17:221–30. tion. Am J Surg Pathol 1979;3:157–71. 40. Kodet R, Newton WA Jr, Hamoudi AB, Asmar L, Jacobs DL, 23. Beckwith JB, Palmer NF. Histopathology and prognosis of Maurer HM. Childhood rhabdomyosarcoma with anaplastic Wilms tumors: results from the First National Wilms’ Tumor (pleomorphic) features: a report of the Intergroup Rhabdo- Study. Cancer 1978;41:1937–48. myosarcoma Study. Am J Surg Pathol 1993;17:443–53. 24. Palmer NF, Foulkes M. Histopathology and prognosis in the 41. Pascasio J, Triche T, Sorensen P, Barr F, Qualman S. p53 second Intergroup Rhabdomyosarcoma Study (IRS II) [Ab- expression correlates with anaplasia in rhabdomyosarcoma stract]. Proc ASCO 1983;2:229. (RMS) [Abstract]. Mod Pathol 2000;11:4P. 25. Palmer NF, Sachs N, Foulkes M. Histology and prognosis in 42. Khan J, Bittner ML, Saal LH, Teichmann U, Azorsa DO, rhabdomyosarcoma: a report of the Intergroup Rhabdomyo- Gooden GC, et al. cDNA microarrays detect activation of a sarcoma Study [Abstract]. Proc SIOP 1981;13:113. myogenic transcription program by the PAX3-FKHR fusion 26. Palmer NF, Sachs N, Foulkes M. Histology and prognosis in . Proc Natl Acad SciUSA1999;96:13264–9. rhabdomyosarcoma (IRS-I) [Abstract]. Proc ASCO 1982;1: 43. Downing JR, Khandekar A, Shurtleff SA, Head DR, Parham 170. DM, Webber BL, et al. Multiplex RT-PCR assay for the dif- 27. Caillaud JM, Gérard-Marchant R, Marsden HB, van Unnik ferential diagnosis of alveolar rhabdomyosarcoma and Ew- AJM, Rodary C, Rey A, et al. Histopathologic classification of ing’s sarcoma. Am J Pathol 1995;146:626–34.

Classification of Rhabdomyosarcomas (D. M. Parham) 513 44. Kelly KM, Womer RB, Sorensen PH, Xiong QB, Barr FG. coma revealed by comparative genomic hybridization and Common and variant gene fusions predict distinct clinical fluorescence in situ hybridization: an intergroup rhabdo- phenotypes in rhabdomyosarcoma. J Clin Oncol 1997;15: myosarcoma study. Genes Chromosomes Cancer 2000;27: 1831–6. 337–44. 45. Kullendorff CM, Donner M, Mertens F, Mandahl N. Chro- 56. Lee W, Han K, Harris CP, Meisner LF. Detection of aneu- mosomal aberrations in a consecutive series of childhood ploidy and possible deletion in paraffin- embedded rhabdo- rhabdomyosarcoma. Med Pediatr Oncol 1998;30:156–9. myosarcoma cells with FISH. Cancer Genet Cytogenet 1993; 46. Scrable H, Witte D, Shimada H, Seemayer T, Wang-Wuu S, 68:99–103. Soukup S, et al. Molecular differential pathology in rhabdo- 57. Smith SS. Stalling of DNA methyltransferase in chromosome myosarcoma. Genes Chromosomes Cancer 1989;1:23–35. stability and chromosome remodelling. Int J Mol Med 1998; 47. Koufos A, Hansen MF, Copeland NG, Jenkins NA, Lampkin 1:147–56. BC, Cavenee WK. Loss of heterozygosity in three embryonal 58. Vilain A, Vogt N, Dutrillaux B, Malfoy B. DNA methylation tumours suggests a common pathogenetic mechanism. Na- and chromosome instability in breast lines. FEBS ture 1985;316:330–4. Lett 1999;460:231–4. 48. Sotelo-Avila C, Gooch WM. Neoplasms associated with the 59. Xu GL, Bestor TH, Bourc’his D, Hsieh CL, Tommerup N, Beckwith-Wiedemann syndrome. Perspect Pediatr Pathol Bugge M, et al. Chromosome instability and immunodefi- 1976;3:255–72. ciency syndrome caused by mutations in a DNA methyl- 49. Scrable H, Cavenee W, Ghavimi F, Lovell M, Morgan K, transferase gene. Nature 1999;402:187–91. Sapienza C. A model for embryonal rhabdomyosarcoma tu- 60. Dias P, Chen B, Dilday B, Palmer H, Hosoi H, Singh S, et al. morigenesis that involves genome imprinting. Proc Natl Strong immunostaining for myogenin in rhabdomyosar- Acad SciUSA1989;86:7480–4. coma is significantly associated with tumors of the alveolar 50. Zhan S, Shapiro DN, Helman LJ. Activation of an imprinted subclass. Am J Pathol 2000;156:399–408. allele of the insulin-like growth factor II gene implicated in 61. Pinto A, Tallini G, Novak RW, Bowen T, Parham DM. Undif- rhabdomyosarcoma. J Clin Invest 1994;94:445–8. ferentiated rhabdomyosarcoma with lymphoid phenotype 51. Chen B, Dias P, Jenkins JJ, Savell VH, Parham DM. Methyl- expression. Med Pediatr Oncol 1997;28:165–70. ation alterations of the MyoD1 upstream region are predic- tive of subclassification of human rhabdomyosarcomas. 62. Boue D, Parham D, Webber B, Maurer H, Qualman S. Clin- Am J Pathol 1998;152:1071–9. icopathologic study of pediatric ectomesenchymoma from 52. Chen B, He L, Savell VH, Jenkins JJ, Parham DM. Inhibition IRS III and IV [Abstract]. Mod Pathol 1997;10:2P. of the interferon-gamma/signal transducers and activators 63. Etcubanas E, Peiper S, Stass S, Green A. Rhabdomyosar- of transcription (STAT) pathway by hypermethylation at a coma, presenting as disseminated malignancy from an un- STAT-binding site in the p21WAF1 promoter region. Cancer known primary site: a retrospective study of ten pediatric Res 2000;60:3290–8. cases. Med Pediatr Oncol 1989;17:39–44. 53. Jung WH, Jung SH, Yoo CJ, Kim YJ, Park C, Kim BS. Flow 64. Coffin CM, Rulon J, Smith L, Bruggers C, White FV. Patho- cytometric analysis of DNA ploidy in childhood rhabdomyo- logic features of rhabdomyosarcoma before and after treat- sarcoma. Yonsei Med J 1994;35:34–42. ment: a clinicopathologic and immunohistochemical analy- 54. Niggli FK, Powell JE, Parkes SE, Ward K, Raafat F, Mann JR, sis. Mod Pathol 1997;10:1175–87. et al. DNA ploidy and proliferative activity (S-phase) in child- 65. Andrassy RJ, Wiener ES, Raney RB, Hays DM, Arndt CA, Lobe hood soft- tissue sarcomas: their value as prognostic indica- TE, et al. Progress in the surgical management of vaginal tors. Br J Cancer 1994;69:1106–10. rhabdomyosarcoma: a 25-year review from the Intergroup 55. Bridge JA, Liu J, Weibolt V, Baker KS, Perry D, Kruger R, et al. Rhabdomyosarcoma Study Group. J Pediatr Surg 1999;34: Novel genomic imbalances in embryonal rhabdomyosar- 731–4.

514 Modern Pathology