review article rhabdomyosarcoma: advances in molecular and

15
Review Article Rhabdomyosarcoma: Advances in Molecular and Cellular Biology Xin Sun, 1,2,3 Wei Guo, 3 Jacson K. Shen, 1,2 Henry J. Mankin, 1,2 Francis J. Hornicek, 1,2 and Zhenfeng Duan 1,2 1 Department of Orthopaedic Surgery, Massachusetts General Hospital, 100 Blossom Street, Boston, MA 02114, USA 2 Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital, 100 Blossom Street, Boston, MA 02114, USA 3 Department of Orthopaedic Oncology, Peking University People’s Hospital, 11 Xizhimen South Street, Xicheng District, Beijing 100044, China Correspondence should be addressed to Zhenfeng Duan; [email protected] Received 4 May 2015; Accepted 16 August 2015 Academic Editor: Enrique de Alava Copyright © 2015 Xin Sun et al. is is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Rhabdomyosarcoma (RMS) is the most common soſt tissue malignancy in childhood and adolescence. e two major histological subtypes of RMS are alveolar RMS, driven by the fusion protein PAX3-FKHR or PAX7-FKHR, and embryonic RMS, which is usually genetically heterogeneous. e prognosis of RMS has improved in the past several decades due to multidisciplinary care. However, in recent years, the treatment of patients with metastatic or refractory RMS has reached a plateau. us, to improve the survival rate of RMS patients and their overall well-being, further understanding of the molecular and cellular biology of RMS and identification of novel therapeutic targets are imperative. In this review, we describe the most recent discoveries in the molecular and cellular biology of RMS, including alterations in oncogenic pathways, miRNA (miR), in vivo models, stem cells, and important signal transduction cascades implicated in the development and progression of RMS. Furthermore, we discuss novel potential targeted therapies that may improve the current treatment of RMS. 1. Introduction Rhabdomyosarcoma (RMS) is the most prevalent soſt tissue tumor in children and adolescents, accounting for 5% of all pediatric tumors [1, 2]. It is estimated that 350 new cases of RMS are diagnosed each year in patients under 20 years of age in the United States [2]. In contrast, RMS is extremely rare in adults. ere is a slight male predominance (1.4 times more common in males than in females), but there are no significant differences in the incidence rates among races or different ethnic groups [3]. As RMS is derived from primitive mesenchymal stem cells directed towards myogenesis, it can arise in a variety of anatomic sites throughout the body [4]. RMS can occur either as a primary malignancy or as a com- ponent of a heterogeneous malignancy, such as a malignant teratomatous tumor [5]. Additionally, a small percentage of cases are associated with known genetic disorders, such as neurofibromatosis type 1 and the Li-Fraumeni familial cancer syndrome [6]. e World Health Organization (WHO) recently revised the classification of RMS subtypes as alveolar rhabdomyo- sarcoma (ARMS), embryonal rhabdomyosarcoma (ERMS), pleomorphic rhabdomyosarcoma (PRMS), and sclerosing/ spindle cell rhabdomyosarcoma (SRMS) in 2013 [7]. ARMS is a high-grade malignancy occurring mostly in adolescents and young adults. e most common site for ARMS is in the deep tissue of extremities. ERMS represents approximately 70% of all childhood RMS, usually afflicting infants or children under 10 years of age. ERMS oſten affects the head and neck regions, especially the orbit. PRMS usually occurs in adult males in the deep tissue of extremities but may occur at any site. In adult patients, the pleomorphic variant is associated with the worst prognosis [8]. Hindawi Publishing Corporation Sarcoma Volume 2015, Article ID 232010, 14 pages http://dx.doi.org/10.1155/2015/232010

Upload: others

Post on 16-Oct-2021

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Review Article Rhabdomyosarcoma: Advances in Molecular and

Review ArticleRhabdomyosarcoma Advances in Molecular andCellular Biology

Xin Sun123 Wei Guo3 Jacson K Shen12 Henry J Mankin12

Francis J Hornicek12 and Zhenfeng Duan12

1Department of Orthopaedic Surgery Massachusetts General Hospital 100 Blossom Street Boston MA 02114 USA2Sarcoma Biology Laboratory Center for Sarcoma and Connective Tissue Oncology Massachusetts General Hospital100 Blossom Street Boston MA 02114 USA3Department of Orthopaedic Oncology Peking University Peoplersquos Hospital 11 Xizhimen South Street Xicheng DistrictBeijing 100044 China

Correspondence should be addressed to Zhenfeng Duan zduanmghharvardedu

Received 4 May 2015 Accepted 16 August 2015

Academic Editor Enrique de Alava

Copyright copy 2015 Xin Sun et al This is an open access article distributed under the Creative Commons Attribution License whichpermits unrestricted use distribution and reproduction in any medium provided the original work is properly cited

Rhabdomyosarcoma (RMS) is the most common soft tissue malignancy in childhood and adolescence The two major histologicalsubtypes of RMS are alveolar RMS driven by the fusion protein PAX3-FKHR or PAX7-FKHR and embryonic RMS which isusually genetically heterogeneous The prognosis of RMS has improved in the past several decades due to multidisciplinary careHowever in recent years the treatment of patients with metastatic or refractory RMS has reached a plateau Thus to improve thesurvival rate of RMS patients and their overall well-being further understanding of the molecular and cellular biology of RMS andidentification of novel therapeutic targets are imperative In this review we describe the most recent discoveries in the molecularand cellular biology of RMS including alterations in oncogenic pathways miRNA (miR) in vivomodels stem cells and importantsignal transduction cascades implicated in the development and progression of RMS Furthermore we discuss novel potentialtargeted therapies that may improve the current treatment of RMS

1 Introduction

Rhabdomyosarcoma (RMS) is the most prevalent soft tissuetumor in children and adolescents accounting for 5 of allpediatric tumors [1 2] It is estimated that 350 new cases ofRMS are diagnosed each year in patients under 20 years ofage in the United States [2] In contrast RMS is extremelyrare in adults There is a slight male predominance (14 timesmore common in males than in females) but there are nosignificant differences in the incidence rates among races ordifferent ethnic groups [3] As RMS is derived from primitivemesenchymal stem cells directed towards myogenesis it canarise in a variety of anatomic sites throughout the body [4]RMS can occur either as a primary malignancy or as a com-ponent of a heterogeneous malignancy such as a malignantteratomatous tumor [5] Additionally a small percentage ofcases are associated with known genetic disorders such as

neurofibromatosis type 1 and the Li-Fraumeni familial cancersyndrome [6]

The World Health Organization (WHO) recently revisedthe classification of RMS subtypes as alveolar rhabdomyo-sarcoma (ARMS) embryonal rhabdomyosarcoma (ERMS)pleomorphic rhabdomyosarcoma (PRMS) and sclerosingspindle cell rhabdomyosarcoma (SRMS) in 2013 [7] ARMS isa high-grademalignancy occurringmostly in adolescents andyoung adultsThemost common site for ARMS is in the deeptissue of extremities ERMS represents approximately 70of all childhood RMS usually afflicting infants or childrenunder 10 years of age ERMS often affects the head and neckregions especially the orbit PRMS usually occurs in adultmales in the deep tissue of extremities but may occur at anysite In adult patients the pleomorphic variant is associatedwith the worst prognosis [8]

Hindawi Publishing CorporationSarcomaVolume 2015 Article ID 232010 14 pageshttpdxdoiorg1011552015232010

2 Sarcoma

Table 1 Alterations of chromosome in RMS by CGH in recent 5 years

Materials Gain Amplification Loss Deletion Reference

25 RMS samples 2p 12q 6p 9q 10q 1p2q 6q 8q 15q 18q 3p 11p 6p Li et al 2009 [16]

13 RMS cell lines

1p213ndash132 1q126q26ndash27 7q213ndash3111q41 2p243 8q241220q132 20q1332

2p243 (MYCN)8p1123ndash1121

(FGFR1) 12q133(CDK4) 19q12 20q

3p142ndash1224q27ndash3236p251ndash243

9p243ndash241 13q143

Missiaglia et al 2009[17]

57 ARMSsamples 12q15 2p24 12q13ndashq14 Barr et al 2009 [18]

128 primaryRMS samples 7 8 11 20

2p241 (MYCN)8p112ndashp111 (FGFR1)12q133ndash141 (CDK4)MDM2 (12q143ndashq15)

Williamson et al2010 [12]

26 frozenprimary ERMSsamples

8 2 11 12 13 19 20

2p21 2q35 2q1422q361 5q352ndashq353

11p112 11q24212q133

6 9 10 14 15 16 181p3623 1q321 3p1424q351ndash352 9p21317q112 22q1331

Paulson et al 2011[13]

39 RMS samples 12q133 12q133ndashq14112q141 17q251

2q1312 12q13312q133ndashq141

9p12ndashp11210q1121ndashq112214q3233 16p112

22q111

1p211 2q141 5q1329p12 9q12 Liu et al 2014 [11 15]

RMS cellsderived fromrefractory RMS

NACA HSD17B6SDR9C7 RDH16GPR182 ZBTB39TAC3 MYO1A

NAB2 STAT6 LRP1

Park et al 2014 [14]

20 RMS samplesof Chinesepatients

12q2431 17q2511q211 7q112312q133ndashq141

9p133 12q133ndashq14112q15 16p1311

5q132 15q11214q3233 (IGHG

IGHM)

1p3633 1p131 2q1115q132 8p231 9p243

16p112Liu et al 2014 [11 15]

Since the 1970s the Intergroup RhabdomyosarcomaStudy Group (IRSG) has conducted a series of clinical trialscomparing risk-base and has established a series of treatmentguidelines [9] Currently multidisciplinary managementincluding chemotherapy and surgery with or without radia-tion has become the standard treatment for RMS The 5-yearsurvival rate of RMS has increased from 25 in 1970 up to60 since 2000 [1 10]However there has been little improve-ment in the oncological outcome of patients with RMS inrecent years Drug resistance andmetastatic disease representthe two most common phenomena for therapy failure Somerandomized chemotherapy trials have failed to improve out-come despite the introduction of newer or more intensivetherapies Thus there is an urgent need for alternative moreeffective treatment strategies Recent molecular and geneticanalysis of these tumors has produced substantial newinsights into molecular cell biology molecular cytogeneticsand tumorigenesis of RMS leading to a better understandingof RMS development at the molecular level These advancesmay ultimately lead to better clinical understanding and topotentially developing more potent targeted therapies Thepurpose of this review is to summarize these most recentfindings in RMS

2 Novel Discoveries of ChromosomalAlterations in RMS

Malignant transformation occurs cytogenetically due to theaccumulation of somatic mutations by the acquisition oftumor-specific chromosomal translocations

21 Gains and Losses of Chromosomes in RMS ComparativeGenomic Hybridization (CGH) analysis has revealed that allRMS have specific gains and losses [11ndash18] (Table 1) ERMSfrequently exhibits gains or losses of specific whole chromo-somes whereas ARMS is characterized by the presence ofregions of genomic amplification [11 19] The focal regionsand genes most of which have frequent gains and ampli-fications include 12q133ndashq141 and 8p112ndash112 and CDK4MYCN GLI MDM2 FGFR1 and FGFR4 respectively Fre-quently the genes differentially expressed in subtypes ofRMS particularly when they are from chromosomal regionsshow a high level of gains in cell lines Previous CGH studieshave shown that ARMS tumors tend to have fewer copynumber variants than ERMS tumors [12 20] For examplefrequent gains were detected in TYROBP HCST LRFN3and ALKBH6 (19q1312) in ERMS but not in ARMS [15]

Sarcoma 3

PAX3

Chromosome 2

PAX3-FKHR

t(213)(q37q14)

NCOA1

PAX7

Chromosome 1

PAX7-FKHR

t(113)(p36q14)

PAX3

Chromosome 2

FKHR

Chromosome 13

PAX3-NCOA1

t(22)(q35q23)

Paired domain Homeodomain

DNA binding domain Transactivation domain

PB

PB

PB

PB HD

HD

HD

HD TAD

TAD

TAD

TAD

TAD

TAD

HD

HD

PB

PB

100 amino acids

Fusion site

Fusion site

FD

DNA binding heterodimerization domain Nuclear receptor interaction domain

LXXLL HAT

HAT

bHLHPAS

Fusion site

(a)

(b)

(c)

Figure 1The chromosomal rearrangements in ARMS 80 of ARMS classified as translocation-positive ARMS carry characteristic chromo-somal translocations demonstrated as t(213)(q35q14) t(113)(p36q14) and (22)(q35p23) In (a) and (b) the translocations fuse the FKHR(a member of the forkheadHNF-3 transcription factor family) locus on chromosome 13 to either PAX3 on chromosome 2 or PAX7 onchromosome 1 In (c) the translocation generated a fusion protein composed of PAX3 and the nuclear receptor coactivator NCOA1 havingsimilar transactivation properties as PAX3FKHR

These studies have identified a number of genetic alterationsin RMSMany of these chromosomal changesmay be respon-sible for tumor progression and proliferation These geneticalterations may be potential treatment targets in RMS [11]

22 Chromosomal Translocations in ARMS Chromosomalanalyses have demonstrated two translocations associatedwith ARMS t(213)(q35q14) and t(113)(p36q14) [21] Initialstudies detected these two gene fusions in 80 of ARMS[22] These characteristic chromosomal translocations areadjacent to the 51015840 DNA-binding domains of PAX a memberof the paired box transcription factor family and the trans-activation domain at the 31015840 end of FKHR a member of theforkheadHNF-3 transcription factor family Approximately75 of these structural rearrangements translocate the PAX3gene at 2q35 to the FKHR gene at 13q14 as t(213)(q35q14)less frequently in the other 25 the t(13)(q36q14) transloca-tion fuses PAX7 to FKHR [23] Previous studies have demon-strated that PAX3FKHR fusion gene status significantlyimproves current risk stratification while the presence ofPAX7FKHR and rarer variant fusion gene products requirefurther investigation [24] The remaining 20 of ARMS isPAX gene fusion-negative (PFN) and forms a more hetero-geneous group which remains a challenge to detect due to

the lack of consistent chromosomal rearrangements PFNARMS has a similar clinical course to ERMS which suggeststhat the fusion gene status provides more accurate informa-tion about patient outcomes than did the histologic subtypeDespite the low overall burden of somatic mutations infusion-positive RMSmultiple genes were recurrently alteredincluding NRAS KRAS HRAS FGFR4 PIK3CA CTNNB1FBXW7 and BCOR [25]

Another novel translocation t(22)(q35p23) was identi-fied in ARMS biopsy samples by gene expression signatures[26] The chromosomal translocation generates a fusion pro-tein composed of PAX3 and the nuclear receptor coacti-vator NCOA1 which has similar transactivation propertiesas PAX3FKHR [26] These biologic effects contribute totumorigenesis bymodulatingmyogenic differentiation alter-ing growth and apoptotic pathways and stimulating motilityand other metastatic pathways (Figure 1)

23 Chromosomal Alterations in ERMS ERMS exhibits a lossof imprinting (LOI) leading to a twofold gene dosage effect[27] Most tumors have at least one 15-Mb region with loss ofheterozygosity (LOH) along chromosome 11 [27] The allelo-type of ERMS demonstrates a high frequency of LOH onchromosomes 11p 11q and 16q [28] ERMS tumorigenesis

4 Sarcoma

Table 2 The histological types of rhabdomyosarcoma

Histological type Predilection population Predilection site Risk category Genetic change

ERMS Infants or children under 10years old Head and neck region Intermediate LOH at chromosome 11p155

ARMS Adolescents and young adults Deep tissue of extremity High t(213)(q35q14) t(113)(p36q14)

PRMS Adult males Throughout the body HighJUN (1p31) MYC (8q24)CCND1 (11q13) INT2 (11q133)MDM2 (12q143ndashq15)

SRMSIn the first decade of life with asecond mode centered aroundthe fifth decade

The extremities headand neck Superior SRF-NCOA2 MYOD1 mutation

ERMS embryonal rhabdomyosarcomaARMS alveolar rhabdomyosarcomaPRMS pleomorphic rhabdomyosarcomaSRMS spindle cellsclerosing rhabdomyosarcoma

can result from the inactivation of the parental bias of chro-mosome 11p15 which is the most common rearrangementin ERMS [29] The proportion of ERMS with LOH alongchromosome 11 is considerably higher than in other subtypes[27] In histopathological analysis ERMS expresses lowPAX3levels and elevated PAX7 levels [30] Hosoi et al identified ahidden 2q35 breakpoint as a novel PAX3 rearrangement incomplex chromosomal translocations in ERMS [31]

24 Chromosomal Alterations in Other RMS There are lim-ited studies on the biological pathways involved in othersubtypes of RMS compared with the two major subtypesARMS andERMS Fluorescence In SituHybridization (FISH)reveals amplification of JUN (1p31) MYC (8q24) CCND1(11q13) INT2 (11q133) MDM2 (12q143ndashq15) and MALT(18q21) in these tumor cells contributing to the pathogenesisof PRMS [32] MYOD1 homozygous mutations are alsoreported as frequent recurrent and pathognomonic eventsin adult-type SRMS [33 34] (Table 2) Furthermore four outof five pediatric tumors showedMYOD1mutations in a study[34] Yoshida et al described a 8q13 locus (NCOA2) generearrangement in a small subset of SRMS occurring uniquelyin the infantilecongenital setting fused with key transcrip-tion factors involved in skeletal muscle differentiation suchas SRF and TEAD1 [35] They also identified NCOA2 as acandidate PAX3 partner geneThePAX3-NCOA2 fusion geneplays a dual role in the tumorigenesis of RMS promotion ofthe proliferation and inhibition of the myogenic differenti-ation of RMS cells [35] As PAX3-NCOA2-induced tumorsgrow more slowly SRMS that are PAX3-NCOA2 fusion-positive could be associated with a very favorable prognosis[36]

Not all RMSs occur as sporadic primary tumors Occa-sionally RMS inherits a mutant gene as part of an establishedfamilial syndrome For example Beckwith-Wiedemann Syn-drome (BWS) with RMS involves dysregulation or alterationof imprinted genes in the 11p155 chromosomal regionincluding IGF2 H19 and CDKN1C (p57KIP2) [37]

3 Cell of Origin in RMS

There have been a number of studies aimed at deciphering thecell origin for RMS [56ndash61]

31 Myogenic Differentiation in the Tumorigenesis Severalstudies have focused on elucidating the mechanisms govern-ing the impaired myogenic program in RMS [62 63] (Fig-ure 2) RMSoriginates as a consequence of regulatory disrup-tion of the growth and differentiation of myogenic precursorcells Progenitor cells reside in muscle and their activationresults in either proper myogenesis or aberrant signalingpathways leading to the development of RMS Based on theskeletal muscle lineage a complete transcriptome analysis ofRMS was performed to compare normal and fetal muscles[56] The high degree of similarity between fetal muscle andRMS expression profiles reflects the undifferentiated myo-genic nature of RMS The genes exclusively upregulated inRMS including FGFR4 NOTCH2 UBE2C UHRF1 andYWHAB genes contribute to the failure of RMS cells to com-plete normal skeletal muscle development and progress toan alternative fate [56] RMS cells represent an arrested statein the development of normal skeletal muscle with regionalsilencing of differentiation factors leading to the maturationdefect in RMS [57] The expression pattern of muscle-specific proteins regulating myogenic differentiation hasbeen extensively examined in RMS The family of myogenictranscription factors (MYOD MYF5 myogenin and MRF4)are considered to be responsible for the determination of stemcells intomyoblasts anddifferentiation intomyocytes [58 59]All these factors are activated during the onset or progressionof myogenesis and are subsequently silenced to reach a finalmuscular differentiation There are statistical differences inthe different subtypes of RMS tumors with MYOD or myo-genin staining patterns Amplification of MDM2 in an RMScell line interferes with MYOD activity and consequentlyinhibits overt muscle cell differentiation [61] Interleukin-4receptor (IL-4R) has also been proposed to be important for

Sarcoma 5

Table 3 Studies identifying stem cells in RMS

Markersubstrate Source Stem cell gene Functional characterization Reference

CD133 Orthotopicxenograft model

OCT4 NANOG c-MYCPAX3 and SOX2

Correlating with poor overallsurvival

Walter et al2011 [38]

PAX-FKHR Bone marrow ofC57BL6 mice

Myf5 MEF2 MyoD andmyogenin

Determining the molecularmyogenic and histologicphenotype of ARMS

Ren et al 2008[39]

V-ATPase RD cell line NANOG and OCT34

Driving mechanisms of areduced sensitivity to anticancerdrugs and activities related toinvasion and metastasis

Salerno et al2014 [40]

Survm-CRAsKYM-1 cell line(FGFR3-positive)

FGFR3Therapeutic effectiveness againstall cell populations and increasedeffectiveness against CSCs

Tanoue et al2014 [41]

Migration

specification

Injured muscle

Terminal differentiation

Fusion

PAX7

Myoblasts

Myoblast

MyoD

MyoD

Myf5

PAX3

Rb

Ezh2

IGF2

HDAC4YY1

YY1

ERK

AKT

RMS

Myogenin

P38 kinase

Myostatin

Progenitor

cell

N-RAS

Myofibers

FGFR4

UBE2C

UHRF1

NOTCH2

YWHAB

Satellite cells

RUNX1 MEF2C

JDP2 NFIC

Myocytes

Restoration of proliferation blockade of

differentiation

Proliferatio

n

Cell circle arrestEarlydifferentiation

TGF-1205731

Figure 2 Myogenic pathways in the tumorigenesis of RMS In aberrant neoplastic condition progenitor cells residing in muscle result inaberrant pathways which lead to malignant transformation and fail to differentiate proliferate uncontrollably and form RMS Sharp arrows(rarr) indicate upregulationactivation and blunt arrows (perp) indicate downregulationinhibition

the maturation of myotubes [64] An IL-4R blockade mighthelp therapeutically to modulate the expression of myogenictranscription factors (MYOD or myogenin) [57 64]

32 Potential Cancer Stem Cell in RMS Cancer Stem Cells(CSCs) may contribute to inherent refractory responses tocurrent therapies andmetastasis [65]Many CSCmodels playan important role in the development of RMS [38ndash41 66](Table 3)

A study explored a potential important role for mesen-chymal stem cells (MSCs) as the cell of origin of ARMS [67]Ren et al confirmed that PAX-FKHR fusion genes commitMSCs to a myogenic lineage by inhibiting terminal differen-tiation and contributing to ARMS formation [39] Anotherstudy showed that PAX-FKHR induces skeletal myogenesisin MSCs by transactivating MYOD and myogenin and trans-forms mesenchymal progenitor cells to the skeletal muscle lin-eage leading to malignant formation resembling ARMS [39]

6 Sarcoma

METCDKN2AB

PAX37-FKHR

MDM2

RAS

Raf

GLI1

PDK1p53Rb

Hh

Myo MEF2

p21

Ptch

FGFR4

IGF1R

p16INK4Ap14ARF

p19ARF

IGF2

FGF

Figure 3 Genetic analyses of RMS have pinpointed several common alterations including inactivation of a master regulator of p53 andRb pathways CDKN2AB and activation of FGFR4 RAS and Hedgehog (Hh) signaling The modifications of these pathways influenceoncogenesis and metastatic potential

As a human hematopoietic CSC marker Walter et alproposed that upregulated CD133 in ERMS can act as aprognostic marker and might help with the development ofnovel targeted therapies for ERMS [38]

4 Signaling Pathway Alterations in RMS

Recent studies have looked into signaling pathway alterationsand their downstream effects in RMSThese new findings notonly helped to improve the understanding of this malignancybut also offered novel potential therapeutic strategies forimproved treatment for patientswithRMS [68 69] (Figure 3)

41 RAS Signaling Pathway RASmutations commonlymain-tain the protein in its GTP bound state and therefore renderit constitutively active in RMS Zhang et al identified RASfamily members NRAS KRAS and HRAS in the RASpathway as themost commonlymutated genes in ERMS [70]the mutation status of these genes is significantly associatedwith the risk of ERMS development [70] A study foundthat a majority of ERMS demonstrated activation of the RASpathway exclusively either by homozygous deletion of NF1or by point mutations in one of the RAS family members asdescribed above [13] Despite remarkable genetic and molec-ular heterogeneity most (93 4144) RMS tumors hijackeda common receptor tyrosine kinaseRASPIK3CA geneticaxis [25] It could occur via two alternative mechanismsrearrangement of a PAX gene and accumulation of mutationsthat were downstream targets of the PAX fusion protein

Skeletal muscle cells have a robust antioxidant defensesystem to protect the DNA lipids and proteins from thedeleterious effects of excess Reactive Oxygen Species (ROS)

Cancer cells also have elevated ROS due to their increasedmetabolic activity oncogenic stimulation andmitochondrialdysfunction These findings implicate RAS mutations andoxidative stress as potential therapeutic targets for high-riskERMS

42 IGF Signaling Pathway There is clear preclinical datathat supports the involvement of the Insulin-like GrowthFactor (IGF) signaling pathway in RMS tumorigenesis andprogression [69] Zhu and Davie demonstrated that IGFpromotes the proliferation of RMS cells while blocking IGFsignaling interferes with cell growth in vivoThe IGF receptorIGF-1R is highly overexpressed both on the cell surface andin the nucleus of RMS cells furthermore RMS cell lines aresensitive to IGF-1R inhibitionThe IGF-2 locus shows a loss ofimprinting in both ERMS andARMS tumors and the expres-sion of PAX3-FKHR has been demonstrated to upregulateIGF-2 and activate IGF pathway in ARMS [69] Mariannaet al found that IGF-2 and tumor suppressors p19Arf andp21Cip1 are overexpressed and prominently upregulated inRMS mouse models [71]

43 TGF-120573 Signaling Pathway The regulatory role of Trans-forming Growth Factor-120573 (TGF-120573) in RMS has been recentlystudied [72] Wang et al demonstrated that the expression ofTGF-1205731 is significantly higher in RMS than in normal skeletalmuscle The inhibition of TGF-1205731 expression by shRNA-expressing vectors reverses themalignant behavior of RMSbyinhibiting cell growth and inducingmyogenic differentiationTGF-1205731 shRNA induces myogenin expression a regulator ofmyogenic differentiation genes Myogenin acts as a target fornegative regulation of myogenesis by TGF-1205731 signals with

Sarcoma 7

a differentiation regulatory cascade These results suggestthat the TGF-1205731 signaling pathway disrupts the differentia-tion of myogenic progenitors leading to the development ofRMS

44 FGF Signaling Pathway Fibroblast Growth Factor (FGF)is crucial in embryonic development and functions to driveproliferation FGFs and their receptors (FGFRs) are essentialregulators in the processes of proliferation antiapoptosisdrug resistance and angiogenesis in RMS [69] RMS over-expresses the receptor tyrosine kinase FGFR4 which causesautophosphorylation and constitutive signaling in correlationwith poor differentiation and decreased survival [73] Recentwork has shown that FGF signaling can prevent ARMS cellsfrom apoptosis induced by targeting the IGF1-R-PI3K-mTORpathway [74]

45 ERK Signaling Pathway Previous studies have shownthat elevated myostatin expression deregulates ExtracellularRegulated Kinase (ERK) signaling and deficient activationof the p38 pathway contributes to the differentiation blockin RMS cells [62] The ERK pathway is frequently highlyactivated in RMS cells from embryonic derivation due to thepresence of activating RASmutations leading to reduced dif-ferentiation by blocking the p38 pathway [75]These findingsindicate that interventions that target themyostatinERKp38network could be an effective way to promote differentiationof RMS

46 Hippo Signaling Pathway Many upstream signal trans-duction proteins in response to cues from the cellular micro-environment regulate the core Hippo pathway [76] TheHippo pathway may limit tumorigenesis by inducing thecytosolic localization of the transcriptional cofactor Yes-Associated Protein 1 (YAP1) High YAP levels and activitiesincrease activated satellite cells and prevent differentiationby activating genes that inhibit differentiation [77] YAP1-TEAD1 was found to upregulate proproliferative and onco-genic genes and maintain the ERMS differentiation blockby interfering with MYOD1 and MEF2 prodifferentiationactivities [76] Inhibition of theHippo pathway is exhibited inRMS through downregulation of Hippo pathway tumor sup-pressors or upregulation of YAP [78] These data suggest thatHippo pathway dysfunction promotes RMS development

47 Notch Signaling Pathway and Wnt Signaling PathwayNotch pathway as an embryonic signaling pathway promotesmuscle stem cell maintenance by inhibiting myogenic earlydifferentiation and expanding pools of progenitor cells dur-ing both embryogenesis and postnatal muscle regenerationSimilar to Notch Wnt has various roles in embryonic fetaland neonatalmyogenesis during skeletalmyogenesis As bothpathways are regulators of myogenic lineage determinationand maturation RMS can arise from disordered regulationof these normal developmental pathways [79]

48 Caveolins in RMS Recent findings have shown that cav-eolins are expressed in a cell stage-dependent manner inRMS [80 81] Caveolins are scaffolding proteins that regulate

several targets and pathways (such as p53 IGF RASERK andTGF-120573myostatin signaling) associated with RMS develop-ment and therefore loss or gain of caveolin function is ableto generate multiple effects on the tumor behaviorThereforerecognizing their precise roles in RMS progression will becrucial to elaborate targeted therapies particularly as oneof the three different isoforms CAV1 could act as a potenttumor suppressor in ARMS Inhibition of CAV1 function cancontribute to aberrant cell proliferation leading to ARMSdevelopment [80]

49 DNA Repairing System Some studies indicate that RMSis characterized by germline mutations of DNA repair genes[82 83] DNA repair gene alterations in RMS occur sec-ondarily to malignant transformation The modificationsin DNA repair enzyme activity can result in resistance toadjuvant treatment in RMS tumor cells which limits theprognosis of RMS There are two pathways for repairingDNA breaks directly without affecting DNA structure andindirectly by DNA phosphodiester backbone cleavage [82](Figure 4) Direct repair includes repair during replicationand enzymatic repair Indirect repair is comprised of exci-sion repair systems including base excision repair (BER)nucleotide excision repair (NER) mismatch repair (MMR)and recombination repair (RR) [83] Defective DNA repairmechanisms result from point mutations or LOH in RMSand contribute to tumorigenesis MGMT and MMR proteinactivity and expression levels are used as predictor indices fortherapy outcome in RMS BER as the major DNA repair sys-tem against damage resulting from cellular metabolism canreverse the cytotoxic effects of alkylation agents used suchas antineoplastics subsequent tumor progression and deathimplicates an RMS mechanism of resistance to chemother-apeutic agents This may provide novel individualized ther-apeutics targeting downregulation of activated DNA repairenzymes or upregulation of DNA repair deficiencies [82]

5 MicroRNA (miRNA miR)Expression in RMS

Muscle-specific and ubiquitously expressed miRs appeardownregulated in RMS tumors and cell lines compared withthe normal counterparts These miRs often play crucial rolesas antioncogenes Inhibition of these miRs contributes toenhanced tumorigenesis through the modulation of diversemolecular pathways Upregulation of prooncogenic miRs hasbeen detected in RMS recently as well [84] (Table 4)

51 Antioncogenic miR in RMS Gain-of-function experimentshave demonstrated that reexpression of selected ldquotumor sup-pressorrdquo miRs impairs the tumorigenic behavior of RMS cells[84 85] As myo-miR family members miR-1 miR-133a andmiR-206 have been demonstrated to be downregulated inRMS and shown to have activity on mRNA expression bytargeting c-Met [42 43 86] Inhibition of thesemyo-miRNAsmay cause aberrant cell proliferation and migration in myo-genesis leading to RMS development especially for ERMSThere have been more miRs found as tumor suppressors inthis malignancy including miR-29 miR-450b-5p miR-203

8 Sarcoma

Table 4 MicroRNAs involved in myogenesis and RMS development

miRNA Expression Target Function Reference

miR-1 and miR-133a Downregulation MYH9Myogenic miRNA inhibit differentiationand promote proliferation in myogenesiscytostatic

Rao et al 2010 [42]

miR-206 Downregulation cMET Promote differentiation and proliferationin myogenesis

Yan et al 2009 [43]

miR-29 Downregulation HADC4 YY1 EZH2 Promote stabilization of RMS phenotype Marchesi et al 2014 [44]

miR-450-5p Downregulation ENOX PAX9 Promote differentiation and progression Sun et al 2014 [45]

miR-203 Downregulation JAK STAT Notch Inhibit differentiation and proliferation inmyogenesis tumor suppressor

Diao et al 2014 [46]

miR-214 Downregulation N-Ras Inhibit tumor cell growth and inducemyogenic differentiation and apoptosis

Huang et al 2014 [47]

miR-183 Upregulation EGR1 Promote migration and metastasis Sarver et al 2010 [48]

PMS2

MGMT

HR

Mismatch Recombination

NHEJ MMEJ

p53

Excision

BER NER

RMS

DNA repair

Direct repair Indirect repair

repair repairrepair

Figure 4The DNA repair systems in RMSThere are two pathways repairing the DNA lesions directly without affecting DNA structure andindirectly by DNA phosphodiester backbone cleavage The modifications in DNA repair enzymes expression or activity lead to resistanceto chemotherapy and radiation in RMS tumor cells MGMT O6-methylguanine-DNA methyltransferase BER base excision repair NERnucleotide excision repair HR homologous recombination NHEJ nonhomologous end-joining MMEJ microhomology-mediated end-joining

and miR-214 [44 46 47 84 87] They are all significantlydownregulated in RMS compared with normal skeletal mus-cles and function to inhibit tumor cell growth and inducemyogenic differentiation and apoptosis in RMS

52 Oncogenic miRNA in RMS The transcription factor EGR1is a tumor suppressor gene that is downregulated in RMS [48]miR-183 functions as an oncogene by targeting EGR1 and pro-moting tumor cell migration Either by direct anti-miR treat-ment or by indirect mechanisms that decrease transcriptmiR-183 targeted treatmentsmight provide a potential optionto RMS patients

The basic strategy of current effective miR-based treat-ment studies is either efficient reexpression of miRs or restor-ing the function of miRs to inhibit the expression of certain

protein-coding genes and promote muscle differentiationMoreover miR expression profiling in tumors and possiblytheir detection in peripheral blood during treatment may beable to predict the response to chemotherapy or radiotherapyand be useful as a prognostic signature for the development oftreatment resistanceThe prognostic value of miR expressionlevels in RMS is a powerful tool in creating a better-tailoredstrategy for particular subsets of patients

53 Long Noncoding RNAs in RMS Long noncoding RNAs(LNCRNAs) are abundant in the mammalian transcriptomeand have been shown to play a role in RMS development [8889]TheH19 gene localized within a chromosomal region onhuman chromosome 11p15 encodes an imprinted untrans-lated RNA [90] H19 opposite tumor suppressor (HOTS)

Sarcoma 9

Table 5 In vivo animal models of RMS

Model TargetModel origin Reference

Inactivation Expression

DrosophilaPAX-FKHR Bipartite Gal4-UAS

expression systemGalindo et al2006 [49]

PAX7-FOXO1 rols Chromosomal deletionDf(3L)vin5

Avirneni-Vadlamudi et al2012 [50]

Zebrafishrag2 promoter c-Myc kRASG12D

Chen andLangenau 2011[51]

MAPKERK andAKTS6K1 PD98059 TPCK Tg(hsp70-HRASG12V) Le et al 2012

[52]

Mouse

Sufu N-myc Sfrp1 Ptch2and cyclin D1 Sufu+minus Lee et al 2007

[53]

Ink4aArf and Trp53 Pax3FKHR Pax3P3Fpwt Keller et al2004 [54]

Wnt120573-cateninsignaling pathway Wnt2 p53minusminusc-fosminusminus Singh et al 2010

[55]

a tumor growth inhibitor is encoded by an imprinted H19antisense transcript Overexpression of HOTS inhibits RMStumor cell growth Silencing HOTS by RNAi was shown toincrease in vitro colony formation as well as in vivo tumorgrowth of RMS [90]

6 In Vivo Models of RMS

Several in vivo models have recently been developed inDrosophila zebrafish and mice to further understand themolecular and cellular biology of RMS tumorigenesis [49ndash55 91ndash94] (Table 5)

61 Drosophila Models in RMS Despite the evolutionarydistance a strong conservation of genes pathways andregulatory molecular networks have been demonstratedbetween flies and humans Many human disease genes haverelated sequences in Drosophila In contrast to mammalianmodels the generation of Drosophila mutants is easy cheapand fast

For a better understanding of the pathogenic conse-quences of PAX-FKHR expression Drosophila was chosenbecause the animalrsquos transparent outer cuticle allows forsimple real-time monitoring of muscle abnormalities elicitedby PAX-FKHR including subtle or focal changes [49] Themutation in the myoblast fusion gene rolling pebbles (rols)was reported to dominantly suppress PAX-FKHR1-inducedlethality by using a Drosophila model of PAX-FKHR1-mediated transformation [50] PAX-FKHR1 signaling upreg-ulates the TANC1 gene and blocks myoblasts from terminaldifferentiation However downregulating the TANC1 genecould cause RMS cells to lose their neoplastic state undergofusion and form differentiation This novel finding uncov-ered a PAX-FKHR1-TANC1 neoplasia axis collaboratively ina Drosophila model and loss-gain-of-function studies inmammalian platforms [50]

62 Zebrafish Models in RMS In contrast to other mod-els the short tumor onset in zebrafish allows for rapididentification of essential genes in various processes suchas tumor growth self-renewal and maintenance There areother advantages for using zebrafish in the study of humandisease and development including the ease and low cost ofraising large numbers of fish and the highly conserved geneticand biochemical pathways between zebrafish and mammals[95]

As zebrafish RMS is highly similar to human ERMSby using fluorescent transgenic approaches it is more con-venient to understand histogenesis and different aspects oftumorigenesis in RMS A robust zebrafish model of RAS-induced RMS has been established sharing twomorphologicand immunophenotypic features resembling ERMS [51] Inone case both are associated with tissue-restricted geneexpression in RMS while in another both comprise a RAS-induced gene signature Cross-species microarray compar-isons confirm that conserved genetic tumor-specific andtissue-restricted pathways such as RAS and p53 pathwaysdrive RMS growth Tg(hsp70-HRASG12V) zebrafish embryoswere generated to evaluate gene expression that mimics RASpathway activation during tumorigenesis to defineRAS targetgenes [52] Another KRASG12D-induced zebrafish embryonalRMS was utilized to assess the therapeutic effects By theblockage of two major downstream signaling pathwaysMAPKERK and AKTS6K1 the model showed that inhibi-tion of translation initiation suppresses tumor cell prolifera-tion [52]

All the above zebrafish cancer models share similar cellu-lar features and molecular pathways with human RMS espe-cially ERMS and can demonstrate a response to therapiesin a similar manner with human RMS Thus a tremendousopportunity is available to implement these animal modelsinto different steps for novel targeted therapeutic develop-ment

10 Sarcoma

63 Mouse Models in RMS Mouse models established fromin vivo studies may contribute to the understanding of thegenetic basis in tumor development and progression andmayhelp to test the efficacies of novel antineoplastic agents

The importance of PAX3PAX7-FKHR fusion proteins inthe progression of ARMS tumorigenesis is evident from bothin vitro and in vivo studies Transgenic mouse models gener-ated with the PAX-FKHR fusion genemimic the formation ofhuman ARMS [67] A mouse model was developed with het-erozygous loss of Sufu a tumor suppressor inHedgehog (Hh)signaling combined with p53 loss driven ERMS with a lowpenetrance (9) [53] These studies indicate that the modelsconsistently formingARMSnot only need the introduction ofthe PAX-FKHR fusion gene but also need to be accompaniedby other genetic events such as p53 pathway disruption [54]

Compared with ARMS ERMS models are more complexto generate because of their lower tumor penetrance andlonger latency JW41 cells from the p53c-fos double mutantmice models resemble human ERMS cells morphologicallyand express similar characteristic markers [55] The overex-pression of the Wnt2 gene identified in JW41 cells confirmedin human RMS cell lines can allow for further analysis of theWnt signaling pathway Both of these results indicate that thedownregulation of theWnt pathway contributes to resistanceto apoptosis and the inhibition of myogenic differentiation inERMS

Using Ptch1 p53 andor Rb1 conditional mouse modelsand controlling prenatal or postnatal myogenic cell of originRubin et al demonstrated that the loss of p53 in maturingmyoblasts related to tumorigenesis of ERMS [96] They alsoindicated that Rb1 alteration with other oncogenic factorswas strongly associated with an undifferentiated phenotypeacting as a modifier In addition they highlighted a sub-set of p53-deficient ERMSs arising from Myf6-expressingmyoblasts which had the same latency with Pax7-expressingmurine satellite cells in the study

7 Potential Therapeutic Targets in RMS

Detection of genomic imbalances and identification of thecrucial effective genes can contribute to identifying novelpotential biomarkers and relevant targets for clinical therapyin RMS

Survivin-responsive conditionally replicating adenovi-ruses can regulate multiple factors (Survm-CRAs) [41]Although Survm-CRA can efficiently replicate and potentlyinduce cell death in RMS cells the cytotoxic effects are morepronounced in RSC-enriched or RSC-purified cells thanin RSC-exiguous or progeny-purified cells Injections ofSurvm-CRAs into tumor nodules generated by transplantingRSC-enriched cells induce significant death in RMS cells andregression of tumor nodulesThe unique therapeutic featuresof Survm-CRA include not only its therapeutic effectivenessagainst RMS but also its increasing effectiveness against CSCsuggesting that Survm-CRA may be a promising anticanceragent [41]

Rapamycin an inhibitor of mTOR can abrogate RMStumor growth in a xenograft mouse model [97] Thetumors in the rapamycin-treatedmice group show significant

reduction of proliferation and invasiveness and inductionof apoptosis The tumor growth inhibition is simultaneousassociated with the diminution of mTOR and Hh pathwayswhich are implicated in the pathogenesis of RMSThe resultsindicate that using rapamycin either alone or in combinationwith traditional chemotherapeutic drugs may represent apotential targeted agent for therapeutic intervention Ampli-fication and mutational activation of Fibroblast Growth Fac-tor Receptor 4 (FGFR4) in RMS cells promote tumor pro-gression [98] Ponatinib is the most potent FGFR4 inhibitorto block wild-type RMS cell growth mutate FGFR4 throughincreasing apoptosis and suppress the FGFR4 downstreamtarget STAT3 Ponatinib treatment slows tumor growth inRMS mouse models expressing mutated FGFR4 [98] Pona-tinib as an FGFR4 inhibitor shows potential to act as aneffective therapeutic agent for RMS tumorsThe expression ofCXC chemokine receptor 4 (CXCR4) CXCR7 and VascularEndothelial Growth Factor (VEGF) indicates poor prognosisin a variety of malignancies including RMS cell lines Mostsamples in a large series of clinical RMS cases show asso-ciation with high expression of these antagonists regardlessof their subtypes [99] However there are significant corre-lations with high expression of CXCR4 and VEGF in bothERMS and ARMS High VEGF expression is predictive ofadverse prognostic factors in RMS as the expression of theseangiogenesis factors was compared with clinicopathologicalparameters and prognosis Considering their overexpressionin RMS these chemokine receptors and VEGF could providepotential molecular therapeutic targets in RMS Some ARMScell lines have undergone apoptosis in response to antineo-plastic drugs such as bortezomib The proapoptotic BH3only family member NOXA acts as an upregulated proteindownstream PAX3-FKHR resulting in increased cell sensi-tivity to bortezomib Apoptosis in response to bortezomibcan be reversed by shRNA knockdown of Noxa PAX3-FKHR upregulation of Noxa creates a potential therapeuticinsight into inducing apoptosis in ARMS cellsThis apoptosispathway could represent a specific targeted therapy againstPAX3-FKHR-expressing ARMS cells [100]

Proton pump activation resulting from intracellular acid-ificationmay be an energetic mechanism to escape apoptosisV-ATPase is an ATP-driven proton pump acidifying theintracellular compartment and transports protons across theplasma membrane Esomeprazole as a V-ATPase inhibitor inthe PPI administration interferes with intensive ion trans-porter activity of RMS cells and induces remarkable cytotoxi-city RMS expresses a higher level ofV-ATPase comparedwithother sarcomasTherefore RMS should be very susceptible toesomeprazole treatment and V-ATPase could be consideredas a promising selective target for treatment [101]

8 Conclusion and Future Prospects

For RMS the current challenges include how to iden-tify promising novel therapeutics and integrate them intoexisting therapy For better understanding of congenitaland epigenetic modifications in the development of RMSthis review summarizes the recent genome-wide studies onmolecular and genetic alterations to decipher the underlying

Sarcoma 11

tumorigenesis mechanisms There is a link between geneticand epigenetic alterations responsible for a variety of tumorcell growth proliferation differentiation apoptosis andtherapy-resistance mechanisms The identification of theprognostic value of the PAX-FKHR fusion status in RMSis one of the most important shifts in the lineage and therisk assessment of RMS which exerts an oncogenic effectthrough multiple pathways and is incorporated into mostrelevant studies The misbalanced expression of a number ofmiRs involved in the regulation of myogenic differentiationalso implicates the tumorsrsquo escape suppressive mechanismsMoreover we described several in vivo models of RMSto explore underlying histogenesis and different aspects oftumorigenesis Recent work on CSCs has also contributedto understanding the cell origins refining molecular char-acterization and elucidating the underlying basis of refrac-tory responses to current adjuvant treatments of subsets oftumorsThese integrative approaches can provide opportuni-ties to identify diagnostic and prognostic biomarkers appliedto the individual targeted therapy and significantly improveRMS prognosis

Abbreviations

RMS RhabdomyosarcomaARMS Alveolar rhabdomyosarcomaERMS Embryonal rhabdomyosarcomaPRMS Pleomorphic rhabdomyosarcomaSRMS Sclerosingspindle cell

rhabdomyosarcomamiRNA miR MicroRNACSC Cancer stem cellMSC Mesenchymal stem cell

Conflict of Interests

The authors declare that there is no conflict of interestsregarding the publication of this paper

Acknowledgments

This work was supported in part by grants from the Gat-tegno and Wechsler funds Dr Duan was supported in partthrough a grant from Sarcoma Foundation of America (SFA)a grant from National Cancer Institute (NCI)National Insti-tutes of Health (NIH) UO1 CA 151452 and a grant from anAcademic Enrichment Fund of MGH Orthopedic Surgery

References

[1] L Yang T Takimoto and J Fujimoto ldquoPrognostic model forpredicting overall survival in children and adolescents withrhabdomyosarcomardquo BMC Cancer vol 14 no 1 article 6542014

[2] D Egas-Bejar and W W Huh ldquoRhabdomyosarcoma in adoles-cent and young adult patients current perspectivesrdquo AdolescentHealth Medicine andTherapeutics vol 5 pp 115ndash125 2014

[3] J C Breneman E Lyden A S Pappo et al ldquoPrognosticfactors and clinical outcomes in children and adolescents withmetastatic rhabdomyosarcomamdasha report from the intergrouprhabdomyosarcoma study IVrdquo Journal of Clinical Oncology vol21 no 1 pp 78ndash84 2003

[4] V D Soleimani and M A Rudnicki ldquoNew insights into theorigin and the genetic basis of rhabdomyosarcomasrdquo CancerCell vol 19 no 2 pp 157ndash159 2011

[5] T Glass D D Cochrane S R Rassekh K Goddard and JHukin ldquoGrowing teratoma syndrome in intracranial non-ger-minomatous germ cell tumors (iNGGCTs) a risk for secondarymalignant transformationmdasha report of two casesrdquo Childrsquos Ner-vous System vol 30 no 5 pp 953ndash957 2014

[6] J Ji C Eng and K Hemminki ldquoFamilial risk for soft tissuetumors a nation-wide epidemiological study from SwedenrdquoJournal of Cancer Research and Clinical Oncology vol 134 no5 pp 617ndash624 2008

[7] C D M Fletcher J A Bridge P Hogendoorn and F MertensWHO Classification of Tumours of Soft Tissue and Bone vol 5IARC Press Paris France 4th edition 2013

[8] I Sultan I Qaddoumi S Yaser C Rodriguez-Galindo and AFerrari ldquoComparing adult and pediatric rhabdomyosarcoma inthe surveillance epidemiology and end results program 1973 to2005 an analysis of 2600 patientsrdquo Journal of Clinical Oncologyvol 27 no 20 pp 3391ndash3397 2009

[9] B Raney W Huh D Hawkins et al ldquoOutcome of patientswith localized orbital sarcoma who relapsed following treat-ment on Intergroup Rhabdomyosarcoma Study Group (IRSG)Protocols-III and -IV 1984ndash1997 a report from the ChildrenrsquosOncology Grouprdquo Pediatric Blood and Cancer vol 60 no 3 pp371ndash376 2013

[10] W Crist E A Gehan A H Ragab et al ldquoThe third intergrouprhabdomyosarcoma studyrdquo Journal of Clinical Oncology vol 13no 3 pp 610ndash630 1995

[11] C Liu D Li J Jiang et al ldquoAnalysis of molecular cytogeneticalteration in rhabdomyosarcoma by array comparative genomichybridizationrdquo PLoS ONE vol 9 no 4 Article ID e94924 2014

[12] DWilliamson E Missiaglia A de Reynies et al ldquoFusion gene-negative alveolar rhabdomyosarcoma is clinically and molec-ularly indistinguishable from embryonal rhabdomyosarcomardquoJournal of Clinical Oncology vol 28 no 13 pp 2151ndash2158 2010

[13] V Paulson G Chandler D Rakheja et al ldquoHigh-resolutionarray CGH identifies common mechanisms that drive embry-onal rhabdomyosarcoma pathogenesisrdquo Genes Chromosomesand Cancer vol 50 no 6 pp 397ndash408 2011

[14] S Park J Lee I-G Do et al ldquoAberrant CDK4 amplificationin refractory rhabdomyosarcoma as identified by genomicprofilingrdquo Scientific Reports vol 4 article 3623 2014

[15] C Liu D Li J Hu et al ldquoChromosomal and genetic imbalancesin Chinese patients with rhabdomyosarcoma detected by high-resolution array comparative genomic hybridizationrdquo Interna-tional Journal of Clinical and Experimental Pathology vol 7 no2 pp 690ndash698 2014

[16] Q-X Li C-X Liu C-P Chun et al ldquoChromosomal imbalancesrevealed in primary rhabdomyo-sarcomas by comparativegenomic hybridizationrdquo Chinese Medical Journal vol 122 no11 pp 1277ndash1282 2009

[17] E Missiaglia J Selfe M Hamdi et al ldquoGenomic imbalancesin rhabdomyosarcoma cell lines affect expression of genesfrequently altered in primary tumors an approach to identifycandidate genes involved in tumor developmentrdquo Genes Chro-mosomes and Cancer vol 48 no 6 pp 455ndash467 2009

12 Sarcoma

[18] F G Barr F Duan L M Smith et al ldquoGenomic and clinicalanalyses of 2p24 and 12q13-q14 amplification in alveolar rhab-domyosarcoma a report from the childrenrsquos oncology grouprdquoGenes Chromosomes and Cancer vol 48 no 8 pp 661ndash6722009

[19] T Gordon AMcManus J Anderson et al ldquoCytogenetic abnor-malities in 42 rhabdomyosarcoma a United Kingdom CancerCytogenetics Group StudyrdquoMedical and Pediatric Oncology vol36 no 2 pp 259ndash267 2001

[20] X Chen E Stewart A A Shelat et al ldquoTargeting oxidativestress in embryonal rhabdomyosarcomardquo Cancer Cell vol 24no 6 pp 710ndash724 2013

[21] S Lizard-Nacol F Mugneret C Volk C Turc-Carel M Favrotand T Philip ldquoTranslocation (213)(q37q14) in alveolar rhab-domyosarcoma a new caserdquo Cancer Genetics and Cytogeneticsvol 25 no 2 pp 373ndash374 1987

[22] P H B Sorensen J C Lynch S J Qualman et al ldquoPAX3-FKHR and PAX7-FKHR gene fusions are prognostic indicatorsin alveolar rhabdomyosarcoma a report from the childrenrsquosoncology grouprdquo Journal of Clinical Oncology vol 20 no 11 pp2672ndash2679 2002

[23] A D Marshall and G C Grosveld ldquoAlveolar rhabdomyo-sarcomamdashthe molecular drivers of PAX37-FOXO1-inducedtumorigenesisrdquo Skeletal Muscle vol 2 no 1 article 25 2012

[24] E Missiaglia D Williamson J Chisholm et al ldquoPAX3FOXO1fusion gene status is the key prognostic molecular markerin rhabdomyosarcoma and significantly improves current riskstratificationrdquo Journal of Clinical Oncology vol 30 no 14 pp1670ndash1677 2012

[25] J F Shern L Chen J Chmielecki et al ldquoComprehensivegenomic analysis of rhabdomyosarcoma reveals a landscape ofalterations affecting a common genetic axis in fusion-positiveand fusion-negative tumorsrdquo Cancer Discovery vol 4 no 2 pp216ndash231 2014

[26] MWachtel M Dettling E Koscielniak et al ldquoGene expressionsignatures identify rhabdomyosarcoma subtypes and detect anovel t(22)(q35p23) translocation fusing PAX3 to NCOA1rdquoCancer Research vol 64 no 16 pp 5539ndash5545 2004

[27] E Davicioni M J Anderson F G Finckenstein et al ldquoMolec-ular classification of rhabdomyosarcomamdashgenotypic and phe-notypic determinants of diagnosis a report from the ChildrenrsquosOncology Grouprdquo The American Journal of Pathology vol 174no 2 pp 550ndash564 2009

[28] M Visser C Sijmons J Bras et al ldquoAllelotype of pediatric rhab-domyosarcomardquo Oncogene vol 15 no 11 pp 1309ndash1314 1997

[29] R Gil-Benso T San-Miguel R C Callaghan et al ldquoChromo-somal and genetic changes produced in tumoral progression ofembryonal rhabdomyosarcomardquo Histopathology vol 62 no 5pp 816ndash819 2013

[30] S X Skapek J Anderson F G Barr et al ldquoPAX-FOXO1 fusionstatus drives unfavorable outcome for children with rhabdom-yosarcoma a childrenrsquos oncology group reportrdquo Pediatric Bloodand Cancer vol 60 no 9 pp 1411ndash1417 2013

[31] HHosoi N Kakazu E Konishi et al ldquoA novel PAX3 rearrange-ment in embryonal rhabdomyosarcomardquo Cancer Genetics andCytogenetics vol 189 no 2 pp 98ndash104 2009

[32] E Takaoka H Sonobe K Akimaru et al ldquoMultiple sites ofhighly amplified DNA sequences detected bymolecular cytoge-netic analysis inHS-RMS-2 a newpleomorphic rhabdomyosar-coma cell linerdquo American Journal of Cancer Research vol 2 no2 pp 141ndash152 2012

[33] K Szuhai D de Jong W Y Leung C D M Fletcher and P CWHogendoorn ldquoTransactivatingmutation of theMYOD1 geneis a frequent event in adult spindle cell rhabdomyosarcomardquoJournal of Pathology vol 232 no 3 pp 300ndash307 2014

[34] N P Agaram C-L Chen L Zhang M P Laquaglia L Wexlerand C R Antonescu ldquoRecurrent MYOD1 mutations in pedi-atric and adult sclerosing and spindle cell rhabdomyosarcomasEvidence for a common pathogenesisrdquoGenes Chromosomes andCancer vol 53 no 9 pp 779ndash787 2014

[35] H Yoshida M Miyachi K Sakamoto et al ldquoPAX3-NCOA2fusion gene has a dual role in promoting the proliferation andinhibiting the myogenic differentiation of rhabdomyosarcomacellsrdquo Oncogene vol 33 pp 5601ndash5608 2013

[36] J M Mosquera A Sboner L Zhang et al ldquoRecurrent NCOA2gene rearrangements in congenitalinfantile spindle cell rhab-domyosarcomardquo Genes Chromosomes and Cancer vol 52 no6 pp 538ndash550 2013

[37] V Thavaraj A Sethi and L S Arya ldquoIncomplete Beckwith-Wiedemann syndrome in a child with orbital rhabdomyosar-comardquo Indian Pediatrics vol 39 no 3 pp 299ndash304 2002

[38] D Walter S Satheesha P Albrecht et al ldquoCD133 positiveembryonal rhabdomyosarcoma stem-like cell population isenriched in rhabdospheresrdquo PLoS ONE vol 6 no 5 Article IDe19506 2011

[39] Y-X Ren F G Finckenstein D A Abdueva et al ldquoMousemesenchymal stem cells expressing PAX-FKHR form alveolarrhabdomyosarcomas by cooperating with secondary muta-tionsrdquo Cancer Research vol 68 no 16 pp 6587ndash6597 2008

[40] M Salerno S Avnet G Bonuccelli S Hosogi D Granchi andN Baldini ldquoImpairment of lysosomal activity as a therapeuticmodality targeting cancer stem cells of embryonal rhabdom-yosarcoma cell line RDrdquo PLoS ONE vol 9 no 10 Article IDe110340 2014

[41] K Tanoue Y Wang M Ikeda et al ldquoSurvivin-responsive con-ditionally replicating adenovirus kills rhabdomyosarcoma stemcells more efficiently than their progenyrdquo Journal of Transla-tional Medicine vol 12 no 1 article 27 2014

[42] P K Rao EMissiaglia L Shields et al ldquoDistinct roles formiR-1and miR-133a in the proliferation and differentiation of rhab-domyosarcoma cellsrdquo The FASEB Journal vol 24 no 9 pp3427ndash3437 2010

[43] D Yan X D Dong X Chen et al ldquoMicroRNA-1206 targets c-met and inhibits rhabdomyosarcomadevelopmentrdquoThe Journalof Biological Chemistry vol 284 no 43 pp 29596ndash29604 2009

[44] I Marchesi A Giordano and L Bagella ldquoRoles of enhancerof zeste homolog 2 from skeletal muscle differentiation torhabdomyosarcoma carcinogenesisrdquoCell Cycle vol 13 no 4 pp516ndash527 2014

[45] M M Sun J F Li L L Guo et al ldquoTGF-1205731 suppression ofmicroRNA-450b-5p expression a novel mechanism for block-ingmyogenic differentiation of rhabdomyosarcomardquoOncogenevol 33 no 16 pp 2075ndash2086 2014

[46] Y Diao X Guo L Jiang et al ldquoMiR-203 a tumor suppressorfrequently down-regulated by promoter hypermethylation inrhabdomyosarcomardquo The Journal of Biological Chemistry vol289 no 1 pp 529ndash539 2014

[47] H-J Huang J Liu HHua et al ldquoMiR-214 andN-ras regulatoryloop suppresses rhabdomyosarcoma cell growth and xenografttumorigenesisrdquo Oncotarget vol 5 no 8 pp 2161ndash2175 2014

Sarcoma 13

[48] A L Sarver L Li and S Subramanian ldquomicroRNA miR-183functions as an oncogene by targeting the transcription factorEGR1 and promoting tumor cell migrationrdquo Cancer Researchvol 70 no 23 pp 9570ndash9580 2010

[49] R L Galindo J A Allport and E N Olson ldquoA Drosophilamodel of the rhabdomyosarcoma initiator PAX7-FKHRrdquo Pro-ceedings of the National Academy of Sciences of the United Statesof America vol 103 no 36 pp 13439ndash13444 2006

[50] U Avirneni-Vadlamudi K A Galindo T R Endicott V Paul-son S Cameron and R L Galindo ldquoDrosophila and mammal-ian models uncover a role for the myoblast fusion gene TANC1in rhabdomyosarcomardquo The Journal of Clinical Investigationvol 122 no 1 pp 403ndash407 2012

[51] E Y Chen andDM Langenau ldquoZebrafishmodels of rhabdom-yosarcomardquoMethods in Cell Biology vol 105 pp 383ndash402 2011

[52] X Le E K Pugach S Hettmer et al ldquoA novel chemicalscreening strategy in zebrafish identifies common pathways inembryogenesis and rhabdomyosarcoma developmentrdquo Devel-opment vol 140 no 11 pp 2354ndash2364 2012

[53] Y Lee R Kawagoe K Sasai et al ldquoLoss of suppressor-of-fusedfunction promotes tumorigenesisrdquoOncogene vol 26 no 44 pp6442ndash6447 2007

[54] C Keller B R Arenkiel C M Coffin N El-Bardeesy R ADePinho and M R Capecchi ldquoAlveolar rhabdomyosarcomasin conditional Pax3Fkhr mice cooperativity of Ink4aARF andTrp53 loss of functionrdquo Genes and Development vol 18 no 21pp 2614ndash2626 2004

[55] S Singh CVinson CMGurley et al ldquoImpairedWnt signalingin embryonal rhabdomyosarcoma cells from p53c-fos doublemutant micerdquo American Journal of Pathology vol 177 no 4 pp2055ndash2066 2010

[56] G J Schaaf J M Ruijter F Van Ruissen et al ldquoFull transcrip-tome analysis of rhabdomyosarcoma normal and fetal skeletalmuscle statistical comparison of multiple SAGE librariesrdquo TheFASEB Journal vol 19 no 3 pp 404ndash406 2005

[57] D S Hawkins A A Gupta and E R Rudzinski ldquoWhat is newin the biology and treatment of pediatric rhabdomyosarcomardquoCurrent Opinion in Pediatrics vol 26 no 1 pp 50ndash56 2014

[58] M Zhang J Truscott and J Davie ldquoLoss of MEF2D expressioninhibits differentiation and contributes to oncogenesis in rhab-domyosarcoma cellsrdquoMolecular Cancer vol 12 no 1 article 1502013

[59] M Ciesla J Dulak and A Jozkowicz ldquomicroRNAs and epi-genetic mechanisms of rhabdomyosarcoma developmentrdquo TheInternational Journal of Biochemistry amp Cell Biology vol 53 pp482ndash492 2014

[60] S Ricaud B Vernus M Duclos et al ldquoInhibition of autocrinesecretion of myostatin enhances terminal differentiation inhuman rhabdomyosarcoma cellsrdquo Oncogene vol 22 no 51 pp8221ndash8232 2003

[61] C S Guo C Degnin T A Fiddler D Stauffer andM JThayerldquoRegulation of MyoD activity and muscle cell differentiation byMDM2 pRb and Sp1rdquoThe Journal of Biological Chemistry vol278 no 25 pp 22615ndash22622 2003

[62] S Rossi E Stoppani P L Puri and A Fanzani ldquoDifferenti-ation of human rhabdomyosarcoma RD cells is regulated byreciprocal functional interactions between myostatin p38 andextracellular regulated kinase signalling pathwaysrdquo EuropeanJournal of Cancer vol 47 no 7 pp 1095ndash1105 2011

[63] Z S Walters B Villarejo-Balcells D Olmos et al ldquoJARID2is a direct target of the PAX3-FOXO1 fusion protein andinhibits myogenic differentiation of rhabdomyosarcoma cellsrdquoOncogene vol 33 no 9 pp 1148ndash1157 2014

[64] T Hosoyama M I Aslam J Abraham et al ldquoIL-4R drives ded-ifferentiation mitogenesis and metastasis in rhabdomyosar-comardquo Clinical Cancer Research vol 17 no 9 pp 2757ndash27662011

[65] F S Dela Cruz ldquoCancer stem cells in pediatric sarcomasrdquoFrontiers in Oncology vol 3 article 168 2013

[66] M J Bissell and W C Hines ldquoWhy donrsquot we get more cancerA proposed role of the microenvironment in restraining cancerprogressionrdquo Nature Medicine vol 17 no 3 pp 320ndash329 2011

[67] E Charytonowicz C Cordon-Cardo I Matushansky and MZiman ldquoAlveolar rhabdomyosarcoma is the cell of origin amesenchymal stem cellrdquoCancer Letters vol 279 no 2 pp 126ndash136 2009

[68] W K Cavenee ldquoMuscling in on rhabdomyosarcomardquo NatureMedicine vol 8 no 11 pp 1200ndash1201 2002

[69] B Zhu and J K Davie ldquoNew insights into signalling-pathwayalterations in rhabdomyosarcomardquo British Journal of Cancervol 112 no 2 pp 227ndash231 2015

[70] M Zhang C M Linardic and D G Kirsch ldquoRAS and ROS inrhabdomyosarcomardquo Cancer Cell vol 24 no 6 pp 689ndash6912013

[71] S C Marianna L Ianzano and G Nicoletti ldquoTumor sup-pressor genes promote rhabdomyosarcoma progression in p53heterozygousrdquo Oncotarget vol 5 no 1 pp 108ndash119 2013

[72] SWang L Guo L Dong et al ldquoTGF-beta1 signal pathwaymaycontribute to rhabdomyosarcoma development by inhibitingdifferentiationrdquo Cancer Science vol 101 no 5 pp 1108ndash11162010

[73] J Wesche K Haglund and E M Haugsten ldquoFibroblast growthfactors and their receptors in cancerrdquo Biochemical Journal vol437 no 2 pp 199ndash213 2011

[74] M Wachtel J Rakic M Okoniewski P Bode F Niggli andB W Schafer ldquoFGFR4 signaling couples to Bim and not Bmfto discriminate subsets of alveolar rhabdomyosarcoma cellsrdquoInternational Journal of Cancer vol 135 no 7 pp 1543ndash15522014

[75] P L Puri ZWu P Zhang et al ldquoInduction of terminal differen-tiation by constitutive activation of p38 MAP kinase in humanrhabdomyosarcoma cellsrdquo Genes and Development vol 14 no5 pp 574ndash584 2000

[76] A M Tremblay E Missiaglia G G Galli et al ldquoThe Hippotransducer YAP1 transforms activated satellite cells and is apotent effector of embryonal rhabdomyosarcoma formationrdquoCancer Cell vol 26 no 2 pp 273ndash287 2014

[77] R N Judson A M Tremblay P Knopp et al ldquoThe hippo path-way member Yap plays a key role in influencing fate decisionsin muscle satellite cellsrdquo Journal of Cell Science vol 125 no 24pp 6009ndash6019 2012

[78] L E S Crose K A Galindo J G Kephart et al ldquoAlve-olar rhabdomyosarcoma-associated PAX3-FOXO1 promotestumorigenesis via Hippo pathway suppressionrdquo Journal ofClinical Investigation vol 124 no 1 pp 285ndash296 2014

[79] B Belyea J G Kephart J Blum D G Kirsch and C MLinardic ldquoEmbryonic signaling pathways and rhabdomyosar-coma contributions to cancer development and opportunitiesfor therapeutic targetingrdquo Sarcoma vol 2012 Article ID 40623913 pages 2012

14 Sarcoma

[80] J Huertas-Martinez S Rello-Varona D Herrero-Martin et alldquoCaveolin-1 is down-regulated in alveolar rhabdomyosarcomasand negatively regulates tumor growthrdquo Oncotarget vol 5 no20 pp 9744ndash9755 2014

[81] S Rossi P L Poliani CMissale EMonti andA Fanzani ldquoCav-eolins in rhabdomyosarcomardquo Journal of Cellular andMolecularMedicine vol 15 no 12 pp 2553ndash2568 2011

[82] P G Tsioli E S Patsouris C Giaginis and S E TheocharisldquoDNA repair systems in rhabdomyosarcomardquo Histology andHistopathology vol 28 no 8 pp 971ndash984 2013

[83] E Gatzidou C Michailidi S Tseleni-Balafouta and S Theo-charis ldquoAn epitome of DNA repair related genes and mecha-nisms in thyroid carcinomardquo Cancer Letters vol 290 no 2 pp139ndash147 2010

[84] R Rota R Ciarapica A Giordano L Miele and F LocatellildquoMicroRNAs in rhabdomyosarcoma pathogenetic implicationsand translational potentialityrdquoMolecular Cancer vol 10 article120 2011

[85] A Gougelet J Perez D Pissaloux et al ldquoMiRNA profiling howto bypass the current difficulties in the diagnosis and treatmentof sarcomasrdquo Sarcoma vol 2011 Article ID 460650 13 pages2011

[86] R Ciarapica G Russo F Verginelli et al ldquoDeregulated expres-sion of miR-26a and Ezh2 in rhabdomyosarcomardquo Cell Cyclevol 8 no 1 pp 172ndash175 2009

[87] L Li A L Sarver S Alamgir and S Subramanian ldquoDownreg-ulation of microRNAs miR-1 -206 and -29 stabilizes PAX3 andCCND2 expression in rhabdomyosarcomardquo Laboratory Investi-gation vol 92 no 4 pp 571ndash583 2012

[88] S-Y Ng G K Bogu B Soh and L W Stanton ldquoThe longnoncoding RNA RMST interacts with SOX2 to regulate neu-rogenesisrdquoMolecular Cell vol 51 no 3 pp 349ndash359 2013

[89] C A Lynch B Tycko T H Bestor and C P Walsh ldquoReac-tivation of a silenced H19 gene in human rhabdomyosarcomaby demethylation of DNA but not by histone hyperacetylationrdquoMolecular Cancer vol 1 article 2 2002

[90] P Onyango and A P Feinberg ldquoA nucleolar protein H19opposite tumor suppressor (HOTS) is a tumor growth inhibitorencoded by a human imprinted H19 antisense transcriptrdquoProceedings of the National Academy of Sciences of the UnitedStates of America vol 108 no 40 pp 16759ndash16764 2011

[91] C E Albacker N Y Storer E M Langdon et al ldquoThe his-tone methyltransferase SUV39H1 suppresses embryonal rhab-domyosarcoma formation in zebrafishrdquo PLoS ONE vol 8 no 5Article ID e64969 2013

[92] N Y Storer R M White A Uong et al ldquoZebrafish rhab-domyosarcoma reflects the developmental stage of oncogeneexpression during myogenesisrdquo Development vol 140 no 14pp 3040ndash3050 2013

[93] M E Hatley W Tang M R Garcia et al ldquoA mouse modelof rhabdomyosarcoma originating from the adipocyte lineagerdquoCancer Cell vol 22 no 4 pp 536ndash546 2012

[94] V Hosur A Kavirayani J Riefler et al ldquoDystrophin and dysfer-lin doublemutantmice a novelmodel for rhabdomyosarcomardquoCancer Genetics vol 205 no 5 pp 232ndash241 2012

[95] W Goessling T E North and L I Zon ldquoNew waves of discov-ery modeling cancer in zebrafishrdquo Journal of Clinical Oncologyvol 25 no 17 pp 2473ndash2479 2007

[96] B P Rubin K Nishijo H-I H Chen et al ldquoEvidence for anunanticipated relationship between undifferentiated pleomor-phic sarcoma and embryonal rhabdomyosarcomardquo Cancer Cellvol 19 no 2 pp 177ndash191 2011

[97] S Z Kaylani J Xu R K Srivastava L Kopelovich J G Presseyand M Athar ldquoRapamycin targeting mTOR and hedgehog sig-naling pathways blocks human rhabdomyosarcoma growth inxenograft murine modelrdquo Biochemical and Biophysical ResearchCommunications vol 435 no 4 pp 557ndash561 2013

[98] S Q Li A T Cheuk J F Shern et al ldquoTargeting wild-type andmutationally activated FGFR4 in rhabdomyosarcoma with theinhibitor ponatinib (AP24534)rdquoPLoSONE vol 8 no 10 ArticleID e76551 2013

[99] K Miyoshi K Kohashi F Fushimi et al ldquoClose correlationbetween CXCR4 and VEGF expression and frequent CXCR7expression in rhabdomyosarcomardquo Human Pathology vol 45no 9 pp 1900ndash1909 2014

[100] ADMarshall F Picchione R I KGeltink andGCGrosveldldquoPAX3-FOXO1 induces up-regulation of Noxa sensitizing alve-olar rhabdomyosarcoma cells to apoptosisrdquo Neoplasia vol 15no 7 pp 738ndash748 2013

[101] F Perut S Avnet C Fotia et al ldquoV-ATPase as an effective thera-peutic target for sarcomasrdquo Experimental Cell Research vol 320no 1 pp 21ndash32 2014

Submit your manuscripts athttpwwwhindawicom

Stem CellsInternational

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

MEDIATORSINFLAMMATION

of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Behavioural Neurology

EndocrinologyInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Disease Markers

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

BioMed Research International

OncologyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Oxidative Medicine and Cellular Longevity

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

PPAR Research

The Scientific World JournalHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Immunology ResearchHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal of

ObesityJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Computational and Mathematical Methods in Medicine

OphthalmologyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Diabetes ResearchJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Research and TreatmentAIDS

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Gastroenterology Research and Practice

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Parkinsonrsquos Disease

Evidence-Based Complementary and Alternative Medicine

Volume 2014Hindawi Publishing Corporationhttpwwwhindawicom

Page 2: Review Article Rhabdomyosarcoma: Advances in Molecular and

2 Sarcoma

Table 1 Alterations of chromosome in RMS by CGH in recent 5 years

Materials Gain Amplification Loss Deletion Reference

25 RMS samples 2p 12q 6p 9q 10q 1p2q 6q 8q 15q 18q 3p 11p 6p Li et al 2009 [16]

13 RMS cell lines

1p213ndash132 1q126q26ndash27 7q213ndash3111q41 2p243 8q241220q132 20q1332

2p243 (MYCN)8p1123ndash1121

(FGFR1) 12q133(CDK4) 19q12 20q

3p142ndash1224q27ndash3236p251ndash243

9p243ndash241 13q143

Missiaglia et al 2009[17]

57 ARMSsamples 12q15 2p24 12q13ndashq14 Barr et al 2009 [18]

128 primaryRMS samples 7 8 11 20

2p241 (MYCN)8p112ndashp111 (FGFR1)12q133ndash141 (CDK4)MDM2 (12q143ndashq15)

Williamson et al2010 [12]

26 frozenprimary ERMSsamples

8 2 11 12 13 19 20

2p21 2q35 2q1422q361 5q352ndashq353

11p112 11q24212q133

6 9 10 14 15 16 181p3623 1q321 3p1424q351ndash352 9p21317q112 22q1331

Paulson et al 2011[13]

39 RMS samples 12q133 12q133ndashq14112q141 17q251

2q1312 12q13312q133ndashq141

9p12ndashp11210q1121ndashq112214q3233 16p112

22q111

1p211 2q141 5q1329p12 9q12 Liu et al 2014 [11 15]

RMS cellsderived fromrefractory RMS

NACA HSD17B6SDR9C7 RDH16GPR182 ZBTB39TAC3 MYO1A

NAB2 STAT6 LRP1

Park et al 2014 [14]

20 RMS samplesof Chinesepatients

12q2431 17q2511q211 7q112312q133ndashq141

9p133 12q133ndashq14112q15 16p1311

5q132 15q11214q3233 (IGHG

IGHM)

1p3633 1p131 2q1115q132 8p231 9p243

16p112Liu et al 2014 [11 15]

Since the 1970s the Intergroup RhabdomyosarcomaStudy Group (IRSG) has conducted a series of clinical trialscomparing risk-base and has established a series of treatmentguidelines [9] Currently multidisciplinary managementincluding chemotherapy and surgery with or without radia-tion has become the standard treatment for RMS The 5-yearsurvival rate of RMS has increased from 25 in 1970 up to60 since 2000 [1 10]However there has been little improve-ment in the oncological outcome of patients with RMS inrecent years Drug resistance andmetastatic disease representthe two most common phenomena for therapy failure Somerandomized chemotherapy trials have failed to improve out-come despite the introduction of newer or more intensivetherapies Thus there is an urgent need for alternative moreeffective treatment strategies Recent molecular and geneticanalysis of these tumors has produced substantial newinsights into molecular cell biology molecular cytogeneticsand tumorigenesis of RMS leading to a better understandingof RMS development at the molecular level These advancesmay ultimately lead to better clinical understanding and topotentially developing more potent targeted therapies Thepurpose of this review is to summarize these most recentfindings in RMS

2 Novel Discoveries of ChromosomalAlterations in RMS

Malignant transformation occurs cytogenetically due to theaccumulation of somatic mutations by the acquisition oftumor-specific chromosomal translocations

21 Gains and Losses of Chromosomes in RMS ComparativeGenomic Hybridization (CGH) analysis has revealed that allRMS have specific gains and losses [11ndash18] (Table 1) ERMSfrequently exhibits gains or losses of specific whole chromo-somes whereas ARMS is characterized by the presence ofregions of genomic amplification [11 19] The focal regionsand genes most of which have frequent gains and ampli-fications include 12q133ndashq141 and 8p112ndash112 and CDK4MYCN GLI MDM2 FGFR1 and FGFR4 respectively Fre-quently the genes differentially expressed in subtypes ofRMS particularly when they are from chromosomal regionsshow a high level of gains in cell lines Previous CGH studieshave shown that ARMS tumors tend to have fewer copynumber variants than ERMS tumors [12 20] For examplefrequent gains were detected in TYROBP HCST LRFN3and ALKBH6 (19q1312) in ERMS but not in ARMS [15]

Sarcoma 3

PAX3

Chromosome 2

PAX3-FKHR

t(213)(q37q14)

NCOA1

PAX7

Chromosome 1

PAX7-FKHR

t(113)(p36q14)

PAX3

Chromosome 2

FKHR

Chromosome 13

PAX3-NCOA1

t(22)(q35q23)

Paired domain Homeodomain

DNA binding domain Transactivation domain

PB

PB

PB

PB HD

HD

HD

HD TAD

TAD

TAD

TAD

TAD

TAD

HD

HD

PB

PB

100 amino acids

Fusion site

Fusion site

FD

DNA binding heterodimerization domain Nuclear receptor interaction domain

LXXLL HAT

HAT

bHLHPAS

Fusion site

(a)

(b)

(c)

Figure 1The chromosomal rearrangements in ARMS 80 of ARMS classified as translocation-positive ARMS carry characteristic chromo-somal translocations demonstrated as t(213)(q35q14) t(113)(p36q14) and (22)(q35p23) In (a) and (b) the translocations fuse the FKHR(a member of the forkheadHNF-3 transcription factor family) locus on chromosome 13 to either PAX3 on chromosome 2 or PAX7 onchromosome 1 In (c) the translocation generated a fusion protein composed of PAX3 and the nuclear receptor coactivator NCOA1 havingsimilar transactivation properties as PAX3FKHR

These studies have identified a number of genetic alterationsin RMSMany of these chromosomal changesmay be respon-sible for tumor progression and proliferation These geneticalterations may be potential treatment targets in RMS [11]

22 Chromosomal Translocations in ARMS Chromosomalanalyses have demonstrated two translocations associatedwith ARMS t(213)(q35q14) and t(113)(p36q14) [21] Initialstudies detected these two gene fusions in 80 of ARMS[22] These characteristic chromosomal translocations areadjacent to the 51015840 DNA-binding domains of PAX a memberof the paired box transcription factor family and the trans-activation domain at the 31015840 end of FKHR a member of theforkheadHNF-3 transcription factor family Approximately75 of these structural rearrangements translocate the PAX3gene at 2q35 to the FKHR gene at 13q14 as t(213)(q35q14)less frequently in the other 25 the t(13)(q36q14) transloca-tion fuses PAX7 to FKHR [23] Previous studies have demon-strated that PAX3FKHR fusion gene status significantlyimproves current risk stratification while the presence ofPAX7FKHR and rarer variant fusion gene products requirefurther investigation [24] The remaining 20 of ARMS isPAX gene fusion-negative (PFN) and forms a more hetero-geneous group which remains a challenge to detect due to

the lack of consistent chromosomal rearrangements PFNARMS has a similar clinical course to ERMS which suggeststhat the fusion gene status provides more accurate informa-tion about patient outcomes than did the histologic subtypeDespite the low overall burden of somatic mutations infusion-positive RMSmultiple genes were recurrently alteredincluding NRAS KRAS HRAS FGFR4 PIK3CA CTNNB1FBXW7 and BCOR [25]

Another novel translocation t(22)(q35p23) was identi-fied in ARMS biopsy samples by gene expression signatures[26] The chromosomal translocation generates a fusion pro-tein composed of PAX3 and the nuclear receptor coacti-vator NCOA1 which has similar transactivation propertiesas PAX3FKHR [26] These biologic effects contribute totumorigenesis bymodulatingmyogenic differentiation alter-ing growth and apoptotic pathways and stimulating motilityand other metastatic pathways (Figure 1)

23 Chromosomal Alterations in ERMS ERMS exhibits a lossof imprinting (LOI) leading to a twofold gene dosage effect[27] Most tumors have at least one 15-Mb region with loss ofheterozygosity (LOH) along chromosome 11 [27] The allelo-type of ERMS demonstrates a high frequency of LOH onchromosomes 11p 11q and 16q [28] ERMS tumorigenesis

4 Sarcoma

Table 2 The histological types of rhabdomyosarcoma

Histological type Predilection population Predilection site Risk category Genetic change

ERMS Infants or children under 10years old Head and neck region Intermediate LOH at chromosome 11p155

ARMS Adolescents and young adults Deep tissue of extremity High t(213)(q35q14) t(113)(p36q14)

PRMS Adult males Throughout the body HighJUN (1p31) MYC (8q24)CCND1 (11q13) INT2 (11q133)MDM2 (12q143ndashq15)

SRMSIn the first decade of life with asecond mode centered aroundthe fifth decade

The extremities headand neck Superior SRF-NCOA2 MYOD1 mutation

ERMS embryonal rhabdomyosarcomaARMS alveolar rhabdomyosarcomaPRMS pleomorphic rhabdomyosarcomaSRMS spindle cellsclerosing rhabdomyosarcoma

can result from the inactivation of the parental bias of chro-mosome 11p15 which is the most common rearrangementin ERMS [29] The proportion of ERMS with LOH alongchromosome 11 is considerably higher than in other subtypes[27] In histopathological analysis ERMS expresses lowPAX3levels and elevated PAX7 levels [30] Hosoi et al identified ahidden 2q35 breakpoint as a novel PAX3 rearrangement incomplex chromosomal translocations in ERMS [31]

24 Chromosomal Alterations in Other RMS There are lim-ited studies on the biological pathways involved in othersubtypes of RMS compared with the two major subtypesARMS andERMS Fluorescence In SituHybridization (FISH)reveals amplification of JUN (1p31) MYC (8q24) CCND1(11q13) INT2 (11q133) MDM2 (12q143ndashq15) and MALT(18q21) in these tumor cells contributing to the pathogenesisof PRMS [32] MYOD1 homozygous mutations are alsoreported as frequent recurrent and pathognomonic eventsin adult-type SRMS [33 34] (Table 2) Furthermore four outof five pediatric tumors showedMYOD1mutations in a study[34] Yoshida et al described a 8q13 locus (NCOA2) generearrangement in a small subset of SRMS occurring uniquelyin the infantilecongenital setting fused with key transcrip-tion factors involved in skeletal muscle differentiation suchas SRF and TEAD1 [35] They also identified NCOA2 as acandidate PAX3 partner geneThePAX3-NCOA2 fusion geneplays a dual role in the tumorigenesis of RMS promotion ofthe proliferation and inhibition of the myogenic differenti-ation of RMS cells [35] As PAX3-NCOA2-induced tumorsgrow more slowly SRMS that are PAX3-NCOA2 fusion-positive could be associated with a very favorable prognosis[36]

Not all RMSs occur as sporadic primary tumors Occa-sionally RMS inherits a mutant gene as part of an establishedfamilial syndrome For example Beckwith-Wiedemann Syn-drome (BWS) with RMS involves dysregulation or alterationof imprinted genes in the 11p155 chromosomal regionincluding IGF2 H19 and CDKN1C (p57KIP2) [37]

3 Cell of Origin in RMS

There have been a number of studies aimed at deciphering thecell origin for RMS [56ndash61]

31 Myogenic Differentiation in the Tumorigenesis Severalstudies have focused on elucidating the mechanisms govern-ing the impaired myogenic program in RMS [62 63] (Fig-ure 2) RMSoriginates as a consequence of regulatory disrup-tion of the growth and differentiation of myogenic precursorcells Progenitor cells reside in muscle and their activationresults in either proper myogenesis or aberrant signalingpathways leading to the development of RMS Based on theskeletal muscle lineage a complete transcriptome analysis ofRMS was performed to compare normal and fetal muscles[56] The high degree of similarity between fetal muscle andRMS expression profiles reflects the undifferentiated myo-genic nature of RMS The genes exclusively upregulated inRMS including FGFR4 NOTCH2 UBE2C UHRF1 andYWHAB genes contribute to the failure of RMS cells to com-plete normal skeletal muscle development and progress toan alternative fate [56] RMS cells represent an arrested statein the development of normal skeletal muscle with regionalsilencing of differentiation factors leading to the maturationdefect in RMS [57] The expression pattern of muscle-specific proteins regulating myogenic differentiation hasbeen extensively examined in RMS The family of myogenictranscription factors (MYOD MYF5 myogenin and MRF4)are considered to be responsible for the determination of stemcells intomyoblasts anddifferentiation intomyocytes [58 59]All these factors are activated during the onset or progressionof myogenesis and are subsequently silenced to reach a finalmuscular differentiation There are statistical differences inthe different subtypes of RMS tumors with MYOD or myo-genin staining patterns Amplification of MDM2 in an RMScell line interferes with MYOD activity and consequentlyinhibits overt muscle cell differentiation [61] Interleukin-4receptor (IL-4R) has also been proposed to be important for

Sarcoma 5

Table 3 Studies identifying stem cells in RMS

Markersubstrate Source Stem cell gene Functional characterization Reference

CD133 Orthotopicxenograft model

OCT4 NANOG c-MYCPAX3 and SOX2

Correlating with poor overallsurvival

Walter et al2011 [38]

PAX-FKHR Bone marrow ofC57BL6 mice

Myf5 MEF2 MyoD andmyogenin

Determining the molecularmyogenic and histologicphenotype of ARMS

Ren et al 2008[39]

V-ATPase RD cell line NANOG and OCT34

Driving mechanisms of areduced sensitivity to anticancerdrugs and activities related toinvasion and metastasis

Salerno et al2014 [40]

Survm-CRAsKYM-1 cell line(FGFR3-positive)

FGFR3Therapeutic effectiveness againstall cell populations and increasedeffectiveness against CSCs

Tanoue et al2014 [41]

Migration

specification

Injured muscle

Terminal differentiation

Fusion

PAX7

Myoblasts

Myoblast

MyoD

MyoD

Myf5

PAX3

Rb

Ezh2

IGF2

HDAC4YY1

YY1

ERK

AKT

RMS

Myogenin

P38 kinase

Myostatin

Progenitor

cell

N-RAS

Myofibers

FGFR4

UBE2C

UHRF1

NOTCH2

YWHAB

Satellite cells

RUNX1 MEF2C

JDP2 NFIC

Myocytes

Restoration of proliferation blockade of

differentiation

Proliferatio

n

Cell circle arrestEarlydifferentiation

TGF-1205731

Figure 2 Myogenic pathways in the tumorigenesis of RMS In aberrant neoplastic condition progenitor cells residing in muscle result inaberrant pathways which lead to malignant transformation and fail to differentiate proliferate uncontrollably and form RMS Sharp arrows(rarr) indicate upregulationactivation and blunt arrows (perp) indicate downregulationinhibition

the maturation of myotubes [64] An IL-4R blockade mighthelp therapeutically to modulate the expression of myogenictranscription factors (MYOD or myogenin) [57 64]

32 Potential Cancer Stem Cell in RMS Cancer Stem Cells(CSCs) may contribute to inherent refractory responses tocurrent therapies andmetastasis [65]Many CSCmodels playan important role in the development of RMS [38ndash41 66](Table 3)

A study explored a potential important role for mesen-chymal stem cells (MSCs) as the cell of origin of ARMS [67]Ren et al confirmed that PAX-FKHR fusion genes commitMSCs to a myogenic lineage by inhibiting terminal differen-tiation and contributing to ARMS formation [39] Anotherstudy showed that PAX-FKHR induces skeletal myogenesisin MSCs by transactivating MYOD and myogenin and trans-forms mesenchymal progenitor cells to the skeletal muscle lin-eage leading to malignant formation resembling ARMS [39]

6 Sarcoma

METCDKN2AB

PAX37-FKHR

MDM2

RAS

Raf

GLI1

PDK1p53Rb

Hh

Myo MEF2

p21

Ptch

FGFR4

IGF1R

p16INK4Ap14ARF

p19ARF

IGF2

FGF

Figure 3 Genetic analyses of RMS have pinpointed several common alterations including inactivation of a master regulator of p53 andRb pathways CDKN2AB and activation of FGFR4 RAS and Hedgehog (Hh) signaling The modifications of these pathways influenceoncogenesis and metastatic potential

As a human hematopoietic CSC marker Walter et alproposed that upregulated CD133 in ERMS can act as aprognostic marker and might help with the development ofnovel targeted therapies for ERMS [38]

4 Signaling Pathway Alterations in RMS

Recent studies have looked into signaling pathway alterationsand their downstream effects in RMSThese new findings notonly helped to improve the understanding of this malignancybut also offered novel potential therapeutic strategies forimproved treatment for patientswithRMS [68 69] (Figure 3)

41 RAS Signaling Pathway RASmutations commonlymain-tain the protein in its GTP bound state and therefore renderit constitutively active in RMS Zhang et al identified RASfamily members NRAS KRAS and HRAS in the RASpathway as themost commonlymutated genes in ERMS [70]the mutation status of these genes is significantly associatedwith the risk of ERMS development [70] A study foundthat a majority of ERMS demonstrated activation of the RASpathway exclusively either by homozygous deletion of NF1or by point mutations in one of the RAS family members asdescribed above [13] Despite remarkable genetic and molec-ular heterogeneity most (93 4144) RMS tumors hijackeda common receptor tyrosine kinaseRASPIK3CA geneticaxis [25] It could occur via two alternative mechanismsrearrangement of a PAX gene and accumulation of mutationsthat were downstream targets of the PAX fusion protein

Skeletal muscle cells have a robust antioxidant defensesystem to protect the DNA lipids and proteins from thedeleterious effects of excess Reactive Oxygen Species (ROS)

Cancer cells also have elevated ROS due to their increasedmetabolic activity oncogenic stimulation andmitochondrialdysfunction These findings implicate RAS mutations andoxidative stress as potential therapeutic targets for high-riskERMS

42 IGF Signaling Pathway There is clear preclinical datathat supports the involvement of the Insulin-like GrowthFactor (IGF) signaling pathway in RMS tumorigenesis andprogression [69] Zhu and Davie demonstrated that IGFpromotes the proliferation of RMS cells while blocking IGFsignaling interferes with cell growth in vivoThe IGF receptorIGF-1R is highly overexpressed both on the cell surface andin the nucleus of RMS cells furthermore RMS cell lines aresensitive to IGF-1R inhibitionThe IGF-2 locus shows a loss ofimprinting in both ERMS andARMS tumors and the expres-sion of PAX3-FKHR has been demonstrated to upregulateIGF-2 and activate IGF pathway in ARMS [69] Mariannaet al found that IGF-2 and tumor suppressors p19Arf andp21Cip1 are overexpressed and prominently upregulated inRMS mouse models [71]

43 TGF-120573 Signaling Pathway The regulatory role of Trans-forming Growth Factor-120573 (TGF-120573) in RMS has been recentlystudied [72] Wang et al demonstrated that the expression ofTGF-1205731 is significantly higher in RMS than in normal skeletalmuscle The inhibition of TGF-1205731 expression by shRNA-expressing vectors reverses themalignant behavior of RMSbyinhibiting cell growth and inducingmyogenic differentiationTGF-1205731 shRNA induces myogenin expression a regulator ofmyogenic differentiation genes Myogenin acts as a target fornegative regulation of myogenesis by TGF-1205731 signals with

Sarcoma 7

a differentiation regulatory cascade These results suggestthat the TGF-1205731 signaling pathway disrupts the differentia-tion of myogenic progenitors leading to the development ofRMS

44 FGF Signaling Pathway Fibroblast Growth Factor (FGF)is crucial in embryonic development and functions to driveproliferation FGFs and their receptors (FGFRs) are essentialregulators in the processes of proliferation antiapoptosisdrug resistance and angiogenesis in RMS [69] RMS over-expresses the receptor tyrosine kinase FGFR4 which causesautophosphorylation and constitutive signaling in correlationwith poor differentiation and decreased survival [73] Recentwork has shown that FGF signaling can prevent ARMS cellsfrom apoptosis induced by targeting the IGF1-R-PI3K-mTORpathway [74]

45 ERK Signaling Pathway Previous studies have shownthat elevated myostatin expression deregulates ExtracellularRegulated Kinase (ERK) signaling and deficient activationof the p38 pathway contributes to the differentiation blockin RMS cells [62] The ERK pathway is frequently highlyactivated in RMS cells from embryonic derivation due to thepresence of activating RASmutations leading to reduced dif-ferentiation by blocking the p38 pathway [75]These findingsindicate that interventions that target themyostatinERKp38network could be an effective way to promote differentiationof RMS

46 Hippo Signaling Pathway Many upstream signal trans-duction proteins in response to cues from the cellular micro-environment regulate the core Hippo pathway [76] TheHippo pathway may limit tumorigenesis by inducing thecytosolic localization of the transcriptional cofactor Yes-Associated Protein 1 (YAP1) High YAP levels and activitiesincrease activated satellite cells and prevent differentiationby activating genes that inhibit differentiation [77] YAP1-TEAD1 was found to upregulate proproliferative and onco-genic genes and maintain the ERMS differentiation blockby interfering with MYOD1 and MEF2 prodifferentiationactivities [76] Inhibition of theHippo pathway is exhibited inRMS through downregulation of Hippo pathway tumor sup-pressors or upregulation of YAP [78] These data suggest thatHippo pathway dysfunction promotes RMS development

47 Notch Signaling Pathway and Wnt Signaling PathwayNotch pathway as an embryonic signaling pathway promotesmuscle stem cell maintenance by inhibiting myogenic earlydifferentiation and expanding pools of progenitor cells dur-ing both embryogenesis and postnatal muscle regenerationSimilar to Notch Wnt has various roles in embryonic fetaland neonatalmyogenesis during skeletalmyogenesis As bothpathways are regulators of myogenic lineage determinationand maturation RMS can arise from disordered regulationof these normal developmental pathways [79]

48 Caveolins in RMS Recent findings have shown that cav-eolins are expressed in a cell stage-dependent manner inRMS [80 81] Caveolins are scaffolding proteins that regulate

several targets and pathways (such as p53 IGF RASERK andTGF-120573myostatin signaling) associated with RMS develop-ment and therefore loss or gain of caveolin function is ableto generate multiple effects on the tumor behaviorThereforerecognizing their precise roles in RMS progression will becrucial to elaborate targeted therapies particularly as oneof the three different isoforms CAV1 could act as a potenttumor suppressor in ARMS Inhibition of CAV1 function cancontribute to aberrant cell proliferation leading to ARMSdevelopment [80]

49 DNA Repairing System Some studies indicate that RMSis characterized by germline mutations of DNA repair genes[82 83] DNA repair gene alterations in RMS occur sec-ondarily to malignant transformation The modificationsin DNA repair enzyme activity can result in resistance toadjuvant treatment in RMS tumor cells which limits theprognosis of RMS There are two pathways for repairingDNA breaks directly without affecting DNA structure andindirectly by DNA phosphodiester backbone cleavage [82](Figure 4) Direct repair includes repair during replicationand enzymatic repair Indirect repair is comprised of exci-sion repair systems including base excision repair (BER)nucleotide excision repair (NER) mismatch repair (MMR)and recombination repair (RR) [83] Defective DNA repairmechanisms result from point mutations or LOH in RMSand contribute to tumorigenesis MGMT and MMR proteinactivity and expression levels are used as predictor indices fortherapy outcome in RMS BER as the major DNA repair sys-tem against damage resulting from cellular metabolism canreverse the cytotoxic effects of alkylation agents used suchas antineoplastics subsequent tumor progression and deathimplicates an RMS mechanism of resistance to chemother-apeutic agents This may provide novel individualized ther-apeutics targeting downregulation of activated DNA repairenzymes or upregulation of DNA repair deficiencies [82]

5 MicroRNA (miRNA miR)Expression in RMS

Muscle-specific and ubiquitously expressed miRs appeardownregulated in RMS tumors and cell lines compared withthe normal counterparts These miRs often play crucial rolesas antioncogenes Inhibition of these miRs contributes toenhanced tumorigenesis through the modulation of diversemolecular pathways Upregulation of prooncogenic miRs hasbeen detected in RMS recently as well [84] (Table 4)

51 Antioncogenic miR in RMS Gain-of-function experimentshave demonstrated that reexpression of selected ldquotumor sup-pressorrdquo miRs impairs the tumorigenic behavior of RMS cells[84 85] As myo-miR family members miR-1 miR-133a andmiR-206 have been demonstrated to be downregulated inRMS and shown to have activity on mRNA expression bytargeting c-Met [42 43 86] Inhibition of thesemyo-miRNAsmay cause aberrant cell proliferation and migration in myo-genesis leading to RMS development especially for ERMSThere have been more miRs found as tumor suppressors inthis malignancy including miR-29 miR-450b-5p miR-203

8 Sarcoma

Table 4 MicroRNAs involved in myogenesis and RMS development

miRNA Expression Target Function Reference

miR-1 and miR-133a Downregulation MYH9Myogenic miRNA inhibit differentiationand promote proliferation in myogenesiscytostatic

Rao et al 2010 [42]

miR-206 Downregulation cMET Promote differentiation and proliferationin myogenesis

Yan et al 2009 [43]

miR-29 Downregulation HADC4 YY1 EZH2 Promote stabilization of RMS phenotype Marchesi et al 2014 [44]

miR-450-5p Downregulation ENOX PAX9 Promote differentiation and progression Sun et al 2014 [45]

miR-203 Downregulation JAK STAT Notch Inhibit differentiation and proliferation inmyogenesis tumor suppressor

Diao et al 2014 [46]

miR-214 Downregulation N-Ras Inhibit tumor cell growth and inducemyogenic differentiation and apoptosis

Huang et al 2014 [47]

miR-183 Upregulation EGR1 Promote migration and metastasis Sarver et al 2010 [48]

PMS2

MGMT

HR

Mismatch Recombination

NHEJ MMEJ

p53

Excision

BER NER

RMS

DNA repair

Direct repair Indirect repair

repair repairrepair

Figure 4The DNA repair systems in RMSThere are two pathways repairing the DNA lesions directly without affecting DNA structure andindirectly by DNA phosphodiester backbone cleavage The modifications in DNA repair enzymes expression or activity lead to resistanceto chemotherapy and radiation in RMS tumor cells MGMT O6-methylguanine-DNA methyltransferase BER base excision repair NERnucleotide excision repair HR homologous recombination NHEJ nonhomologous end-joining MMEJ microhomology-mediated end-joining

and miR-214 [44 46 47 84 87] They are all significantlydownregulated in RMS compared with normal skeletal mus-cles and function to inhibit tumor cell growth and inducemyogenic differentiation and apoptosis in RMS

52 Oncogenic miRNA in RMS The transcription factor EGR1is a tumor suppressor gene that is downregulated in RMS [48]miR-183 functions as an oncogene by targeting EGR1 and pro-moting tumor cell migration Either by direct anti-miR treat-ment or by indirect mechanisms that decrease transcriptmiR-183 targeted treatmentsmight provide a potential optionto RMS patients

The basic strategy of current effective miR-based treat-ment studies is either efficient reexpression of miRs or restor-ing the function of miRs to inhibit the expression of certain

protein-coding genes and promote muscle differentiationMoreover miR expression profiling in tumors and possiblytheir detection in peripheral blood during treatment may beable to predict the response to chemotherapy or radiotherapyand be useful as a prognostic signature for the development oftreatment resistanceThe prognostic value of miR expressionlevels in RMS is a powerful tool in creating a better-tailoredstrategy for particular subsets of patients

53 Long Noncoding RNAs in RMS Long noncoding RNAs(LNCRNAs) are abundant in the mammalian transcriptomeand have been shown to play a role in RMS development [8889]TheH19 gene localized within a chromosomal region onhuman chromosome 11p15 encodes an imprinted untrans-lated RNA [90] H19 opposite tumor suppressor (HOTS)

Sarcoma 9

Table 5 In vivo animal models of RMS

Model TargetModel origin Reference

Inactivation Expression

DrosophilaPAX-FKHR Bipartite Gal4-UAS

expression systemGalindo et al2006 [49]

PAX7-FOXO1 rols Chromosomal deletionDf(3L)vin5

Avirneni-Vadlamudi et al2012 [50]

Zebrafishrag2 promoter c-Myc kRASG12D

Chen andLangenau 2011[51]

MAPKERK andAKTS6K1 PD98059 TPCK Tg(hsp70-HRASG12V) Le et al 2012

[52]

Mouse

Sufu N-myc Sfrp1 Ptch2and cyclin D1 Sufu+minus Lee et al 2007

[53]

Ink4aArf and Trp53 Pax3FKHR Pax3P3Fpwt Keller et al2004 [54]

Wnt120573-cateninsignaling pathway Wnt2 p53minusminusc-fosminusminus Singh et al 2010

[55]

a tumor growth inhibitor is encoded by an imprinted H19antisense transcript Overexpression of HOTS inhibits RMStumor cell growth Silencing HOTS by RNAi was shown toincrease in vitro colony formation as well as in vivo tumorgrowth of RMS [90]

6 In Vivo Models of RMS

Several in vivo models have recently been developed inDrosophila zebrafish and mice to further understand themolecular and cellular biology of RMS tumorigenesis [49ndash55 91ndash94] (Table 5)

61 Drosophila Models in RMS Despite the evolutionarydistance a strong conservation of genes pathways andregulatory molecular networks have been demonstratedbetween flies and humans Many human disease genes haverelated sequences in Drosophila In contrast to mammalianmodels the generation of Drosophila mutants is easy cheapand fast

For a better understanding of the pathogenic conse-quences of PAX-FKHR expression Drosophila was chosenbecause the animalrsquos transparent outer cuticle allows forsimple real-time monitoring of muscle abnormalities elicitedby PAX-FKHR including subtle or focal changes [49] Themutation in the myoblast fusion gene rolling pebbles (rols)was reported to dominantly suppress PAX-FKHR1-inducedlethality by using a Drosophila model of PAX-FKHR1-mediated transformation [50] PAX-FKHR1 signaling upreg-ulates the TANC1 gene and blocks myoblasts from terminaldifferentiation However downregulating the TANC1 genecould cause RMS cells to lose their neoplastic state undergofusion and form differentiation This novel finding uncov-ered a PAX-FKHR1-TANC1 neoplasia axis collaboratively ina Drosophila model and loss-gain-of-function studies inmammalian platforms [50]

62 Zebrafish Models in RMS In contrast to other mod-els the short tumor onset in zebrafish allows for rapididentification of essential genes in various processes suchas tumor growth self-renewal and maintenance There areother advantages for using zebrafish in the study of humandisease and development including the ease and low cost ofraising large numbers of fish and the highly conserved geneticand biochemical pathways between zebrafish and mammals[95]

As zebrafish RMS is highly similar to human ERMSby using fluorescent transgenic approaches it is more con-venient to understand histogenesis and different aspects oftumorigenesis in RMS A robust zebrafish model of RAS-induced RMS has been established sharing twomorphologicand immunophenotypic features resembling ERMS [51] Inone case both are associated with tissue-restricted geneexpression in RMS while in another both comprise a RAS-induced gene signature Cross-species microarray compar-isons confirm that conserved genetic tumor-specific andtissue-restricted pathways such as RAS and p53 pathwaysdrive RMS growth Tg(hsp70-HRASG12V) zebrafish embryoswere generated to evaluate gene expression that mimics RASpathway activation during tumorigenesis to defineRAS targetgenes [52] Another KRASG12D-induced zebrafish embryonalRMS was utilized to assess the therapeutic effects By theblockage of two major downstream signaling pathwaysMAPKERK and AKTS6K1 the model showed that inhibi-tion of translation initiation suppresses tumor cell prolifera-tion [52]

All the above zebrafish cancer models share similar cellu-lar features and molecular pathways with human RMS espe-cially ERMS and can demonstrate a response to therapiesin a similar manner with human RMS Thus a tremendousopportunity is available to implement these animal modelsinto different steps for novel targeted therapeutic develop-ment

10 Sarcoma

63 Mouse Models in RMS Mouse models established fromin vivo studies may contribute to the understanding of thegenetic basis in tumor development and progression andmayhelp to test the efficacies of novel antineoplastic agents

The importance of PAX3PAX7-FKHR fusion proteins inthe progression of ARMS tumorigenesis is evident from bothin vitro and in vivo studies Transgenic mouse models gener-ated with the PAX-FKHR fusion genemimic the formation ofhuman ARMS [67] A mouse model was developed with het-erozygous loss of Sufu a tumor suppressor inHedgehog (Hh)signaling combined with p53 loss driven ERMS with a lowpenetrance (9) [53] These studies indicate that the modelsconsistently formingARMSnot only need the introduction ofthe PAX-FKHR fusion gene but also need to be accompaniedby other genetic events such as p53 pathway disruption [54]

Compared with ARMS ERMS models are more complexto generate because of their lower tumor penetrance andlonger latency JW41 cells from the p53c-fos double mutantmice models resemble human ERMS cells morphologicallyand express similar characteristic markers [55] The overex-pression of the Wnt2 gene identified in JW41 cells confirmedin human RMS cell lines can allow for further analysis of theWnt signaling pathway Both of these results indicate that thedownregulation of theWnt pathway contributes to resistanceto apoptosis and the inhibition of myogenic differentiation inERMS

Using Ptch1 p53 andor Rb1 conditional mouse modelsand controlling prenatal or postnatal myogenic cell of originRubin et al demonstrated that the loss of p53 in maturingmyoblasts related to tumorigenesis of ERMS [96] They alsoindicated that Rb1 alteration with other oncogenic factorswas strongly associated with an undifferentiated phenotypeacting as a modifier In addition they highlighted a sub-set of p53-deficient ERMSs arising from Myf6-expressingmyoblasts which had the same latency with Pax7-expressingmurine satellite cells in the study

7 Potential Therapeutic Targets in RMS

Detection of genomic imbalances and identification of thecrucial effective genes can contribute to identifying novelpotential biomarkers and relevant targets for clinical therapyin RMS

Survivin-responsive conditionally replicating adenovi-ruses can regulate multiple factors (Survm-CRAs) [41]Although Survm-CRA can efficiently replicate and potentlyinduce cell death in RMS cells the cytotoxic effects are morepronounced in RSC-enriched or RSC-purified cells thanin RSC-exiguous or progeny-purified cells Injections ofSurvm-CRAs into tumor nodules generated by transplantingRSC-enriched cells induce significant death in RMS cells andregression of tumor nodulesThe unique therapeutic featuresof Survm-CRA include not only its therapeutic effectivenessagainst RMS but also its increasing effectiveness against CSCsuggesting that Survm-CRA may be a promising anticanceragent [41]

Rapamycin an inhibitor of mTOR can abrogate RMStumor growth in a xenograft mouse model [97] Thetumors in the rapamycin-treatedmice group show significant

reduction of proliferation and invasiveness and inductionof apoptosis The tumor growth inhibition is simultaneousassociated with the diminution of mTOR and Hh pathwayswhich are implicated in the pathogenesis of RMSThe resultsindicate that using rapamycin either alone or in combinationwith traditional chemotherapeutic drugs may represent apotential targeted agent for therapeutic intervention Ampli-fication and mutational activation of Fibroblast Growth Fac-tor Receptor 4 (FGFR4) in RMS cells promote tumor pro-gression [98] Ponatinib is the most potent FGFR4 inhibitorto block wild-type RMS cell growth mutate FGFR4 throughincreasing apoptosis and suppress the FGFR4 downstreamtarget STAT3 Ponatinib treatment slows tumor growth inRMS mouse models expressing mutated FGFR4 [98] Pona-tinib as an FGFR4 inhibitor shows potential to act as aneffective therapeutic agent for RMS tumorsThe expression ofCXC chemokine receptor 4 (CXCR4) CXCR7 and VascularEndothelial Growth Factor (VEGF) indicates poor prognosisin a variety of malignancies including RMS cell lines Mostsamples in a large series of clinical RMS cases show asso-ciation with high expression of these antagonists regardlessof their subtypes [99] However there are significant corre-lations with high expression of CXCR4 and VEGF in bothERMS and ARMS High VEGF expression is predictive ofadverse prognostic factors in RMS as the expression of theseangiogenesis factors was compared with clinicopathologicalparameters and prognosis Considering their overexpressionin RMS these chemokine receptors and VEGF could providepotential molecular therapeutic targets in RMS Some ARMScell lines have undergone apoptosis in response to antineo-plastic drugs such as bortezomib The proapoptotic BH3only family member NOXA acts as an upregulated proteindownstream PAX3-FKHR resulting in increased cell sensi-tivity to bortezomib Apoptosis in response to bortezomibcan be reversed by shRNA knockdown of Noxa PAX3-FKHR upregulation of Noxa creates a potential therapeuticinsight into inducing apoptosis in ARMS cellsThis apoptosispathway could represent a specific targeted therapy againstPAX3-FKHR-expressing ARMS cells [100]

Proton pump activation resulting from intracellular acid-ificationmay be an energetic mechanism to escape apoptosisV-ATPase is an ATP-driven proton pump acidifying theintracellular compartment and transports protons across theplasma membrane Esomeprazole as a V-ATPase inhibitor inthe PPI administration interferes with intensive ion trans-porter activity of RMS cells and induces remarkable cytotoxi-city RMS expresses a higher level ofV-ATPase comparedwithother sarcomasTherefore RMS should be very susceptible toesomeprazole treatment and V-ATPase could be consideredas a promising selective target for treatment [101]

8 Conclusion and Future Prospects

For RMS the current challenges include how to iden-tify promising novel therapeutics and integrate them intoexisting therapy For better understanding of congenitaland epigenetic modifications in the development of RMSthis review summarizes the recent genome-wide studies onmolecular and genetic alterations to decipher the underlying

Sarcoma 11

tumorigenesis mechanisms There is a link between geneticand epigenetic alterations responsible for a variety of tumorcell growth proliferation differentiation apoptosis andtherapy-resistance mechanisms The identification of theprognostic value of the PAX-FKHR fusion status in RMSis one of the most important shifts in the lineage and therisk assessment of RMS which exerts an oncogenic effectthrough multiple pathways and is incorporated into mostrelevant studies The misbalanced expression of a number ofmiRs involved in the regulation of myogenic differentiationalso implicates the tumorsrsquo escape suppressive mechanismsMoreover we described several in vivo models of RMSto explore underlying histogenesis and different aspects oftumorigenesis Recent work on CSCs has also contributedto understanding the cell origins refining molecular char-acterization and elucidating the underlying basis of refrac-tory responses to current adjuvant treatments of subsets oftumorsThese integrative approaches can provide opportuni-ties to identify diagnostic and prognostic biomarkers appliedto the individual targeted therapy and significantly improveRMS prognosis

Abbreviations

RMS RhabdomyosarcomaARMS Alveolar rhabdomyosarcomaERMS Embryonal rhabdomyosarcomaPRMS Pleomorphic rhabdomyosarcomaSRMS Sclerosingspindle cell

rhabdomyosarcomamiRNA miR MicroRNACSC Cancer stem cellMSC Mesenchymal stem cell

Conflict of Interests

The authors declare that there is no conflict of interestsregarding the publication of this paper

Acknowledgments

This work was supported in part by grants from the Gat-tegno and Wechsler funds Dr Duan was supported in partthrough a grant from Sarcoma Foundation of America (SFA)a grant from National Cancer Institute (NCI)National Insti-tutes of Health (NIH) UO1 CA 151452 and a grant from anAcademic Enrichment Fund of MGH Orthopedic Surgery

References

[1] L Yang T Takimoto and J Fujimoto ldquoPrognostic model forpredicting overall survival in children and adolescents withrhabdomyosarcomardquo BMC Cancer vol 14 no 1 article 6542014

[2] D Egas-Bejar and W W Huh ldquoRhabdomyosarcoma in adoles-cent and young adult patients current perspectivesrdquo AdolescentHealth Medicine andTherapeutics vol 5 pp 115ndash125 2014

[3] J C Breneman E Lyden A S Pappo et al ldquoPrognosticfactors and clinical outcomes in children and adolescents withmetastatic rhabdomyosarcomamdasha report from the intergrouprhabdomyosarcoma study IVrdquo Journal of Clinical Oncology vol21 no 1 pp 78ndash84 2003

[4] V D Soleimani and M A Rudnicki ldquoNew insights into theorigin and the genetic basis of rhabdomyosarcomasrdquo CancerCell vol 19 no 2 pp 157ndash159 2011

[5] T Glass D D Cochrane S R Rassekh K Goddard and JHukin ldquoGrowing teratoma syndrome in intracranial non-ger-minomatous germ cell tumors (iNGGCTs) a risk for secondarymalignant transformationmdasha report of two casesrdquo Childrsquos Ner-vous System vol 30 no 5 pp 953ndash957 2014

[6] J Ji C Eng and K Hemminki ldquoFamilial risk for soft tissuetumors a nation-wide epidemiological study from SwedenrdquoJournal of Cancer Research and Clinical Oncology vol 134 no5 pp 617ndash624 2008

[7] C D M Fletcher J A Bridge P Hogendoorn and F MertensWHO Classification of Tumours of Soft Tissue and Bone vol 5IARC Press Paris France 4th edition 2013

[8] I Sultan I Qaddoumi S Yaser C Rodriguez-Galindo and AFerrari ldquoComparing adult and pediatric rhabdomyosarcoma inthe surveillance epidemiology and end results program 1973 to2005 an analysis of 2600 patientsrdquo Journal of Clinical Oncologyvol 27 no 20 pp 3391ndash3397 2009

[9] B Raney W Huh D Hawkins et al ldquoOutcome of patientswith localized orbital sarcoma who relapsed following treat-ment on Intergroup Rhabdomyosarcoma Study Group (IRSG)Protocols-III and -IV 1984ndash1997 a report from the ChildrenrsquosOncology Grouprdquo Pediatric Blood and Cancer vol 60 no 3 pp371ndash376 2013

[10] W Crist E A Gehan A H Ragab et al ldquoThe third intergrouprhabdomyosarcoma studyrdquo Journal of Clinical Oncology vol 13no 3 pp 610ndash630 1995

[11] C Liu D Li J Jiang et al ldquoAnalysis of molecular cytogeneticalteration in rhabdomyosarcoma by array comparative genomichybridizationrdquo PLoS ONE vol 9 no 4 Article ID e94924 2014

[12] DWilliamson E Missiaglia A de Reynies et al ldquoFusion gene-negative alveolar rhabdomyosarcoma is clinically and molec-ularly indistinguishable from embryonal rhabdomyosarcomardquoJournal of Clinical Oncology vol 28 no 13 pp 2151ndash2158 2010

[13] V Paulson G Chandler D Rakheja et al ldquoHigh-resolutionarray CGH identifies common mechanisms that drive embry-onal rhabdomyosarcoma pathogenesisrdquo Genes Chromosomesand Cancer vol 50 no 6 pp 397ndash408 2011

[14] S Park J Lee I-G Do et al ldquoAberrant CDK4 amplificationin refractory rhabdomyosarcoma as identified by genomicprofilingrdquo Scientific Reports vol 4 article 3623 2014

[15] C Liu D Li J Hu et al ldquoChromosomal and genetic imbalancesin Chinese patients with rhabdomyosarcoma detected by high-resolution array comparative genomic hybridizationrdquo Interna-tional Journal of Clinical and Experimental Pathology vol 7 no2 pp 690ndash698 2014

[16] Q-X Li C-X Liu C-P Chun et al ldquoChromosomal imbalancesrevealed in primary rhabdomyo-sarcomas by comparativegenomic hybridizationrdquo Chinese Medical Journal vol 122 no11 pp 1277ndash1282 2009

[17] E Missiaglia J Selfe M Hamdi et al ldquoGenomic imbalancesin rhabdomyosarcoma cell lines affect expression of genesfrequently altered in primary tumors an approach to identifycandidate genes involved in tumor developmentrdquo Genes Chro-mosomes and Cancer vol 48 no 6 pp 455ndash467 2009

12 Sarcoma

[18] F G Barr F Duan L M Smith et al ldquoGenomic and clinicalanalyses of 2p24 and 12q13-q14 amplification in alveolar rhab-domyosarcoma a report from the childrenrsquos oncology grouprdquoGenes Chromosomes and Cancer vol 48 no 8 pp 661ndash6722009

[19] T Gordon AMcManus J Anderson et al ldquoCytogenetic abnor-malities in 42 rhabdomyosarcoma a United Kingdom CancerCytogenetics Group StudyrdquoMedical and Pediatric Oncology vol36 no 2 pp 259ndash267 2001

[20] X Chen E Stewart A A Shelat et al ldquoTargeting oxidativestress in embryonal rhabdomyosarcomardquo Cancer Cell vol 24no 6 pp 710ndash724 2013

[21] S Lizard-Nacol F Mugneret C Volk C Turc-Carel M Favrotand T Philip ldquoTranslocation (213)(q37q14) in alveolar rhab-domyosarcoma a new caserdquo Cancer Genetics and Cytogeneticsvol 25 no 2 pp 373ndash374 1987

[22] P H B Sorensen J C Lynch S J Qualman et al ldquoPAX3-FKHR and PAX7-FKHR gene fusions are prognostic indicatorsin alveolar rhabdomyosarcoma a report from the childrenrsquosoncology grouprdquo Journal of Clinical Oncology vol 20 no 11 pp2672ndash2679 2002

[23] A D Marshall and G C Grosveld ldquoAlveolar rhabdomyo-sarcomamdashthe molecular drivers of PAX37-FOXO1-inducedtumorigenesisrdquo Skeletal Muscle vol 2 no 1 article 25 2012

[24] E Missiaglia D Williamson J Chisholm et al ldquoPAX3FOXO1fusion gene status is the key prognostic molecular markerin rhabdomyosarcoma and significantly improves current riskstratificationrdquo Journal of Clinical Oncology vol 30 no 14 pp1670ndash1677 2012

[25] J F Shern L Chen J Chmielecki et al ldquoComprehensivegenomic analysis of rhabdomyosarcoma reveals a landscape ofalterations affecting a common genetic axis in fusion-positiveand fusion-negative tumorsrdquo Cancer Discovery vol 4 no 2 pp216ndash231 2014

[26] MWachtel M Dettling E Koscielniak et al ldquoGene expressionsignatures identify rhabdomyosarcoma subtypes and detect anovel t(22)(q35p23) translocation fusing PAX3 to NCOA1rdquoCancer Research vol 64 no 16 pp 5539ndash5545 2004

[27] E Davicioni M J Anderson F G Finckenstein et al ldquoMolec-ular classification of rhabdomyosarcomamdashgenotypic and phe-notypic determinants of diagnosis a report from the ChildrenrsquosOncology Grouprdquo The American Journal of Pathology vol 174no 2 pp 550ndash564 2009

[28] M Visser C Sijmons J Bras et al ldquoAllelotype of pediatric rhab-domyosarcomardquo Oncogene vol 15 no 11 pp 1309ndash1314 1997

[29] R Gil-Benso T San-Miguel R C Callaghan et al ldquoChromo-somal and genetic changes produced in tumoral progression ofembryonal rhabdomyosarcomardquo Histopathology vol 62 no 5pp 816ndash819 2013

[30] S X Skapek J Anderson F G Barr et al ldquoPAX-FOXO1 fusionstatus drives unfavorable outcome for children with rhabdom-yosarcoma a childrenrsquos oncology group reportrdquo Pediatric Bloodand Cancer vol 60 no 9 pp 1411ndash1417 2013

[31] HHosoi N Kakazu E Konishi et al ldquoA novel PAX3 rearrange-ment in embryonal rhabdomyosarcomardquo Cancer Genetics andCytogenetics vol 189 no 2 pp 98ndash104 2009

[32] E Takaoka H Sonobe K Akimaru et al ldquoMultiple sites ofhighly amplified DNA sequences detected bymolecular cytoge-netic analysis inHS-RMS-2 a newpleomorphic rhabdomyosar-coma cell linerdquo American Journal of Cancer Research vol 2 no2 pp 141ndash152 2012

[33] K Szuhai D de Jong W Y Leung C D M Fletcher and P CWHogendoorn ldquoTransactivatingmutation of theMYOD1 geneis a frequent event in adult spindle cell rhabdomyosarcomardquoJournal of Pathology vol 232 no 3 pp 300ndash307 2014

[34] N P Agaram C-L Chen L Zhang M P Laquaglia L Wexlerand C R Antonescu ldquoRecurrent MYOD1 mutations in pedi-atric and adult sclerosing and spindle cell rhabdomyosarcomasEvidence for a common pathogenesisrdquoGenes Chromosomes andCancer vol 53 no 9 pp 779ndash787 2014

[35] H Yoshida M Miyachi K Sakamoto et al ldquoPAX3-NCOA2fusion gene has a dual role in promoting the proliferation andinhibiting the myogenic differentiation of rhabdomyosarcomacellsrdquo Oncogene vol 33 pp 5601ndash5608 2013

[36] J M Mosquera A Sboner L Zhang et al ldquoRecurrent NCOA2gene rearrangements in congenitalinfantile spindle cell rhab-domyosarcomardquo Genes Chromosomes and Cancer vol 52 no6 pp 538ndash550 2013

[37] V Thavaraj A Sethi and L S Arya ldquoIncomplete Beckwith-Wiedemann syndrome in a child with orbital rhabdomyosar-comardquo Indian Pediatrics vol 39 no 3 pp 299ndash304 2002

[38] D Walter S Satheesha P Albrecht et al ldquoCD133 positiveembryonal rhabdomyosarcoma stem-like cell population isenriched in rhabdospheresrdquo PLoS ONE vol 6 no 5 Article IDe19506 2011

[39] Y-X Ren F G Finckenstein D A Abdueva et al ldquoMousemesenchymal stem cells expressing PAX-FKHR form alveolarrhabdomyosarcomas by cooperating with secondary muta-tionsrdquo Cancer Research vol 68 no 16 pp 6587ndash6597 2008

[40] M Salerno S Avnet G Bonuccelli S Hosogi D Granchi andN Baldini ldquoImpairment of lysosomal activity as a therapeuticmodality targeting cancer stem cells of embryonal rhabdom-yosarcoma cell line RDrdquo PLoS ONE vol 9 no 10 Article IDe110340 2014

[41] K Tanoue Y Wang M Ikeda et al ldquoSurvivin-responsive con-ditionally replicating adenovirus kills rhabdomyosarcoma stemcells more efficiently than their progenyrdquo Journal of Transla-tional Medicine vol 12 no 1 article 27 2014

[42] P K Rao EMissiaglia L Shields et al ldquoDistinct roles formiR-1and miR-133a in the proliferation and differentiation of rhab-domyosarcoma cellsrdquo The FASEB Journal vol 24 no 9 pp3427ndash3437 2010

[43] D Yan X D Dong X Chen et al ldquoMicroRNA-1206 targets c-met and inhibits rhabdomyosarcomadevelopmentrdquoThe Journalof Biological Chemistry vol 284 no 43 pp 29596ndash29604 2009

[44] I Marchesi A Giordano and L Bagella ldquoRoles of enhancerof zeste homolog 2 from skeletal muscle differentiation torhabdomyosarcoma carcinogenesisrdquoCell Cycle vol 13 no 4 pp516ndash527 2014

[45] M M Sun J F Li L L Guo et al ldquoTGF-1205731 suppression ofmicroRNA-450b-5p expression a novel mechanism for block-ingmyogenic differentiation of rhabdomyosarcomardquoOncogenevol 33 no 16 pp 2075ndash2086 2014

[46] Y Diao X Guo L Jiang et al ldquoMiR-203 a tumor suppressorfrequently down-regulated by promoter hypermethylation inrhabdomyosarcomardquo The Journal of Biological Chemistry vol289 no 1 pp 529ndash539 2014

[47] H-J Huang J Liu HHua et al ldquoMiR-214 andN-ras regulatoryloop suppresses rhabdomyosarcoma cell growth and xenografttumorigenesisrdquo Oncotarget vol 5 no 8 pp 2161ndash2175 2014

Sarcoma 13

[48] A L Sarver L Li and S Subramanian ldquomicroRNA miR-183functions as an oncogene by targeting the transcription factorEGR1 and promoting tumor cell migrationrdquo Cancer Researchvol 70 no 23 pp 9570ndash9580 2010

[49] R L Galindo J A Allport and E N Olson ldquoA Drosophilamodel of the rhabdomyosarcoma initiator PAX7-FKHRrdquo Pro-ceedings of the National Academy of Sciences of the United Statesof America vol 103 no 36 pp 13439ndash13444 2006

[50] U Avirneni-Vadlamudi K A Galindo T R Endicott V Paul-son S Cameron and R L Galindo ldquoDrosophila and mammal-ian models uncover a role for the myoblast fusion gene TANC1in rhabdomyosarcomardquo The Journal of Clinical Investigationvol 122 no 1 pp 403ndash407 2012

[51] E Y Chen andDM Langenau ldquoZebrafishmodels of rhabdom-yosarcomardquoMethods in Cell Biology vol 105 pp 383ndash402 2011

[52] X Le E K Pugach S Hettmer et al ldquoA novel chemicalscreening strategy in zebrafish identifies common pathways inembryogenesis and rhabdomyosarcoma developmentrdquo Devel-opment vol 140 no 11 pp 2354ndash2364 2012

[53] Y Lee R Kawagoe K Sasai et al ldquoLoss of suppressor-of-fusedfunction promotes tumorigenesisrdquoOncogene vol 26 no 44 pp6442ndash6447 2007

[54] C Keller B R Arenkiel C M Coffin N El-Bardeesy R ADePinho and M R Capecchi ldquoAlveolar rhabdomyosarcomasin conditional Pax3Fkhr mice cooperativity of Ink4aARF andTrp53 loss of functionrdquo Genes and Development vol 18 no 21pp 2614ndash2626 2004

[55] S Singh CVinson CMGurley et al ldquoImpairedWnt signalingin embryonal rhabdomyosarcoma cells from p53c-fos doublemutant micerdquo American Journal of Pathology vol 177 no 4 pp2055ndash2066 2010

[56] G J Schaaf J M Ruijter F Van Ruissen et al ldquoFull transcrip-tome analysis of rhabdomyosarcoma normal and fetal skeletalmuscle statistical comparison of multiple SAGE librariesrdquo TheFASEB Journal vol 19 no 3 pp 404ndash406 2005

[57] D S Hawkins A A Gupta and E R Rudzinski ldquoWhat is newin the biology and treatment of pediatric rhabdomyosarcomardquoCurrent Opinion in Pediatrics vol 26 no 1 pp 50ndash56 2014

[58] M Zhang J Truscott and J Davie ldquoLoss of MEF2D expressioninhibits differentiation and contributes to oncogenesis in rhab-domyosarcoma cellsrdquoMolecular Cancer vol 12 no 1 article 1502013

[59] M Ciesla J Dulak and A Jozkowicz ldquomicroRNAs and epi-genetic mechanisms of rhabdomyosarcoma developmentrdquo TheInternational Journal of Biochemistry amp Cell Biology vol 53 pp482ndash492 2014

[60] S Ricaud B Vernus M Duclos et al ldquoInhibition of autocrinesecretion of myostatin enhances terminal differentiation inhuman rhabdomyosarcoma cellsrdquo Oncogene vol 22 no 51 pp8221ndash8232 2003

[61] C S Guo C Degnin T A Fiddler D Stauffer andM JThayerldquoRegulation of MyoD activity and muscle cell differentiation byMDM2 pRb and Sp1rdquoThe Journal of Biological Chemistry vol278 no 25 pp 22615ndash22622 2003

[62] S Rossi E Stoppani P L Puri and A Fanzani ldquoDifferenti-ation of human rhabdomyosarcoma RD cells is regulated byreciprocal functional interactions between myostatin p38 andextracellular regulated kinase signalling pathwaysrdquo EuropeanJournal of Cancer vol 47 no 7 pp 1095ndash1105 2011

[63] Z S Walters B Villarejo-Balcells D Olmos et al ldquoJARID2is a direct target of the PAX3-FOXO1 fusion protein andinhibits myogenic differentiation of rhabdomyosarcoma cellsrdquoOncogene vol 33 no 9 pp 1148ndash1157 2014

[64] T Hosoyama M I Aslam J Abraham et al ldquoIL-4R drives ded-ifferentiation mitogenesis and metastasis in rhabdomyosar-comardquo Clinical Cancer Research vol 17 no 9 pp 2757ndash27662011

[65] F S Dela Cruz ldquoCancer stem cells in pediatric sarcomasrdquoFrontiers in Oncology vol 3 article 168 2013

[66] M J Bissell and W C Hines ldquoWhy donrsquot we get more cancerA proposed role of the microenvironment in restraining cancerprogressionrdquo Nature Medicine vol 17 no 3 pp 320ndash329 2011

[67] E Charytonowicz C Cordon-Cardo I Matushansky and MZiman ldquoAlveolar rhabdomyosarcoma is the cell of origin amesenchymal stem cellrdquoCancer Letters vol 279 no 2 pp 126ndash136 2009

[68] W K Cavenee ldquoMuscling in on rhabdomyosarcomardquo NatureMedicine vol 8 no 11 pp 1200ndash1201 2002

[69] B Zhu and J K Davie ldquoNew insights into signalling-pathwayalterations in rhabdomyosarcomardquo British Journal of Cancervol 112 no 2 pp 227ndash231 2015

[70] M Zhang C M Linardic and D G Kirsch ldquoRAS and ROS inrhabdomyosarcomardquo Cancer Cell vol 24 no 6 pp 689ndash6912013

[71] S C Marianna L Ianzano and G Nicoletti ldquoTumor sup-pressor genes promote rhabdomyosarcoma progression in p53heterozygousrdquo Oncotarget vol 5 no 1 pp 108ndash119 2013

[72] SWang L Guo L Dong et al ldquoTGF-beta1 signal pathwaymaycontribute to rhabdomyosarcoma development by inhibitingdifferentiationrdquo Cancer Science vol 101 no 5 pp 1108ndash11162010

[73] J Wesche K Haglund and E M Haugsten ldquoFibroblast growthfactors and their receptors in cancerrdquo Biochemical Journal vol437 no 2 pp 199ndash213 2011

[74] M Wachtel J Rakic M Okoniewski P Bode F Niggli andB W Schafer ldquoFGFR4 signaling couples to Bim and not Bmfto discriminate subsets of alveolar rhabdomyosarcoma cellsrdquoInternational Journal of Cancer vol 135 no 7 pp 1543ndash15522014

[75] P L Puri ZWu P Zhang et al ldquoInduction of terminal differen-tiation by constitutive activation of p38 MAP kinase in humanrhabdomyosarcoma cellsrdquo Genes and Development vol 14 no5 pp 574ndash584 2000

[76] A M Tremblay E Missiaglia G G Galli et al ldquoThe Hippotransducer YAP1 transforms activated satellite cells and is apotent effector of embryonal rhabdomyosarcoma formationrdquoCancer Cell vol 26 no 2 pp 273ndash287 2014

[77] R N Judson A M Tremblay P Knopp et al ldquoThe hippo path-way member Yap plays a key role in influencing fate decisionsin muscle satellite cellsrdquo Journal of Cell Science vol 125 no 24pp 6009ndash6019 2012

[78] L E S Crose K A Galindo J G Kephart et al ldquoAlve-olar rhabdomyosarcoma-associated PAX3-FOXO1 promotestumorigenesis via Hippo pathway suppressionrdquo Journal ofClinical Investigation vol 124 no 1 pp 285ndash296 2014

[79] B Belyea J G Kephart J Blum D G Kirsch and C MLinardic ldquoEmbryonic signaling pathways and rhabdomyosar-coma contributions to cancer development and opportunitiesfor therapeutic targetingrdquo Sarcoma vol 2012 Article ID 40623913 pages 2012

14 Sarcoma

[80] J Huertas-Martinez S Rello-Varona D Herrero-Martin et alldquoCaveolin-1 is down-regulated in alveolar rhabdomyosarcomasand negatively regulates tumor growthrdquo Oncotarget vol 5 no20 pp 9744ndash9755 2014

[81] S Rossi P L Poliani CMissale EMonti andA Fanzani ldquoCav-eolins in rhabdomyosarcomardquo Journal of Cellular andMolecularMedicine vol 15 no 12 pp 2553ndash2568 2011

[82] P G Tsioli E S Patsouris C Giaginis and S E TheocharisldquoDNA repair systems in rhabdomyosarcomardquo Histology andHistopathology vol 28 no 8 pp 971ndash984 2013

[83] E Gatzidou C Michailidi S Tseleni-Balafouta and S Theo-charis ldquoAn epitome of DNA repair related genes and mecha-nisms in thyroid carcinomardquo Cancer Letters vol 290 no 2 pp139ndash147 2010

[84] R Rota R Ciarapica A Giordano L Miele and F LocatellildquoMicroRNAs in rhabdomyosarcoma pathogenetic implicationsand translational potentialityrdquoMolecular Cancer vol 10 article120 2011

[85] A Gougelet J Perez D Pissaloux et al ldquoMiRNA profiling howto bypass the current difficulties in the diagnosis and treatmentof sarcomasrdquo Sarcoma vol 2011 Article ID 460650 13 pages2011

[86] R Ciarapica G Russo F Verginelli et al ldquoDeregulated expres-sion of miR-26a and Ezh2 in rhabdomyosarcomardquo Cell Cyclevol 8 no 1 pp 172ndash175 2009

[87] L Li A L Sarver S Alamgir and S Subramanian ldquoDownreg-ulation of microRNAs miR-1 -206 and -29 stabilizes PAX3 andCCND2 expression in rhabdomyosarcomardquo Laboratory Investi-gation vol 92 no 4 pp 571ndash583 2012

[88] S-Y Ng G K Bogu B Soh and L W Stanton ldquoThe longnoncoding RNA RMST interacts with SOX2 to regulate neu-rogenesisrdquoMolecular Cell vol 51 no 3 pp 349ndash359 2013

[89] C A Lynch B Tycko T H Bestor and C P Walsh ldquoReac-tivation of a silenced H19 gene in human rhabdomyosarcomaby demethylation of DNA but not by histone hyperacetylationrdquoMolecular Cancer vol 1 article 2 2002

[90] P Onyango and A P Feinberg ldquoA nucleolar protein H19opposite tumor suppressor (HOTS) is a tumor growth inhibitorencoded by a human imprinted H19 antisense transcriptrdquoProceedings of the National Academy of Sciences of the UnitedStates of America vol 108 no 40 pp 16759ndash16764 2011

[91] C E Albacker N Y Storer E M Langdon et al ldquoThe his-tone methyltransferase SUV39H1 suppresses embryonal rhab-domyosarcoma formation in zebrafishrdquo PLoS ONE vol 8 no 5Article ID e64969 2013

[92] N Y Storer R M White A Uong et al ldquoZebrafish rhab-domyosarcoma reflects the developmental stage of oncogeneexpression during myogenesisrdquo Development vol 140 no 14pp 3040ndash3050 2013

[93] M E Hatley W Tang M R Garcia et al ldquoA mouse modelof rhabdomyosarcoma originating from the adipocyte lineagerdquoCancer Cell vol 22 no 4 pp 536ndash546 2012

[94] V Hosur A Kavirayani J Riefler et al ldquoDystrophin and dysfer-lin doublemutantmice a novelmodel for rhabdomyosarcomardquoCancer Genetics vol 205 no 5 pp 232ndash241 2012

[95] W Goessling T E North and L I Zon ldquoNew waves of discov-ery modeling cancer in zebrafishrdquo Journal of Clinical Oncologyvol 25 no 17 pp 2473ndash2479 2007

[96] B P Rubin K Nishijo H-I H Chen et al ldquoEvidence for anunanticipated relationship between undifferentiated pleomor-phic sarcoma and embryonal rhabdomyosarcomardquo Cancer Cellvol 19 no 2 pp 177ndash191 2011

[97] S Z Kaylani J Xu R K Srivastava L Kopelovich J G Presseyand M Athar ldquoRapamycin targeting mTOR and hedgehog sig-naling pathways blocks human rhabdomyosarcoma growth inxenograft murine modelrdquo Biochemical and Biophysical ResearchCommunications vol 435 no 4 pp 557ndash561 2013

[98] S Q Li A T Cheuk J F Shern et al ldquoTargeting wild-type andmutationally activated FGFR4 in rhabdomyosarcoma with theinhibitor ponatinib (AP24534)rdquoPLoSONE vol 8 no 10 ArticleID e76551 2013

[99] K Miyoshi K Kohashi F Fushimi et al ldquoClose correlationbetween CXCR4 and VEGF expression and frequent CXCR7expression in rhabdomyosarcomardquo Human Pathology vol 45no 9 pp 1900ndash1909 2014

[100] ADMarshall F Picchione R I KGeltink andGCGrosveldldquoPAX3-FOXO1 induces up-regulation of Noxa sensitizing alve-olar rhabdomyosarcoma cells to apoptosisrdquo Neoplasia vol 15no 7 pp 738ndash748 2013

[101] F Perut S Avnet C Fotia et al ldquoV-ATPase as an effective thera-peutic target for sarcomasrdquo Experimental Cell Research vol 320no 1 pp 21ndash32 2014

Submit your manuscripts athttpwwwhindawicom

Stem CellsInternational

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

MEDIATORSINFLAMMATION

of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Behavioural Neurology

EndocrinologyInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Disease Markers

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

BioMed Research International

OncologyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Oxidative Medicine and Cellular Longevity

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

PPAR Research

The Scientific World JournalHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Immunology ResearchHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal of

ObesityJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Computational and Mathematical Methods in Medicine

OphthalmologyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Diabetes ResearchJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Research and TreatmentAIDS

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Gastroenterology Research and Practice

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Parkinsonrsquos Disease

Evidence-Based Complementary and Alternative Medicine

Volume 2014Hindawi Publishing Corporationhttpwwwhindawicom

Page 3: Review Article Rhabdomyosarcoma: Advances in Molecular and

Sarcoma 3

PAX3

Chromosome 2

PAX3-FKHR

t(213)(q37q14)

NCOA1

PAX7

Chromosome 1

PAX7-FKHR

t(113)(p36q14)

PAX3

Chromosome 2

FKHR

Chromosome 13

PAX3-NCOA1

t(22)(q35q23)

Paired domain Homeodomain

DNA binding domain Transactivation domain

PB

PB

PB

PB HD

HD

HD

HD TAD

TAD

TAD

TAD

TAD

TAD

HD

HD

PB

PB

100 amino acids

Fusion site

Fusion site

FD

DNA binding heterodimerization domain Nuclear receptor interaction domain

LXXLL HAT

HAT

bHLHPAS

Fusion site

(a)

(b)

(c)

Figure 1The chromosomal rearrangements in ARMS 80 of ARMS classified as translocation-positive ARMS carry characteristic chromo-somal translocations demonstrated as t(213)(q35q14) t(113)(p36q14) and (22)(q35p23) In (a) and (b) the translocations fuse the FKHR(a member of the forkheadHNF-3 transcription factor family) locus on chromosome 13 to either PAX3 on chromosome 2 or PAX7 onchromosome 1 In (c) the translocation generated a fusion protein composed of PAX3 and the nuclear receptor coactivator NCOA1 havingsimilar transactivation properties as PAX3FKHR

These studies have identified a number of genetic alterationsin RMSMany of these chromosomal changesmay be respon-sible for tumor progression and proliferation These geneticalterations may be potential treatment targets in RMS [11]

22 Chromosomal Translocations in ARMS Chromosomalanalyses have demonstrated two translocations associatedwith ARMS t(213)(q35q14) and t(113)(p36q14) [21] Initialstudies detected these two gene fusions in 80 of ARMS[22] These characteristic chromosomal translocations areadjacent to the 51015840 DNA-binding domains of PAX a memberof the paired box transcription factor family and the trans-activation domain at the 31015840 end of FKHR a member of theforkheadHNF-3 transcription factor family Approximately75 of these structural rearrangements translocate the PAX3gene at 2q35 to the FKHR gene at 13q14 as t(213)(q35q14)less frequently in the other 25 the t(13)(q36q14) transloca-tion fuses PAX7 to FKHR [23] Previous studies have demon-strated that PAX3FKHR fusion gene status significantlyimproves current risk stratification while the presence ofPAX7FKHR and rarer variant fusion gene products requirefurther investigation [24] The remaining 20 of ARMS isPAX gene fusion-negative (PFN) and forms a more hetero-geneous group which remains a challenge to detect due to

the lack of consistent chromosomal rearrangements PFNARMS has a similar clinical course to ERMS which suggeststhat the fusion gene status provides more accurate informa-tion about patient outcomes than did the histologic subtypeDespite the low overall burden of somatic mutations infusion-positive RMSmultiple genes were recurrently alteredincluding NRAS KRAS HRAS FGFR4 PIK3CA CTNNB1FBXW7 and BCOR [25]

Another novel translocation t(22)(q35p23) was identi-fied in ARMS biopsy samples by gene expression signatures[26] The chromosomal translocation generates a fusion pro-tein composed of PAX3 and the nuclear receptor coacti-vator NCOA1 which has similar transactivation propertiesas PAX3FKHR [26] These biologic effects contribute totumorigenesis bymodulatingmyogenic differentiation alter-ing growth and apoptotic pathways and stimulating motilityand other metastatic pathways (Figure 1)

23 Chromosomal Alterations in ERMS ERMS exhibits a lossof imprinting (LOI) leading to a twofold gene dosage effect[27] Most tumors have at least one 15-Mb region with loss ofheterozygosity (LOH) along chromosome 11 [27] The allelo-type of ERMS demonstrates a high frequency of LOH onchromosomes 11p 11q and 16q [28] ERMS tumorigenesis

4 Sarcoma

Table 2 The histological types of rhabdomyosarcoma

Histological type Predilection population Predilection site Risk category Genetic change

ERMS Infants or children under 10years old Head and neck region Intermediate LOH at chromosome 11p155

ARMS Adolescents and young adults Deep tissue of extremity High t(213)(q35q14) t(113)(p36q14)

PRMS Adult males Throughout the body HighJUN (1p31) MYC (8q24)CCND1 (11q13) INT2 (11q133)MDM2 (12q143ndashq15)

SRMSIn the first decade of life with asecond mode centered aroundthe fifth decade

The extremities headand neck Superior SRF-NCOA2 MYOD1 mutation

ERMS embryonal rhabdomyosarcomaARMS alveolar rhabdomyosarcomaPRMS pleomorphic rhabdomyosarcomaSRMS spindle cellsclerosing rhabdomyosarcoma

can result from the inactivation of the parental bias of chro-mosome 11p15 which is the most common rearrangementin ERMS [29] The proportion of ERMS with LOH alongchromosome 11 is considerably higher than in other subtypes[27] In histopathological analysis ERMS expresses lowPAX3levels and elevated PAX7 levels [30] Hosoi et al identified ahidden 2q35 breakpoint as a novel PAX3 rearrangement incomplex chromosomal translocations in ERMS [31]

24 Chromosomal Alterations in Other RMS There are lim-ited studies on the biological pathways involved in othersubtypes of RMS compared with the two major subtypesARMS andERMS Fluorescence In SituHybridization (FISH)reveals amplification of JUN (1p31) MYC (8q24) CCND1(11q13) INT2 (11q133) MDM2 (12q143ndashq15) and MALT(18q21) in these tumor cells contributing to the pathogenesisof PRMS [32] MYOD1 homozygous mutations are alsoreported as frequent recurrent and pathognomonic eventsin adult-type SRMS [33 34] (Table 2) Furthermore four outof five pediatric tumors showedMYOD1mutations in a study[34] Yoshida et al described a 8q13 locus (NCOA2) generearrangement in a small subset of SRMS occurring uniquelyin the infantilecongenital setting fused with key transcrip-tion factors involved in skeletal muscle differentiation suchas SRF and TEAD1 [35] They also identified NCOA2 as acandidate PAX3 partner geneThePAX3-NCOA2 fusion geneplays a dual role in the tumorigenesis of RMS promotion ofthe proliferation and inhibition of the myogenic differenti-ation of RMS cells [35] As PAX3-NCOA2-induced tumorsgrow more slowly SRMS that are PAX3-NCOA2 fusion-positive could be associated with a very favorable prognosis[36]

Not all RMSs occur as sporadic primary tumors Occa-sionally RMS inherits a mutant gene as part of an establishedfamilial syndrome For example Beckwith-Wiedemann Syn-drome (BWS) with RMS involves dysregulation or alterationof imprinted genes in the 11p155 chromosomal regionincluding IGF2 H19 and CDKN1C (p57KIP2) [37]

3 Cell of Origin in RMS

There have been a number of studies aimed at deciphering thecell origin for RMS [56ndash61]

31 Myogenic Differentiation in the Tumorigenesis Severalstudies have focused on elucidating the mechanisms govern-ing the impaired myogenic program in RMS [62 63] (Fig-ure 2) RMSoriginates as a consequence of regulatory disrup-tion of the growth and differentiation of myogenic precursorcells Progenitor cells reside in muscle and their activationresults in either proper myogenesis or aberrant signalingpathways leading to the development of RMS Based on theskeletal muscle lineage a complete transcriptome analysis ofRMS was performed to compare normal and fetal muscles[56] The high degree of similarity between fetal muscle andRMS expression profiles reflects the undifferentiated myo-genic nature of RMS The genes exclusively upregulated inRMS including FGFR4 NOTCH2 UBE2C UHRF1 andYWHAB genes contribute to the failure of RMS cells to com-plete normal skeletal muscle development and progress toan alternative fate [56] RMS cells represent an arrested statein the development of normal skeletal muscle with regionalsilencing of differentiation factors leading to the maturationdefect in RMS [57] The expression pattern of muscle-specific proteins regulating myogenic differentiation hasbeen extensively examined in RMS The family of myogenictranscription factors (MYOD MYF5 myogenin and MRF4)are considered to be responsible for the determination of stemcells intomyoblasts anddifferentiation intomyocytes [58 59]All these factors are activated during the onset or progressionof myogenesis and are subsequently silenced to reach a finalmuscular differentiation There are statistical differences inthe different subtypes of RMS tumors with MYOD or myo-genin staining patterns Amplification of MDM2 in an RMScell line interferes with MYOD activity and consequentlyinhibits overt muscle cell differentiation [61] Interleukin-4receptor (IL-4R) has also been proposed to be important for

Sarcoma 5

Table 3 Studies identifying stem cells in RMS

Markersubstrate Source Stem cell gene Functional characterization Reference

CD133 Orthotopicxenograft model

OCT4 NANOG c-MYCPAX3 and SOX2

Correlating with poor overallsurvival

Walter et al2011 [38]

PAX-FKHR Bone marrow ofC57BL6 mice

Myf5 MEF2 MyoD andmyogenin

Determining the molecularmyogenic and histologicphenotype of ARMS

Ren et al 2008[39]

V-ATPase RD cell line NANOG and OCT34

Driving mechanisms of areduced sensitivity to anticancerdrugs and activities related toinvasion and metastasis

Salerno et al2014 [40]

Survm-CRAsKYM-1 cell line(FGFR3-positive)

FGFR3Therapeutic effectiveness againstall cell populations and increasedeffectiveness against CSCs

Tanoue et al2014 [41]

Migration

specification

Injured muscle

Terminal differentiation

Fusion

PAX7

Myoblasts

Myoblast

MyoD

MyoD

Myf5

PAX3

Rb

Ezh2

IGF2

HDAC4YY1

YY1

ERK

AKT

RMS

Myogenin

P38 kinase

Myostatin

Progenitor

cell

N-RAS

Myofibers

FGFR4

UBE2C

UHRF1

NOTCH2

YWHAB

Satellite cells

RUNX1 MEF2C

JDP2 NFIC

Myocytes

Restoration of proliferation blockade of

differentiation

Proliferatio

n

Cell circle arrestEarlydifferentiation

TGF-1205731

Figure 2 Myogenic pathways in the tumorigenesis of RMS In aberrant neoplastic condition progenitor cells residing in muscle result inaberrant pathways which lead to malignant transformation and fail to differentiate proliferate uncontrollably and form RMS Sharp arrows(rarr) indicate upregulationactivation and blunt arrows (perp) indicate downregulationinhibition

the maturation of myotubes [64] An IL-4R blockade mighthelp therapeutically to modulate the expression of myogenictranscription factors (MYOD or myogenin) [57 64]

32 Potential Cancer Stem Cell in RMS Cancer Stem Cells(CSCs) may contribute to inherent refractory responses tocurrent therapies andmetastasis [65]Many CSCmodels playan important role in the development of RMS [38ndash41 66](Table 3)

A study explored a potential important role for mesen-chymal stem cells (MSCs) as the cell of origin of ARMS [67]Ren et al confirmed that PAX-FKHR fusion genes commitMSCs to a myogenic lineage by inhibiting terminal differen-tiation and contributing to ARMS formation [39] Anotherstudy showed that PAX-FKHR induces skeletal myogenesisin MSCs by transactivating MYOD and myogenin and trans-forms mesenchymal progenitor cells to the skeletal muscle lin-eage leading to malignant formation resembling ARMS [39]

6 Sarcoma

METCDKN2AB

PAX37-FKHR

MDM2

RAS

Raf

GLI1

PDK1p53Rb

Hh

Myo MEF2

p21

Ptch

FGFR4

IGF1R

p16INK4Ap14ARF

p19ARF

IGF2

FGF

Figure 3 Genetic analyses of RMS have pinpointed several common alterations including inactivation of a master regulator of p53 andRb pathways CDKN2AB and activation of FGFR4 RAS and Hedgehog (Hh) signaling The modifications of these pathways influenceoncogenesis and metastatic potential

As a human hematopoietic CSC marker Walter et alproposed that upregulated CD133 in ERMS can act as aprognostic marker and might help with the development ofnovel targeted therapies for ERMS [38]

4 Signaling Pathway Alterations in RMS

Recent studies have looked into signaling pathway alterationsand their downstream effects in RMSThese new findings notonly helped to improve the understanding of this malignancybut also offered novel potential therapeutic strategies forimproved treatment for patientswithRMS [68 69] (Figure 3)

41 RAS Signaling Pathway RASmutations commonlymain-tain the protein in its GTP bound state and therefore renderit constitutively active in RMS Zhang et al identified RASfamily members NRAS KRAS and HRAS in the RASpathway as themost commonlymutated genes in ERMS [70]the mutation status of these genes is significantly associatedwith the risk of ERMS development [70] A study foundthat a majority of ERMS demonstrated activation of the RASpathway exclusively either by homozygous deletion of NF1or by point mutations in one of the RAS family members asdescribed above [13] Despite remarkable genetic and molec-ular heterogeneity most (93 4144) RMS tumors hijackeda common receptor tyrosine kinaseRASPIK3CA geneticaxis [25] It could occur via two alternative mechanismsrearrangement of a PAX gene and accumulation of mutationsthat were downstream targets of the PAX fusion protein

Skeletal muscle cells have a robust antioxidant defensesystem to protect the DNA lipids and proteins from thedeleterious effects of excess Reactive Oxygen Species (ROS)

Cancer cells also have elevated ROS due to their increasedmetabolic activity oncogenic stimulation andmitochondrialdysfunction These findings implicate RAS mutations andoxidative stress as potential therapeutic targets for high-riskERMS

42 IGF Signaling Pathway There is clear preclinical datathat supports the involvement of the Insulin-like GrowthFactor (IGF) signaling pathway in RMS tumorigenesis andprogression [69] Zhu and Davie demonstrated that IGFpromotes the proliferation of RMS cells while blocking IGFsignaling interferes with cell growth in vivoThe IGF receptorIGF-1R is highly overexpressed both on the cell surface andin the nucleus of RMS cells furthermore RMS cell lines aresensitive to IGF-1R inhibitionThe IGF-2 locus shows a loss ofimprinting in both ERMS andARMS tumors and the expres-sion of PAX3-FKHR has been demonstrated to upregulateIGF-2 and activate IGF pathway in ARMS [69] Mariannaet al found that IGF-2 and tumor suppressors p19Arf andp21Cip1 are overexpressed and prominently upregulated inRMS mouse models [71]

43 TGF-120573 Signaling Pathway The regulatory role of Trans-forming Growth Factor-120573 (TGF-120573) in RMS has been recentlystudied [72] Wang et al demonstrated that the expression ofTGF-1205731 is significantly higher in RMS than in normal skeletalmuscle The inhibition of TGF-1205731 expression by shRNA-expressing vectors reverses themalignant behavior of RMSbyinhibiting cell growth and inducingmyogenic differentiationTGF-1205731 shRNA induces myogenin expression a regulator ofmyogenic differentiation genes Myogenin acts as a target fornegative regulation of myogenesis by TGF-1205731 signals with

Sarcoma 7

a differentiation regulatory cascade These results suggestthat the TGF-1205731 signaling pathway disrupts the differentia-tion of myogenic progenitors leading to the development ofRMS

44 FGF Signaling Pathway Fibroblast Growth Factor (FGF)is crucial in embryonic development and functions to driveproliferation FGFs and their receptors (FGFRs) are essentialregulators in the processes of proliferation antiapoptosisdrug resistance and angiogenesis in RMS [69] RMS over-expresses the receptor tyrosine kinase FGFR4 which causesautophosphorylation and constitutive signaling in correlationwith poor differentiation and decreased survival [73] Recentwork has shown that FGF signaling can prevent ARMS cellsfrom apoptosis induced by targeting the IGF1-R-PI3K-mTORpathway [74]

45 ERK Signaling Pathway Previous studies have shownthat elevated myostatin expression deregulates ExtracellularRegulated Kinase (ERK) signaling and deficient activationof the p38 pathway contributes to the differentiation blockin RMS cells [62] The ERK pathway is frequently highlyactivated in RMS cells from embryonic derivation due to thepresence of activating RASmutations leading to reduced dif-ferentiation by blocking the p38 pathway [75]These findingsindicate that interventions that target themyostatinERKp38network could be an effective way to promote differentiationof RMS

46 Hippo Signaling Pathway Many upstream signal trans-duction proteins in response to cues from the cellular micro-environment regulate the core Hippo pathway [76] TheHippo pathway may limit tumorigenesis by inducing thecytosolic localization of the transcriptional cofactor Yes-Associated Protein 1 (YAP1) High YAP levels and activitiesincrease activated satellite cells and prevent differentiationby activating genes that inhibit differentiation [77] YAP1-TEAD1 was found to upregulate proproliferative and onco-genic genes and maintain the ERMS differentiation blockby interfering with MYOD1 and MEF2 prodifferentiationactivities [76] Inhibition of theHippo pathway is exhibited inRMS through downregulation of Hippo pathway tumor sup-pressors or upregulation of YAP [78] These data suggest thatHippo pathway dysfunction promotes RMS development

47 Notch Signaling Pathway and Wnt Signaling PathwayNotch pathway as an embryonic signaling pathway promotesmuscle stem cell maintenance by inhibiting myogenic earlydifferentiation and expanding pools of progenitor cells dur-ing both embryogenesis and postnatal muscle regenerationSimilar to Notch Wnt has various roles in embryonic fetaland neonatalmyogenesis during skeletalmyogenesis As bothpathways are regulators of myogenic lineage determinationand maturation RMS can arise from disordered regulationof these normal developmental pathways [79]

48 Caveolins in RMS Recent findings have shown that cav-eolins are expressed in a cell stage-dependent manner inRMS [80 81] Caveolins are scaffolding proteins that regulate

several targets and pathways (such as p53 IGF RASERK andTGF-120573myostatin signaling) associated with RMS develop-ment and therefore loss or gain of caveolin function is ableto generate multiple effects on the tumor behaviorThereforerecognizing their precise roles in RMS progression will becrucial to elaborate targeted therapies particularly as oneof the three different isoforms CAV1 could act as a potenttumor suppressor in ARMS Inhibition of CAV1 function cancontribute to aberrant cell proliferation leading to ARMSdevelopment [80]

49 DNA Repairing System Some studies indicate that RMSis characterized by germline mutations of DNA repair genes[82 83] DNA repair gene alterations in RMS occur sec-ondarily to malignant transformation The modificationsin DNA repair enzyme activity can result in resistance toadjuvant treatment in RMS tumor cells which limits theprognosis of RMS There are two pathways for repairingDNA breaks directly without affecting DNA structure andindirectly by DNA phosphodiester backbone cleavage [82](Figure 4) Direct repair includes repair during replicationand enzymatic repair Indirect repair is comprised of exci-sion repair systems including base excision repair (BER)nucleotide excision repair (NER) mismatch repair (MMR)and recombination repair (RR) [83] Defective DNA repairmechanisms result from point mutations or LOH in RMSand contribute to tumorigenesis MGMT and MMR proteinactivity and expression levels are used as predictor indices fortherapy outcome in RMS BER as the major DNA repair sys-tem against damage resulting from cellular metabolism canreverse the cytotoxic effects of alkylation agents used suchas antineoplastics subsequent tumor progression and deathimplicates an RMS mechanism of resistance to chemother-apeutic agents This may provide novel individualized ther-apeutics targeting downregulation of activated DNA repairenzymes or upregulation of DNA repair deficiencies [82]

5 MicroRNA (miRNA miR)Expression in RMS

Muscle-specific and ubiquitously expressed miRs appeardownregulated in RMS tumors and cell lines compared withthe normal counterparts These miRs often play crucial rolesas antioncogenes Inhibition of these miRs contributes toenhanced tumorigenesis through the modulation of diversemolecular pathways Upregulation of prooncogenic miRs hasbeen detected in RMS recently as well [84] (Table 4)

51 Antioncogenic miR in RMS Gain-of-function experimentshave demonstrated that reexpression of selected ldquotumor sup-pressorrdquo miRs impairs the tumorigenic behavior of RMS cells[84 85] As myo-miR family members miR-1 miR-133a andmiR-206 have been demonstrated to be downregulated inRMS and shown to have activity on mRNA expression bytargeting c-Met [42 43 86] Inhibition of thesemyo-miRNAsmay cause aberrant cell proliferation and migration in myo-genesis leading to RMS development especially for ERMSThere have been more miRs found as tumor suppressors inthis malignancy including miR-29 miR-450b-5p miR-203

8 Sarcoma

Table 4 MicroRNAs involved in myogenesis and RMS development

miRNA Expression Target Function Reference

miR-1 and miR-133a Downregulation MYH9Myogenic miRNA inhibit differentiationand promote proliferation in myogenesiscytostatic

Rao et al 2010 [42]

miR-206 Downregulation cMET Promote differentiation and proliferationin myogenesis

Yan et al 2009 [43]

miR-29 Downregulation HADC4 YY1 EZH2 Promote stabilization of RMS phenotype Marchesi et al 2014 [44]

miR-450-5p Downregulation ENOX PAX9 Promote differentiation and progression Sun et al 2014 [45]

miR-203 Downregulation JAK STAT Notch Inhibit differentiation and proliferation inmyogenesis tumor suppressor

Diao et al 2014 [46]

miR-214 Downregulation N-Ras Inhibit tumor cell growth and inducemyogenic differentiation and apoptosis

Huang et al 2014 [47]

miR-183 Upregulation EGR1 Promote migration and metastasis Sarver et al 2010 [48]

PMS2

MGMT

HR

Mismatch Recombination

NHEJ MMEJ

p53

Excision

BER NER

RMS

DNA repair

Direct repair Indirect repair

repair repairrepair

Figure 4The DNA repair systems in RMSThere are two pathways repairing the DNA lesions directly without affecting DNA structure andindirectly by DNA phosphodiester backbone cleavage The modifications in DNA repair enzymes expression or activity lead to resistanceto chemotherapy and radiation in RMS tumor cells MGMT O6-methylguanine-DNA methyltransferase BER base excision repair NERnucleotide excision repair HR homologous recombination NHEJ nonhomologous end-joining MMEJ microhomology-mediated end-joining

and miR-214 [44 46 47 84 87] They are all significantlydownregulated in RMS compared with normal skeletal mus-cles and function to inhibit tumor cell growth and inducemyogenic differentiation and apoptosis in RMS

52 Oncogenic miRNA in RMS The transcription factor EGR1is a tumor suppressor gene that is downregulated in RMS [48]miR-183 functions as an oncogene by targeting EGR1 and pro-moting tumor cell migration Either by direct anti-miR treat-ment or by indirect mechanisms that decrease transcriptmiR-183 targeted treatmentsmight provide a potential optionto RMS patients

The basic strategy of current effective miR-based treat-ment studies is either efficient reexpression of miRs or restor-ing the function of miRs to inhibit the expression of certain

protein-coding genes and promote muscle differentiationMoreover miR expression profiling in tumors and possiblytheir detection in peripheral blood during treatment may beable to predict the response to chemotherapy or radiotherapyand be useful as a prognostic signature for the development oftreatment resistanceThe prognostic value of miR expressionlevels in RMS is a powerful tool in creating a better-tailoredstrategy for particular subsets of patients

53 Long Noncoding RNAs in RMS Long noncoding RNAs(LNCRNAs) are abundant in the mammalian transcriptomeand have been shown to play a role in RMS development [8889]TheH19 gene localized within a chromosomal region onhuman chromosome 11p15 encodes an imprinted untrans-lated RNA [90] H19 opposite tumor suppressor (HOTS)

Sarcoma 9

Table 5 In vivo animal models of RMS

Model TargetModel origin Reference

Inactivation Expression

DrosophilaPAX-FKHR Bipartite Gal4-UAS

expression systemGalindo et al2006 [49]

PAX7-FOXO1 rols Chromosomal deletionDf(3L)vin5

Avirneni-Vadlamudi et al2012 [50]

Zebrafishrag2 promoter c-Myc kRASG12D

Chen andLangenau 2011[51]

MAPKERK andAKTS6K1 PD98059 TPCK Tg(hsp70-HRASG12V) Le et al 2012

[52]

Mouse

Sufu N-myc Sfrp1 Ptch2and cyclin D1 Sufu+minus Lee et al 2007

[53]

Ink4aArf and Trp53 Pax3FKHR Pax3P3Fpwt Keller et al2004 [54]

Wnt120573-cateninsignaling pathway Wnt2 p53minusminusc-fosminusminus Singh et al 2010

[55]

a tumor growth inhibitor is encoded by an imprinted H19antisense transcript Overexpression of HOTS inhibits RMStumor cell growth Silencing HOTS by RNAi was shown toincrease in vitro colony formation as well as in vivo tumorgrowth of RMS [90]

6 In Vivo Models of RMS

Several in vivo models have recently been developed inDrosophila zebrafish and mice to further understand themolecular and cellular biology of RMS tumorigenesis [49ndash55 91ndash94] (Table 5)

61 Drosophila Models in RMS Despite the evolutionarydistance a strong conservation of genes pathways andregulatory molecular networks have been demonstratedbetween flies and humans Many human disease genes haverelated sequences in Drosophila In contrast to mammalianmodels the generation of Drosophila mutants is easy cheapand fast

For a better understanding of the pathogenic conse-quences of PAX-FKHR expression Drosophila was chosenbecause the animalrsquos transparent outer cuticle allows forsimple real-time monitoring of muscle abnormalities elicitedby PAX-FKHR including subtle or focal changes [49] Themutation in the myoblast fusion gene rolling pebbles (rols)was reported to dominantly suppress PAX-FKHR1-inducedlethality by using a Drosophila model of PAX-FKHR1-mediated transformation [50] PAX-FKHR1 signaling upreg-ulates the TANC1 gene and blocks myoblasts from terminaldifferentiation However downregulating the TANC1 genecould cause RMS cells to lose their neoplastic state undergofusion and form differentiation This novel finding uncov-ered a PAX-FKHR1-TANC1 neoplasia axis collaboratively ina Drosophila model and loss-gain-of-function studies inmammalian platforms [50]

62 Zebrafish Models in RMS In contrast to other mod-els the short tumor onset in zebrafish allows for rapididentification of essential genes in various processes suchas tumor growth self-renewal and maintenance There areother advantages for using zebrafish in the study of humandisease and development including the ease and low cost ofraising large numbers of fish and the highly conserved geneticand biochemical pathways between zebrafish and mammals[95]

As zebrafish RMS is highly similar to human ERMSby using fluorescent transgenic approaches it is more con-venient to understand histogenesis and different aspects oftumorigenesis in RMS A robust zebrafish model of RAS-induced RMS has been established sharing twomorphologicand immunophenotypic features resembling ERMS [51] Inone case both are associated with tissue-restricted geneexpression in RMS while in another both comprise a RAS-induced gene signature Cross-species microarray compar-isons confirm that conserved genetic tumor-specific andtissue-restricted pathways such as RAS and p53 pathwaysdrive RMS growth Tg(hsp70-HRASG12V) zebrafish embryoswere generated to evaluate gene expression that mimics RASpathway activation during tumorigenesis to defineRAS targetgenes [52] Another KRASG12D-induced zebrafish embryonalRMS was utilized to assess the therapeutic effects By theblockage of two major downstream signaling pathwaysMAPKERK and AKTS6K1 the model showed that inhibi-tion of translation initiation suppresses tumor cell prolifera-tion [52]

All the above zebrafish cancer models share similar cellu-lar features and molecular pathways with human RMS espe-cially ERMS and can demonstrate a response to therapiesin a similar manner with human RMS Thus a tremendousopportunity is available to implement these animal modelsinto different steps for novel targeted therapeutic develop-ment

10 Sarcoma

63 Mouse Models in RMS Mouse models established fromin vivo studies may contribute to the understanding of thegenetic basis in tumor development and progression andmayhelp to test the efficacies of novel antineoplastic agents

The importance of PAX3PAX7-FKHR fusion proteins inthe progression of ARMS tumorigenesis is evident from bothin vitro and in vivo studies Transgenic mouse models gener-ated with the PAX-FKHR fusion genemimic the formation ofhuman ARMS [67] A mouse model was developed with het-erozygous loss of Sufu a tumor suppressor inHedgehog (Hh)signaling combined with p53 loss driven ERMS with a lowpenetrance (9) [53] These studies indicate that the modelsconsistently formingARMSnot only need the introduction ofthe PAX-FKHR fusion gene but also need to be accompaniedby other genetic events such as p53 pathway disruption [54]

Compared with ARMS ERMS models are more complexto generate because of their lower tumor penetrance andlonger latency JW41 cells from the p53c-fos double mutantmice models resemble human ERMS cells morphologicallyand express similar characteristic markers [55] The overex-pression of the Wnt2 gene identified in JW41 cells confirmedin human RMS cell lines can allow for further analysis of theWnt signaling pathway Both of these results indicate that thedownregulation of theWnt pathway contributes to resistanceto apoptosis and the inhibition of myogenic differentiation inERMS

Using Ptch1 p53 andor Rb1 conditional mouse modelsand controlling prenatal or postnatal myogenic cell of originRubin et al demonstrated that the loss of p53 in maturingmyoblasts related to tumorigenesis of ERMS [96] They alsoindicated that Rb1 alteration with other oncogenic factorswas strongly associated with an undifferentiated phenotypeacting as a modifier In addition they highlighted a sub-set of p53-deficient ERMSs arising from Myf6-expressingmyoblasts which had the same latency with Pax7-expressingmurine satellite cells in the study

7 Potential Therapeutic Targets in RMS

Detection of genomic imbalances and identification of thecrucial effective genes can contribute to identifying novelpotential biomarkers and relevant targets for clinical therapyin RMS

Survivin-responsive conditionally replicating adenovi-ruses can regulate multiple factors (Survm-CRAs) [41]Although Survm-CRA can efficiently replicate and potentlyinduce cell death in RMS cells the cytotoxic effects are morepronounced in RSC-enriched or RSC-purified cells thanin RSC-exiguous or progeny-purified cells Injections ofSurvm-CRAs into tumor nodules generated by transplantingRSC-enriched cells induce significant death in RMS cells andregression of tumor nodulesThe unique therapeutic featuresof Survm-CRA include not only its therapeutic effectivenessagainst RMS but also its increasing effectiveness against CSCsuggesting that Survm-CRA may be a promising anticanceragent [41]

Rapamycin an inhibitor of mTOR can abrogate RMStumor growth in a xenograft mouse model [97] Thetumors in the rapamycin-treatedmice group show significant

reduction of proliferation and invasiveness and inductionof apoptosis The tumor growth inhibition is simultaneousassociated with the diminution of mTOR and Hh pathwayswhich are implicated in the pathogenesis of RMSThe resultsindicate that using rapamycin either alone or in combinationwith traditional chemotherapeutic drugs may represent apotential targeted agent for therapeutic intervention Ampli-fication and mutational activation of Fibroblast Growth Fac-tor Receptor 4 (FGFR4) in RMS cells promote tumor pro-gression [98] Ponatinib is the most potent FGFR4 inhibitorto block wild-type RMS cell growth mutate FGFR4 throughincreasing apoptosis and suppress the FGFR4 downstreamtarget STAT3 Ponatinib treatment slows tumor growth inRMS mouse models expressing mutated FGFR4 [98] Pona-tinib as an FGFR4 inhibitor shows potential to act as aneffective therapeutic agent for RMS tumorsThe expression ofCXC chemokine receptor 4 (CXCR4) CXCR7 and VascularEndothelial Growth Factor (VEGF) indicates poor prognosisin a variety of malignancies including RMS cell lines Mostsamples in a large series of clinical RMS cases show asso-ciation with high expression of these antagonists regardlessof their subtypes [99] However there are significant corre-lations with high expression of CXCR4 and VEGF in bothERMS and ARMS High VEGF expression is predictive ofadverse prognostic factors in RMS as the expression of theseangiogenesis factors was compared with clinicopathologicalparameters and prognosis Considering their overexpressionin RMS these chemokine receptors and VEGF could providepotential molecular therapeutic targets in RMS Some ARMScell lines have undergone apoptosis in response to antineo-plastic drugs such as bortezomib The proapoptotic BH3only family member NOXA acts as an upregulated proteindownstream PAX3-FKHR resulting in increased cell sensi-tivity to bortezomib Apoptosis in response to bortezomibcan be reversed by shRNA knockdown of Noxa PAX3-FKHR upregulation of Noxa creates a potential therapeuticinsight into inducing apoptosis in ARMS cellsThis apoptosispathway could represent a specific targeted therapy againstPAX3-FKHR-expressing ARMS cells [100]

Proton pump activation resulting from intracellular acid-ificationmay be an energetic mechanism to escape apoptosisV-ATPase is an ATP-driven proton pump acidifying theintracellular compartment and transports protons across theplasma membrane Esomeprazole as a V-ATPase inhibitor inthe PPI administration interferes with intensive ion trans-porter activity of RMS cells and induces remarkable cytotoxi-city RMS expresses a higher level ofV-ATPase comparedwithother sarcomasTherefore RMS should be very susceptible toesomeprazole treatment and V-ATPase could be consideredas a promising selective target for treatment [101]

8 Conclusion and Future Prospects

For RMS the current challenges include how to iden-tify promising novel therapeutics and integrate them intoexisting therapy For better understanding of congenitaland epigenetic modifications in the development of RMSthis review summarizes the recent genome-wide studies onmolecular and genetic alterations to decipher the underlying

Sarcoma 11

tumorigenesis mechanisms There is a link between geneticand epigenetic alterations responsible for a variety of tumorcell growth proliferation differentiation apoptosis andtherapy-resistance mechanisms The identification of theprognostic value of the PAX-FKHR fusion status in RMSis one of the most important shifts in the lineage and therisk assessment of RMS which exerts an oncogenic effectthrough multiple pathways and is incorporated into mostrelevant studies The misbalanced expression of a number ofmiRs involved in the regulation of myogenic differentiationalso implicates the tumorsrsquo escape suppressive mechanismsMoreover we described several in vivo models of RMSto explore underlying histogenesis and different aspects oftumorigenesis Recent work on CSCs has also contributedto understanding the cell origins refining molecular char-acterization and elucidating the underlying basis of refrac-tory responses to current adjuvant treatments of subsets oftumorsThese integrative approaches can provide opportuni-ties to identify diagnostic and prognostic biomarkers appliedto the individual targeted therapy and significantly improveRMS prognosis

Abbreviations

RMS RhabdomyosarcomaARMS Alveolar rhabdomyosarcomaERMS Embryonal rhabdomyosarcomaPRMS Pleomorphic rhabdomyosarcomaSRMS Sclerosingspindle cell

rhabdomyosarcomamiRNA miR MicroRNACSC Cancer stem cellMSC Mesenchymal stem cell

Conflict of Interests

The authors declare that there is no conflict of interestsregarding the publication of this paper

Acknowledgments

This work was supported in part by grants from the Gat-tegno and Wechsler funds Dr Duan was supported in partthrough a grant from Sarcoma Foundation of America (SFA)a grant from National Cancer Institute (NCI)National Insti-tutes of Health (NIH) UO1 CA 151452 and a grant from anAcademic Enrichment Fund of MGH Orthopedic Surgery

References

[1] L Yang T Takimoto and J Fujimoto ldquoPrognostic model forpredicting overall survival in children and adolescents withrhabdomyosarcomardquo BMC Cancer vol 14 no 1 article 6542014

[2] D Egas-Bejar and W W Huh ldquoRhabdomyosarcoma in adoles-cent and young adult patients current perspectivesrdquo AdolescentHealth Medicine andTherapeutics vol 5 pp 115ndash125 2014

[3] J C Breneman E Lyden A S Pappo et al ldquoPrognosticfactors and clinical outcomes in children and adolescents withmetastatic rhabdomyosarcomamdasha report from the intergrouprhabdomyosarcoma study IVrdquo Journal of Clinical Oncology vol21 no 1 pp 78ndash84 2003

[4] V D Soleimani and M A Rudnicki ldquoNew insights into theorigin and the genetic basis of rhabdomyosarcomasrdquo CancerCell vol 19 no 2 pp 157ndash159 2011

[5] T Glass D D Cochrane S R Rassekh K Goddard and JHukin ldquoGrowing teratoma syndrome in intracranial non-ger-minomatous germ cell tumors (iNGGCTs) a risk for secondarymalignant transformationmdasha report of two casesrdquo Childrsquos Ner-vous System vol 30 no 5 pp 953ndash957 2014

[6] J Ji C Eng and K Hemminki ldquoFamilial risk for soft tissuetumors a nation-wide epidemiological study from SwedenrdquoJournal of Cancer Research and Clinical Oncology vol 134 no5 pp 617ndash624 2008

[7] C D M Fletcher J A Bridge P Hogendoorn and F MertensWHO Classification of Tumours of Soft Tissue and Bone vol 5IARC Press Paris France 4th edition 2013

[8] I Sultan I Qaddoumi S Yaser C Rodriguez-Galindo and AFerrari ldquoComparing adult and pediatric rhabdomyosarcoma inthe surveillance epidemiology and end results program 1973 to2005 an analysis of 2600 patientsrdquo Journal of Clinical Oncologyvol 27 no 20 pp 3391ndash3397 2009

[9] B Raney W Huh D Hawkins et al ldquoOutcome of patientswith localized orbital sarcoma who relapsed following treat-ment on Intergroup Rhabdomyosarcoma Study Group (IRSG)Protocols-III and -IV 1984ndash1997 a report from the ChildrenrsquosOncology Grouprdquo Pediatric Blood and Cancer vol 60 no 3 pp371ndash376 2013

[10] W Crist E A Gehan A H Ragab et al ldquoThe third intergrouprhabdomyosarcoma studyrdquo Journal of Clinical Oncology vol 13no 3 pp 610ndash630 1995

[11] C Liu D Li J Jiang et al ldquoAnalysis of molecular cytogeneticalteration in rhabdomyosarcoma by array comparative genomichybridizationrdquo PLoS ONE vol 9 no 4 Article ID e94924 2014

[12] DWilliamson E Missiaglia A de Reynies et al ldquoFusion gene-negative alveolar rhabdomyosarcoma is clinically and molec-ularly indistinguishable from embryonal rhabdomyosarcomardquoJournal of Clinical Oncology vol 28 no 13 pp 2151ndash2158 2010

[13] V Paulson G Chandler D Rakheja et al ldquoHigh-resolutionarray CGH identifies common mechanisms that drive embry-onal rhabdomyosarcoma pathogenesisrdquo Genes Chromosomesand Cancer vol 50 no 6 pp 397ndash408 2011

[14] S Park J Lee I-G Do et al ldquoAberrant CDK4 amplificationin refractory rhabdomyosarcoma as identified by genomicprofilingrdquo Scientific Reports vol 4 article 3623 2014

[15] C Liu D Li J Hu et al ldquoChromosomal and genetic imbalancesin Chinese patients with rhabdomyosarcoma detected by high-resolution array comparative genomic hybridizationrdquo Interna-tional Journal of Clinical and Experimental Pathology vol 7 no2 pp 690ndash698 2014

[16] Q-X Li C-X Liu C-P Chun et al ldquoChromosomal imbalancesrevealed in primary rhabdomyo-sarcomas by comparativegenomic hybridizationrdquo Chinese Medical Journal vol 122 no11 pp 1277ndash1282 2009

[17] E Missiaglia J Selfe M Hamdi et al ldquoGenomic imbalancesin rhabdomyosarcoma cell lines affect expression of genesfrequently altered in primary tumors an approach to identifycandidate genes involved in tumor developmentrdquo Genes Chro-mosomes and Cancer vol 48 no 6 pp 455ndash467 2009

12 Sarcoma

[18] F G Barr F Duan L M Smith et al ldquoGenomic and clinicalanalyses of 2p24 and 12q13-q14 amplification in alveolar rhab-domyosarcoma a report from the childrenrsquos oncology grouprdquoGenes Chromosomes and Cancer vol 48 no 8 pp 661ndash6722009

[19] T Gordon AMcManus J Anderson et al ldquoCytogenetic abnor-malities in 42 rhabdomyosarcoma a United Kingdom CancerCytogenetics Group StudyrdquoMedical and Pediatric Oncology vol36 no 2 pp 259ndash267 2001

[20] X Chen E Stewart A A Shelat et al ldquoTargeting oxidativestress in embryonal rhabdomyosarcomardquo Cancer Cell vol 24no 6 pp 710ndash724 2013

[21] S Lizard-Nacol F Mugneret C Volk C Turc-Carel M Favrotand T Philip ldquoTranslocation (213)(q37q14) in alveolar rhab-domyosarcoma a new caserdquo Cancer Genetics and Cytogeneticsvol 25 no 2 pp 373ndash374 1987

[22] P H B Sorensen J C Lynch S J Qualman et al ldquoPAX3-FKHR and PAX7-FKHR gene fusions are prognostic indicatorsin alveolar rhabdomyosarcoma a report from the childrenrsquosoncology grouprdquo Journal of Clinical Oncology vol 20 no 11 pp2672ndash2679 2002

[23] A D Marshall and G C Grosveld ldquoAlveolar rhabdomyo-sarcomamdashthe molecular drivers of PAX37-FOXO1-inducedtumorigenesisrdquo Skeletal Muscle vol 2 no 1 article 25 2012

[24] E Missiaglia D Williamson J Chisholm et al ldquoPAX3FOXO1fusion gene status is the key prognostic molecular markerin rhabdomyosarcoma and significantly improves current riskstratificationrdquo Journal of Clinical Oncology vol 30 no 14 pp1670ndash1677 2012

[25] J F Shern L Chen J Chmielecki et al ldquoComprehensivegenomic analysis of rhabdomyosarcoma reveals a landscape ofalterations affecting a common genetic axis in fusion-positiveand fusion-negative tumorsrdquo Cancer Discovery vol 4 no 2 pp216ndash231 2014

[26] MWachtel M Dettling E Koscielniak et al ldquoGene expressionsignatures identify rhabdomyosarcoma subtypes and detect anovel t(22)(q35p23) translocation fusing PAX3 to NCOA1rdquoCancer Research vol 64 no 16 pp 5539ndash5545 2004

[27] E Davicioni M J Anderson F G Finckenstein et al ldquoMolec-ular classification of rhabdomyosarcomamdashgenotypic and phe-notypic determinants of diagnosis a report from the ChildrenrsquosOncology Grouprdquo The American Journal of Pathology vol 174no 2 pp 550ndash564 2009

[28] M Visser C Sijmons J Bras et al ldquoAllelotype of pediatric rhab-domyosarcomardquo Oncogene vol 15 no 11 pp 1309ndash1314 1997

[29] R Gil-Benso T San-Miguel R C Callaghan et al ldquoChromo-somal and genetic changes produced in tumoral progression ofembryonal rhabdomyosarcomardquo Histopathology vol 62 no 5pp 816ndash819 2013

[30] S X Skapek J Anderson F G Barr et al ldquoPAX-FOXO1 fusionstatus drives unfavorable outcome for children with rhabdom-yosarcoma a childrenrsquos oncology group reportrdquo Pediatric Bloodand Cancer vol 60 no 9 pp 1411ndash1417 2013

[31] HHosoi N Kakazu E Konishi et al ldquoA novel PAX3 rearrange-ment in embryonal rhabdomyosarcomardquo Cancer Genetics andCytogenetics vol 189 no 2 pp 98ndash104 2009

[32] E Takaoka H Sonobe K Akimaru et al ldquoMultiple sites ofhighly amplified DNA sequences detected bymolecular cytoge-netic analysis inHS-RMS-2 a newpleomorphic rhabdomyosar-coma cell linerdquo American Journal of Cancer Research vol 2 no2 pp 141ndash152 2012

[33] K Szuhai D de Jong W Y Leung C D M Fletcher and P CWHogendoorn ldquoTransactivatingmutation of theMYOD1 geneis a frequent event in adult spindle cell rhabdomyosarcomardquoJournal of Pathology vol 232 no 3 pp 300ndash307 2014

[34] N P Agaram C-L Chen L Zhang M P Laquaglia L Wexlerand C R Antonescu ldquoRecurrent MYOD1 mutations in pedi-atric and adult sclerosing and spindle cell rhabdomyosarcomasEvidence for a common pathogenesisrdquoGenes Chromosomes andCancer vol 53 no 9 pp 779ndash787 2014

[35] H Yoshida M Miyachi K Sakamoto et al ldquoPAX3-NCOA2fusion gene has a dual role in promoting the proliferation andinhibiting the myogenic differentiation of rhabdomyosarcomacellsrdquo Oncogene vol 33 pp 5601ndash5608 2013

[36] J M Mosquera A Sboner L Zhang et al ldquoRecurrent NCOA2gene rearrangements in congenitalinfantile spindle cell rhab-domyosarcomardquo Genes Chromosomes and Cancer vol 52 no6 pp 538ndash550 2013

[37] V Thavaraj A Sethi and L S Arya ldquoIncomplete Beckwith-Wiedemann syndrome in a child with orbital rhabdomyosar-comardquo Indian Pediatrics vol 39 no 3 pp 299ndash304 2002

[38] D Walter S Satheesha P Albrecht et al ldquoCD133 positiveembryonal rhabdomyosarcoma stem-like cell population isenriched in rhabdospheresrdquo PLoS ONE vol 6 no 5 Article IDe19506 2011

[39] Y-X Ren F G Finckenstein D A Abdueva et al ldquoMousemesenchymal stem cells expressing PAX-FKHR form alveolarrhabdomyosarcomas by cooperating with secondary muta-tionsrdquo Cancer Research vol 68 no 16 pp 6587ndash6597 2008

[40] M Salerno S Avnet G Bonuccelli S Hosogi D Granchi andN Baldini ldquoImpairment of lysosomal activity as a therapeuticmodality targeting cancer stem cells of embryonal rhabdom-yosarcoma cell line RDrdquo PLoS ONE vol 9 no 10 Article IDe110340 2014

[41] K Tanoue Y Wang M Ikeda et al ldquoSurvivin-responsive con-ditionally replicating adenovirus kills rhabdomyosarcoma stemcells more efficiently than their progenyrdquo Journal of Transla-tional Medicine vol 12 no 1 article 27 2014

[42] P K Rao EMissiaglia L Shields et al ldquoDistinct roles formiR-1and miR-133a in the proliferation and differentiation of rhab-domyosarcoma cellsrdquo The FASEB Journal vol 24 no 9 pp3427ndash3437 2010

[43] D Yan X D Dong X Chen et al ldquoMicroRNA-1206 targets c-met and inhibits rhabdomyosarcomadevelopmentrdquoThe Journalof Biological Chemistry vol 284 no 43 pp 29596ndash29604 2009

[44] I Marchesi A Giordano and L Bagella ldquoRoles of enhancerof zeste homolog 2 from skeletal muscle differentiation torhabdomyosarcoma carcinogenesisrdquoCell Cycle vol 13 no 4 pp516ndash527 2014

[45] M M Sun J F Li L L Guo et al ldquoTGF-1205731 suppression ofmicroRNA-450b-5p expression a novel mechanism for block-ingmyogenic differentiation of rhabdomyosarcomardquoOncogenevol 33 no 16 pp 2075ndash2086 2014

[46] Y Diao X Guo L Jiang et al ldquoMiR-203 a tumor suppressorfrequently down-regulated by promoter hypermethylation inrhabdomyosarcomardquo The Journal of Biological Chemistry vol289 no 1 pp 529ndash539 2014

[47] H-J Huang J Liu HHua et al ldquoMiR-214 andN-ras regulatoryloop suppresses rhabdomyosarcoma cell growth and xenografttumorigenesisrdquo Oncotarget vol 5 no 8 pp 2161ndash2175 2014

Sarcoma 13

[48] A L Sarver L Li and S Subramanian ldquomicroRNA miR-183functions as an oncogene by targeting the transcription factorEGR1 and promoting tumor cell migrationrdquo Cancer Researchvol 70 no 23 pp 9570ndash9580 2010

[49] R L Galindo J A Allport and E N Olson ldquoA Drosophilamodel of the rhabdomyosarcoma initiator PAX7-FKHRrdquo Pro-ceedings of the National Academy of Sciences of the United Statesof America vol 103 no 36 pp 13439ndash13444 2006

[50] U Avirneni-Vadlamudi K A Galindo T R Endicott V Paul-son S Cameron and R L Galindo ldquoDrosophila and mammal-ian models uncover a role for the myoblast fusion gene TANC1in rhabdomyosarcomardquo The Journal of Clinical Investigationvol 122 no 1 pp 403ndash407 2012

[51] E Y Chen andDM Langenau ldquoZebrafishmodels of rhabdom-yosarcomardquoMethods in Cell Biology vol 105 pp 383ndash402 2011

[52] X Le E K Pugach S Hettmer et al ldquoA novel chemicalscreening strategy in zebrafish identifies common pathways inembryogenesis and rhabdomyosarcoma developmentrdquo Devel-opment vol 140 no 11 pp 2354ndash2364 2012

[53] Y Lee R Kawagoe K Sasai et al ldquoLoss of suppressor-of-fusedfunction promotes tumorigenesisrdquoOncogene vol 26 no 44 pp6442ndash6447 2007

[54] C Keller B R Arenkiel C M Coffin N El-Bardeesy R ADePinho and M R Capecchi ldquoAlveolar rhabdomyosarcomasin conditional Pax3Fkhr mice cooperativity of Ink4aARF andTrp53 loss of functionrdquo Genes and Development vol 18 no 21pp 2614ndash2626 2004

[55] S Singh CVinson CMGurley et al ldquoImpairedWnt signalingin embryonal rhabdomyosarcoma cells from p53c-fos doublemutant micerdquo American Journal of Pathology vol 177 no 4 pp2055ndash2066 2010

[56] G J Schaaf J M Ruijter F Van Ruissen et al ldquoFull transcrip-tome analysis of rhabdomyosarcoma normal and fetal skeletalmuscle statistical comparison of multiple SAGE librariesrdquo TheFASEB Journal vol 19 no 3 pp 404ndash406 2005

[57] D S Hawkins A A Gupta and E R Rudzinski ldquoWhat is newin the biology and treatment of pediatric rhabdomyosarcomardquoCurrent Opinion in Pediatrics vol 26 no 1 pp 50ndash56 2014

[58] M Zhang J Truscott and J Davie ldquoLoss of MEF2D expressioninhibits differentiation and contributes to oncogenesis in rhab-domyosarcoma cellsrdquoMolecular Cancer vol 12 no 1 article 1502013

[59] M Ciesla J Dulak and A Jozkowicz ldquomicroRNAs and epi-genetic mechanisms of rhabdomyosarcoma developmentrdquo TheInternational Journal of Biochemistry amp Cell Biology vol 53 pp482ndash492 2014

[60] S Ricaud B Vernus M Duclos et al ldquoInhibition of autocrinesecretion of myostatin enhances terminal differentiation inhuman rhabdomyosarcoma cellsrdquo Oncogene vol 22 no 51 pp8221ndash8232 2003

[61] C S Guo C Degnin T A Fiddler D Stauffer andM JThayerldquoRegulation of MyoD activity and muscle cell differentiation byMDM2 pRb and Sp1rdquoThe Journal of Biological Chemistry vol278 no 25 pp 22615ndash22622 2003

[62] S Rossi E Stoppani P L Puri and A Fanzani ldquoDifferenti-ation of human rhabdomyosarcoma RD cells is regulated byreciprocal functional interactions between myostatin p38 andextracellular regulated kinase signalling pathwaysrdquo EuropeanJournal of Cancer vol 47 no 7 pp 1095ndash1105 2011

[63] Z S Walters B Villarejo-Balcells D Olmos et al ldquoJARID2is a direct target of the PAX3-FOXO1 fusion protein andinhibits myogenic differentiation of rhabdomyosarcoma cellsrdquoOncogene vol 33 no 9 pp 1148ndash1157 2014

[64] T Hosoyama M I Aslam J Abraham et al ldquoIL-4R drives ded-ifferentiation mitogenesis and metastasis in rhabdomyosar-comardquo Clinical Cancer Research vol 17 no 9 pp 2757ndash27662011

[65] F S Dela Cruz ldquoCancer stem cells in pediatric sarcomasrdquoFrontiers in Oncology vol 3 article 168 2013

[66] M J Bissell and W C Hines ldquoWhy donrsquot we get more cancerA proposed role of the microenvironment in restraining cancerprogressionrdquo Nature Medicine vol 17 no 3 pp 320ndash329 2011

[67] E Charytonowicz C Cordon-Cardo I Matushansky and MZiman ldquoAlveolar rhabdomyosarcoma is the cell of origin amesenchymal stem cellrdquoCancer Letters vol 279 no 2 pp 126ndash136 2009

[68] W K Cavenee ldquoMuscling in on rhabdomyosarcomardquo NatureMedicine vol 8 no 11 pp 1200ndash1201 2002

[69] B Zhu and J K Davie ldquoNew insights into signalling-pathwayalterations in rhabdomyosarcomardquo British Journal of Cancervol 112 no 2 pp 227ndash231 2015

[70] M Zhang C M Linardic and D G Kirsch ldquoRAS and ROS inrhabdomyosarcomardquo Cancer Cell vol 24 no 6 pp 689ndash6912013

[71] S C Marianna L Ianzano and G Nicoletti ldquoTumor sup-pressor genes promote rhabdomyosarcoma progression in p53heterozygousrdquo Oncotarget vol 5 no 1 pp 108ndash119 2013

[72] SWang L Guo L Dong et al ldquoTGF-beta1 signal pathwaymaycontribute to rhabdomyosarcoma development by inhibitingdifferentiationrdquo Cancer Science vol 101 no 5 pp 1108ndash11162010

[73] J Wesche K Haglund and E M Haugsten ldquoFibroblast growthfactors and their receptors in cancerrdquo Biochemical Journal vol437 no 2 pp 199ndash213 2011

[74] M Wachtel J Rakic M Okoniewski P Bode F Niggli andB W Schafer ldquoFGFR4 signaling couples to Bim and not Bmfto discriminate subsets of alveolar rhabdomyosarcoma cellsrdquoInternational Journal of Cancer vol 135 no 7 pp 1543ndash15522014

[75] P L Puri ZWu P Zhang et al ldquoInduction of terminal differen-tiation by constitutive activation of p38 MAP kinase in humanrhabdomyosarcoma cellsrdquo Genes and Development vol 14 no5 pp 574ndash584 2000

[76] A M Tremblay E Missiaglia G G Galli et al ldquoThe Hippotransducer YAP1 transforms activated satellite cells and is apotent effector of embryonal rhabdomyosarcoma formationrdquoCancer Cell vol 26 no 2 pp 273ndash287 2014

[77] R N Judson A M Tremblay P Knopp et al ldquoThe hippo path-way member Yap plays a key role in influencing fate decisionsin muscle satellite cellsrdquo Journal of Cell Science vol 125 no 24pp 6009ndash6019 2012

[78] L E S Crose K A Galindo J G Kephart et al ldquoAlve-olar rhabdomyosarcoma-associated PAX3-FOXO1 promotestumorigenesis via Hippo pathway suppressionrdquo Journal ofClinical Investigation vol 124 no 1 pp 285ndash296 2014

[79] B Belyea J G Kephart J Blum D G Kirsch and C MLinardic ldquoEmbryonic signaling pathways and rhabdomyosar-coma contributions to cancer development and opportunitiesfor therapeutic targetingrdquo Sarcoma vol 2012 Article ID 40623913 pages 2012

14 Sarcoma

[80] J Huertas-Martinez S Rello-Varona D Herrero-Martin et alldquoCaveolin-1 is down-regulated in alveolar rhabdomyosarcomasand negatively regulates tumor growthrdquo Oncotarget vol 5 no20 pp 9744ndash9755 2014

[81] S Rossi P L Poliani CMissale EMonti andA Fanzani ldquoCav-eolins in rhabdomyosarcomardquo Journal of Cellular andMolecularMedicine vol 15 no 12 pp 2553ndash2568 2011

[82] P G Tsioli E S Patsouris C Giaginis and S E TheocharisldquoDNA repair systems in rhabdomyosarcomardquo Histology andHistopathology vol 28 no 8 pp 971ndash984 2013

[83] E Gatzidou C Michailidi S Tseleni-Balafouta and S Theo-charis ldquoAn epitome of DNA repair related genes and mecha-nisms in thyroid carcinomardquo Cancer Letters vol 290 no 2 pp139ndash147 2010

[84] R Rota R Ciarapica A Giordano L Miele and F LocatellildquoMicroRNAs in rhabdomyosarcoma pathogenetic implicationsand translational potentialityrdquoMolecular Cancer vol 10 article120 2011

[85] A Gougelet J Perez D Pissaloux et al ldquoMiRNA profiling howto bypass the current difficulties in the diagnosis and treatmentof sarcomasrdquo Sarcoma vol 2011 Article ID 460650 13 pages2011

[86] R Ciarapica G Russo F Verginelli et al ldquoDeregulated expres-sion of miR-26a and Ezh2 in rhabdomyosarcomardquo Cell Cyclevol 8 no 1 pp 172ndash175 2009

[87] L Li A L Sarver S Alamgir and S Subramanian ldquoDownreg-ulation of microRNAs miR-1 -206 and -29 stabilizes PAX3 andCCND2 expression in rhabdomyosarcomardquo Laboratory Investi-gation vol 92 no 4 pp 571ndash583 2012

[88] S-Y Ng G K Bogu B Soh and L W Stanton ldquoThe longnoncoding RNA RMST interacts with SOX2 to regulate neu-rogenesisrdquoMolecular Cell vol 51 no 3 pp 349ndash359 2013

[89] C A Lynch B Tycko T H Bestor and C P Walsh ldquoReac-tivation of a silenced H19 gene in human rhabdomyosarcomaby demethylation of DNA but not by histone hyperacetylationrdquoMolecular Cancer vol 1 article 2 2002

[90] P Onyango and A P Feinberg ldquoA nucleolar protein H19opposite tumor suppressor (HOTS) is a tumor growth inhibitorencoded by a human imprinted H19 antisense transcriptrdquoProceedings of the National Academy of Sciences of the UnitedStates of America vol 108 no 40 pp 16759ndash16764 2011

[91] C E Albacker N Y Storer E M Langdon et al ldquoThe his-tone methyltransferase SUV39H1 suppresses embryonal rhab-domyosarcoma formation in zebrafishrdquo PLoS ONE vol 8 no 5Article ID e64969 2013

[92] N Y Storer R M White A Uong et al ldquoZebrafish rhab-domyosarcoma reflects the developmental stage of oncogeneexpression during myogenesisrdquo Development vol 140 no 14pp 3040ndash3050 2013

[93] M E Hatley W Tang M R Garcia et al ldquoA mouse modelof rhabdomyosarcoma originating from the adipocyte lineagerdquoCancer Cell vol 22 no 4 pp 536ndash546 2012

[94] V Hosur A Kavirayani J Riefler et al ldquoDystrophin and dysfer-lin doublemutantmice a novelmodel for rhabdomyosarcomardquoCancer Genetics vol 205 no 5 pp 232ndash241 2012

[95] W Goessling T E North and L I Zon ldquoNew waves of discov-ery modeling cancer in zebrafishrdquo Journal of Clinical Oncologyvol 25 no 17 pp 2473ndash2479 2007

[96] B P Rubin K Nishijo H-I H Chen et al ldquoEvidence for anunanticipated relationship between undifferentiated pleomor-phic sarcoma and embryonal rhabdomyosarcomardquo Cancer Cellvol 19 no 2 pp 177ndash191 2011

[97] S Z Kaylani J Xu R K Srivastava L Kopelovich J G Presseyand M Athar ldquoRapamycin targeting mTOR and hedgehog sig-naling pathways blocks human rhabdomyosarcoma growth inxenograft murine modelrdquo Biochemical and Biophysical ResearchCommunications vol 435 no 4 pp 557ndash561 2013

[98] S Q Li A T Cheuk J F Shern et al ldquoTargeting wild-type andmutationally activated FGFR4 in rhabdomyosarcoma with theinhibitor ponatinib (AP24534)rdquoPLoSONE vol 8 no 10 ArticleID e76551 2013

[99] K Miyoshi K Kohashi F Fushimi et al ldquoClose correlationbetween CXCR4 and VEGF expression and frequent CXCR7expression in rhabdomyosarcomardquo Human Pathology vol 45no 9 pp 1900ndash1909 2014

[100] ADMarshall F Picchione R I KGeltink andGCGrosveldldquoPAX3-FOXO1 induces up-regulation of Noxa sensitizing alve-olar rhabdomyosarcoma cells to apoptosisrdquo Neoplasia vol 15no 7 pp 738ndash748 2013

[101] F Perut S Avnet C Fotia et al ldquoV-ATPase as an effective thera-peutic target for sarcomasrdquo Experimental Cell Research vol 320no 1 pp 21ndash32 2014

Submit your manuscripts athttpwwwhindawicom

Stem CellsInternational

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

MEDIATORSINFLAMMATION

of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Behavioural Neurology

EndocrinologyInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Disease Markers

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

BioMed Research International

OncologyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Oxidative Medicine and Cellular Longevity

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

PPAR Research

The Scientific World JournalHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Immunology ResearchHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal of

ObesityJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Computational and Mathematical Methods in Medicine

OphthalmologyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Diabetes ResearchJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Research and TreatmentAIDS

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Gastroenterology Research and Practice

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Parkinsonrsquos Disease

Evidence-Based Complementary and Alternative Medicine

Volume 2014Hindawi Publishing Corporationhttpwwwhindawicom

Page 4: Review Article Rhabdomyosarcoma: Advances in Molecular and

4 Sarcoma

Table 2 The histological types of rhabdomyosarcoma

Histological type Predilection population Predilection site Risk category Genetic change

ERMS Infants or children under 10years old Head and neck region Intermediate LOH at chromosome 11p155

ARMS Adolescents and young adults Deep tissue of extremity High t(213)(q35q14) t(113)(p36q14)

PRMS Adult males Throughout the body HighJUN (1p31) MYC (8q24)CCND1 (11q13) INT2 (11q133)MDM2 (12q143ndashq15)

SRMSIn the first decade of life with asecond mode centered aroundthe fifth decade

The extremities headand neck Superior SRF-NCOA2 MYOD1 mutation

ERMS embryonal rhabdomyosarcomaARMS alveolar rhabdomyosarcomaPRMS pleomorphic rhabdomyosarcomaSRMS spindle cellsclerosing rhabdomyosarcoma

can result from the inactivation of the parental bias of chro-mosome 11p15 which is the most common rearrangementin ERMS [29] The proportion of ERMS with LOH alongchromosome 11 is considerably higher than in other subtypes[27] In histopathological analysis ERMS expresses lowPAX3levels and elevated PAX7 levels [30] Hosoi et al identified ahidden 2q35 breakpoint as a novel PAX3 rearrangement incomplex chromosomal translocations in ERMS [31]

24 Chromosomal Alterations in Other RMS There are lim-ited studies on the biological pathways involved in othersubtypes of RMS compared with the two major subtypesARMS andERMS Fluorescence In SituHybridization (FISH)reveals amplification of JUN (1p31) MYC (8q24) CCND1(11q13) INT2 (11q133) MDM2 (12q143ndashq15) and MALT(18q21) in these tumor cells contributing to the pathogenesisof PRMS [32] MYOD1 homozygous mutations are alsoreported as frequent recurrent and pathognomonic eventsin adult-type SRMS [33 34] (Table 2) Furthermore four outof five pediatric tumors showedMYOD1mutations in a study[34] Yoshida et al described a 8q13 locus (NCOA2) generearrangement in a small subset of SRMS occurring uniquelyin the infantilecongenital setting fused with key transcrip-tion factors involved in skeletal muscle differentiation suchas SRF and TEAD1 [35] They also identified NCOA2 as acandidate PAX3 partner geneThePAX3-NCOA2 fusion geneplays a dual role in the tumorigenesis of RMS promotion ofthe proliferation and inhibition of the myogenic differenti-ation of RMS cells [35] As PAX3-NCOA2-induced tumorsgrow more slowly SRMS that are PAX3-NCOA2 fusion-positive could be associated with a very favorable prognosis[36]

Not all RMSs occur as sporadic primary tumors Occa-sionally RMS inherits a mutant gene as part of an establishedfamilial syndrome For example Beckwith-Wiedemann Syn-drome (BWS) with RMS involves dysregulation or alterationof imprinted genes in the 11p155 chromosomal regionincluding IGF2 H19 and CDKN1C (p57KIP2) [37]

3 Cell of Origin in RMS

There have been a number of studies aimed at deciphering thecell origin for RMS [56ndash61]

31 Myogenic Differentiation in the Tumorigenesis Severalstudies have focused on elucidating the mechanisms govern-ing the impaired myogenic program in RMS [62 63] (Fig-ure 2) RMSoriginates as a consequence of regulatory disrup-tion of the growth and differentiation of myogenic precursorcells Progenitor cells reside in muscle and their activationresults in either proper myogenesis or aberrant signalingpathways leading to the development of RMS Based on theskeletal muscle lineage a complete transcriptome analysis ofRMS was performed to compare normal and fetal muscles[56] The high degree of similarity between fetal muscle andRMS expression profiles reflects the undifferentiated myo-genic nature of RMS The genes exclusively upregulated inRMS including FGFR4 NOTCH2 UBE2C UHRF1 andYWHAB genes contribute to the failure of RMS cells to com-plete normal skeletal muscle development and progress toan alternative fate [56] RMS cells represent an arrested statein the development of normal skeletal muscle with regionalsilencing of differentiation factors leading to the maturationdefect in RMS [57] The expression pattern of muscle-specific proteins regulating myogenic differentiation hasbeen extensively examined in RMS The family of myogenictranscription factors (MYOD MYF5 myogenin and MRF4)are considered to be responsible for the determination of stemcells intomyoblasts anddifferentiation intomyocytes [58 59]All these factors are activated during the onset or progressionof myogenesis and are subsequently silenced to reach a finalmuscular differentiation There are statistical differences inthe different subtypes of RMS tumors with MYOD or myo-genin staining patterns Amplification of MDM2 in an RMScell line interferes with MYOD activity and consequentlyinhibits overt muscle cell differentiation [61] Interleukin-4receptor (IL-4R) has also been proposed to be important for

Sarcoma 5

Table 3 Studies identifying stem cells in RMS

Markersubstrate Source Stem cell gene Functional characterization Reference

CD133 Orthotopicxenograft model

OCT4 NANOG c-MYCPAX3 and SOX2

Correlating with poor overallsurvival

Walter et al2011 [38]

PAX-FKHR Bone marrow ofC57BL6 mice

Myf5 MEF2 MyoD andmyogenin

Determining the molecularmyogenic and histologicphenotype of ARMS

Ren et al 2008[39]

V-ATPase RD cell line NANOG and OCT34

Driving mechanisms of areduced sensitivity to anticancerdrugs and activities related toinvasion and metastasis

Salerno et al2014 [40]

Survm-CRAsKYM-1 cell line(FGFR3-positive)

FGFR3Therapeutic effectiveness againstall cell populations and increasedeffectiveness against CSCs

Tanoue et al2014 [41]

Migration

specification

Injured muscle

Terminal differentiation

Fusion

PAX7

Myoblasts

Myoblast

MyoD

MyoD

Myf5

PAX3

Rb

Ezh2

IGF2

HDAC4YY1

YY1

ERK

AKT

RMS

Myogenin

P38 kinase

Myostatin

Progenitor

cell

N-RAS

Myofibers

FGFR4

UBE2C

UHRF1

NOTCH2

YWHAB

Satellite cells

RUNX1 MEF2C

JDP2 NFIC

Myocytes

Restoration of proliferation blockade of

differentiation

Proliferatio

n

Cell circle arrestEarlydifferentiation

TGF-1205731

Figure 2 Myogenic pathways in the tumorigenesis of RMS In aberrant neoplastic condition progenitor cells residing in muscle result inaberrant pathways which lead to malignant transformation and fail to differentiate proliferate uncontrollably and form RMS Sharp arrows(rarr) indicate upregulationactivation and blunt arrows (perp) indicate downregulationinhibition

the maturation of myotubes [64] An IL-4R blockade mighthelp therapeutically to modulate the expression of myogenictranscription factors (MYOD or myogenin) [57 64]

32 Potential Cancer Stem Cell in RMS Cancer Stem Cells(CSCs) may contribute to inherent refractory responses tocurrent therapies andmetastasis [65]Many CSCmodels playan important role in the development of RMS [38ndash41 66](Table 3)

A study explored a potential important role for mesen-chymal stem cells (MSCs) as the cell of origin of ARMS [67]Ren et al confirmed that PAX-FKHR fusion genes commitMSCs to a myogenic lineage by inhibiting terminal differen-tiation and contributing to ARMS formation [39] Anotherstudy showed that PAX-FKHR induces skeletal myogenesisin MSCs by transactivating MYOD and myogenin and trans-forms mesenchymal progenitor cells to the skeletal muscle lin-eage leading to malignant formation resembling ARMS [39]

6 Sarcoma

METCDKN2AB

PAX37-FKHR

MDM2

RAS

Raf

GLI1

PDK1p53Rb

Hh

Myo MEF2

p21

Ptch

FGFR4

IGF1R

p16INK4Ap14ARF

p19ARF

IGF2

FGF

Figure 3 Genetic analyses of RMS have pinpointed several common alterations including inactivation of a master regulator of p53 andRb pathways CDKN2AB and activation of FGFR4 RAS and Hedgehog (Hh) signaling The modifications of these pathways influenceoncogenesis and metastatic potential

As a human hematopoietic CSC marker Walter et alproposed that upregulated CD133 in ERMS can act as aprognostic marker and might help with the development ofnovel targeted therapies for ERMS [38]

4 Signaling Pathway Alterations in RMS

Recent studies have looked into signaling pathway alterationsand their downstream effects in RMSThese new findings notonly helped to improve the understanding of this malignancybut also offered novel potential therapeutic strategies forimproved treatment for patientswithRMS [68 69] (Figure 3)

41 RAS Signaling Pathway RASmutations commonlymain-tain the protein in its GTP bound state and therefore renderit constitutively active in RMS Zhang et al identified RASfamily members NRAS KRAS and HRAS in the RASpathway as themost commonlymutated genes in ERMS [70]the mutation status of these genes is significantly associatedwith the risk of ERMS development [70] A study foundthat a majority of ERMS demonstrated activation of the RASpathway exclusively either by homozygous deletion of NF1or by point mutations in one of the RAS family members asdescribed above [13] Despite remarkable genetic and molec-ular heterogeneity most (93 4144) RMS tumors hijackeda common receptor tyrosine kinaseRASPIK3CA geneticaxis [25] It could occur via two alternative mechanismsrearrangement of a PAX gene and accumulation of mutationsthat were downstream targets of the PAX fusion protein

Skeletal muscle cells have a robust antioxidant defensesystem to protect the DNA lipids and proteins from thedeleterious effects of excess Reactive Oxygen Species (ROS)

Cancer cells also have elevated ROS due to their increasedmetabolic activity oncogenic stimulation andmitochondrialdysfunction These findings implicate RAS mutations andoxidative stress as potential therapeutic targets for high-riskERMS

42 IGF Signaling Pathway There is clear preclinical datathat supports the involvement of the Insulin-like GrowthFactor (IGF) signaling pathway in RMS tumorigenesis andprogression [69] Zhu and Davie demonstrated that IGFpromotes the proliferation of RMS cells while blocking IGFsignaling interferes with cell growth in vivoThe IGF receptorIGF-1R is highly overexpressed both on the cell surface andin the nucleus of RMS cells furthermore RMS cell lines aresensitive to IGF-1R inhibitionThe IGF-2 locus shows a loss ofimprinting in both ERMS andARMS tumors and the expres-sion of PAX3-FKHR has been demonstrated to upregulateIGF-2 and activate IGF pathway in ARMS [69] Mariannaet al found that IGF-2 and tumor suppressors p19Arf andp21Cip1 are overexpressed and prominently upregulated inRMS mouse models [71]

43 TGF-120573 Signaling Pathway The regulatory role of Trans-forming Growth Factor-120573 (TGF-120573) in RMS has been recentlystudied [72] Wang et al demonstrated that the expression ofTGF-1205731 is significantly higher in RMS than in normal skeletalmuscle The inhibition of TGF-1205731 expression by shRNA-expressing vectors reverses themalignant behavior of RMSbyinhibiting cell growth and inducingmyogenic differentiationTGF-1205731 shRNA induces myogenin expression a regulator ofmyogenic differentiation genes Myogenin acts as a target fornegative regulation of myogenesis by TGF-1205731 signals with

Sarcoma 7

a differentiation regulatory cascade These results suggestthat the TGF-1205731 signaling pathway disrupts the differentia-tion of myogenic progenitors leading to the development ofRMS

44 FGF Signaling Pathway Fibroblast Growth Factor (FGF)is crucial in embryonic development and functions to driveproliferation FGFs and their receptors (FGFRs) are essentialregulators in the processes of proliferation antiapoptosisdrug resistance and angiogenesis in RMS [69] RMS over-expresses the receptor tyrosine kinase FGFR4 which causesautophosphorylation and constitutive signaling in correlationwith poor differentiation and decreased survival [73] Recentwork has shown that FGF signaling can prevent ARMS cellsfrom apoptosis induced by targeting the IGF1-R-PI3K-mTORpathway [74]

45 ERK Signaling Pathway Previous studies have shownthat elevated myostatin expression deregulates ExtracellularRegulated Kinase (ERK) signaling and deficient activationof the p38 pathway contributes to the differentiation blockin RMS cells [62] The ERK pathway is frequently highlyactivated in RMS cells from embryonic derivation due to thepresence of activating RASmutations leading to reduced dif-ferentiation by blocking the p38 pathway [75]These findingsindicate that interventions that target themyostatinERKp38network could be an effective way to promote differentiationof RMS

46 Hippo Signaling Pathway Many upstream signal trans-duction proteins in response to cues from the cellular micro-environment regulate the core Hippo pathway [76] TheHippo pathway may limit tumorigenesis by inducing thecytosolic localization of the transcriptional cofactor Yes-Associated Protein 1 (YAP1) High YAP levels and activitiesincrease activated satellite cells and prevent differentiationby activating genes that inhibit differentiation [77] YAP1-TEAD1 was found to upregulate proproliferative and onco-genic genes and maintain the ERMS differentiation blockby interfering with MYOD1 and MEF2 prodifferentiationactivities [76] Inhibition of theHippo pathway is exhibited inRMS through downregulation of Hippo pathway tumor sup-pressors or upregulation of YAP [78] These data suggest thatHippo pathway dysfunction promotes RMS development

47 Notch Signaling Pathway and Wnt Signaling PathwayNotch pathway as an embryonic signaling pathway promotesmuscle stem cell maintenance by inhibiting myogenic earlydifferentiation and expanding pools of progenitor cells dur-ing both embryogenesis and postnatal muscle regenerationSimilar to Notch Wnt has various roles in embryonic fetaland neonatalmyogenesis during skeletalmyogenesis As bothpathways are regulators of myogenic lineage determinationand maturation RMS can arise from disordered regulationof these normal developmental pathways [79]

48 Caveolins in RMS Recent findings have shown that cav-eolins are expressed in a cell stage-dependent manner inRMS [80 81] Caveolins are scaffolding proteins that regulate

several targets and pathways (such as p53 IGF RASERK andTGF-120573myostatin signaling) associated with RMS develop-ment and therefore loss or gain of caveolin function is ableto generate multiple effects on the tumor behaviorThereforerecognizing their precise roles in RMS progression will becrucial to elaborate targeted therapies particularly as oneof the three different isoforms CAV1 could act as a potenttumor suppressor in ARMS Inhibition of CAV1 function cancontribute to aberrant cell proliferation leading to ARMSdevelopment [80]

49 DNA Repairing System Some studies indicate that RMSis characterized by germline mutations of DNA repair genes[82 83] DNA repair gene alterations in RMS occur sec-ondarily to malignant transformation The modificationsin DNA repair enzyme activity can result in resistance toadjuvant treatment in RMS tumor cells which limits theprognosis of RMS There are two pathways for repairingDNA breaks directly without affecting DNA structure andindirectly by DNA phosphodiester backbone cleavage [82](Figure 4) Direct repair includes repair during replicationand enzymatic repair Indirect repair is comprised of exci-sion repair systems including base excision repair (BER)nucleotide excision repair (NER) mismatch repair (MMR)and recombination repair (RR) [83] Defective DNA repairmechanisms result from point mutations or LOH in RMSand contribute to tumorigenesis MGMT and MMR proteinactivity and expression levels are used as predictor indices fortherapy outcome in RMS BER as the major DNA repair sys-tem against damage resulting from cellular metabolism canreverse the cytotoxic effects of alkylation agents used suchas antineoplastics subsequent tumor progression and deathimplicates an RMS mechanism of resistance to chemother-apeutic agents This may provide novel individualized ther-apeutics targeting downregulation of activated DNA repairenzymes or upregulation of DNA repair deficiencies [82]

5 MicroRNA (miRNA miR)Expression in RMS

Muscle-specific and ubiquitously expressed miRs appeardownregulated in RMS tumors and cell lines compared withthe normal counterparts These miRs often play crucial rolesas antioncogenes Inhibition of these miRs contributes toenhanced tumorigenesis through the modulation of diversemolecular pathways Upregulation of prooncogenic miRs hasbeen detected in RMS recently as well [84] (Table 4)

51 Antioncogenic miR in RMS Gain-of-function experimentshave demonstrated that reexpression of selected ldquotumor sup-pressorrdquo miRs impairs the tumorigenic behavior of RMS cells[84 85] As myo-miR family members miR-1 miR-133a andmiR-206 have been demonstrated to be downregulated inRMS and shown to have activity on mRNA expression bytargeting c-Met [42 43 86] Inhibition of thesemyo-miRNAsmay cause aberrant cell proliferation and migration in myo-genesis leading to RMS development especially for ERMSThere have been more miRs found as tumor suppressors inthis malignancy including miR-29 miR-450b-5p miR-203

8 Sarcoma

Table 4 MicroRNAs involved in myogenesis and RMS development

miRNA Expression Target Function Reference

miR-1 and miR-133a Downregulation MYH9Myogenic miRNA inhibit differentiationand promote proliferation in myogenesiscytostatic

Rao et al 2010 [42]

miR-206 Downregulation cMET Promote differentiation and proliferationin myogenesis

Yan et al 2009 [43]

miR-29 Downregulation HADC4 YY1 EZH2 Promote stabilization of RMS phenotype Marchesi et al 2014 [44]

miR-450-5p Downregulation ENOX PAX9 Promote differentiation and progression Sun et al 2014 [45]

miR-203 Downregulation JAK STAT Notch Inhibit differentiation and proliferation inmyogenesis tumor suppressor

Diao et al 2014 [46]

miR-214 Downregulation N-Ras Inhibit tumor cell growth and inducemyogenic differentiation and apoptosis

Huang et al 2014 [47]

miR-183 Upregulation EGR1 Promote migration and metastasis Sarver et al 2010 [48]

PMS2

MGMT

HR

Mismatch Recombination

NHEJ MMEJ

p53

Excision

BER NER

RMS

DNA repair

Direct repair Indirect repair

repair repairrepair

Figure 4The DNA repair systems in RMSThere are two pathways repairing the DNA lesions directly without affecting DNA structure andindirectly by DNA phosphodiester backbone cleavage The modifications in DNA repair enzymes expression or activity lead to resistanceto chemotherapy and radiation in RMS tumor cells MGMT O6-methylguanine-DNA methyltransferase BER base excision repair NERnucleotide excision repair HR homologous recombination NHEJ nonhomologous end-joining MMEJ microhomology-mediated end-joining

and miR-214 [44 46 47 84 87] They are all significantlydownregulated in RMS compared with normal skeletal mus-cles and function to inhibit tumor cell growth and inducemyogenic differentiation and apoptosis in RMS

52 Oncogenic miRNA in RMS The transcription factor EGR1is a tumor suppressor gene that is downregulated in RMS [48]miR-183 functions as an oncogene by targeting EGR1 and pro-moting tumor cell migration Either by direct anti-miR treat-ment or by indirect mechanisms that decrease transcriptmiR-183 targeted treatmentsmight provide a potential optionto RMS patients

The basic strategy of current effective miR-based treat-ment studies is either efficient reexpression of miRs or restor-ing the function of miRs to inhibit the expression of certain

protein-coding genes and promote muscle differentiationMoreover miR expression profiling in tumors and possiblytheir detection in peripheral blood during treatment may beable to predict the response to chemotherapy or radiotherapyand be useful as a prognostic signature for the development oftreatment resistanceThe prognostic value of miR expressionlevels in RMS is a powerful tool in creating a better-tailoredstrategy for particular subsets of patients

53 Long Noncoding RNAs in RMS Long noncoding RNAs(LNCRNAs) are abundant in the mammalian transcriptomeand have been shown to play a role in RMS development [8889]TheH19 gene localized within a chromosomal region onhuman chromosome 11p15 encodes an imprinted untrans-lated RNA [90] H19 opposite tumor suppressor (HOTS)

Sarcoma 9

Table 5 In vivo animal models of RMS

Model TargetModel origin Reference

Inactivation Expression

DrosophilaPAX-FKHR Bipartite Gal4-UAS

expression systemGalindo et al2006 [49]

PAX7-FOXO1 rols Chromosomal deletionDf(3L)vin5

Avirneni-Vadlamudi et al2012 [50]

Zebrafishrag2 promoter c-Myc kRASG12D

Chen andLangenau 2011[51]

MAPKERK andAKTS6K1 PD98059 TPCK Tg(hsp70-HRASG12V) Le et al 2012

[52]

Mouse

Sufu N-myc Sfrp1 Ptch2and cyclin D1 Sufu+minus Lee et al 2007

[53]

Ink4aArf and Trp53 Pax3FKHR Pax3P3Fpwt Keller et al2004 [54]

Wnt120573-cateninsignaling pathway Wnt2 p53minusminusc-fosminusminus Singh et al 2010

[55]

a tumor growth inhibitor is encoded by an imprinted H19antisense transcript Overexpression of HOTS inhibits RMStumor cell growth Silencing HOTS by RNAi was shown toincrease in vitro colony formation as well as in vivo tumorgrowth of RMS [90]

6 In Vivo Models of RMS

Several in vivo models have recently been developed inDrosophila zebrafish and mice to further understand themolecular and cellular biology of RMS tumorigenesis [49ndash55 91ndash94] (Table 5)

61 Drosophila Models in RMS Despite the evolutionarydistance a strong conservation of genes pathways andregulatory molecular networks have been demonstratedbetween flies and humans Many human disease genes haverelated sequences in Drosophila In contrast to mammalianmodels the generation of Drosophila mutants is easy cheapand fast

For a better understanding of the pathogenic conse-quences of PAX-FKHR expression Drosophila was chosenbecause the animalrsquos transparent outer cuticle allows forsimple real-time monitoring of muscle abnormalities elicitedby PAX-FKHR including subtle or focal changes [49] Themutation in the myoblast fusion gene rolling pebbles (rols)was reported to dominantly suppress PAX-FKHR1-inducedlethality by using a Drosophila model of PAX-FKHR1-mediated transformation [50] PAX-FKHR1 signaling upreg-ulates the TANC1 gene and blocks myoblasts from terminaldifferentiation However downregulating the TANC1 genecould cause RMS cells to lose their neoplastic state undergofusion and form differentiation This novel finding uncov-ered a PAX-FKHR1-TANC1 neoplasia axis collaboratively ina Drosophila model and loss-gain-of-function studies inmammalian platforms [50]

62 Zebrafish Models in RMS In contrast to other mod-els the short tumor onset in zebrafish allows for rapididentification of essential genes in various processes suchas tumor growth self-renewal and maintenance There areother advantages for using zebrafish in the study of humandisease and development including the ease and low cost ofraising large numbers of fish and the highly conserved geneticand biochemical pathways between zebrafish and mammals[95]

As zebrafish RMS is highly similar to human ERMSby using fluorescent transgenic approaches it is more con-venient to understand histogenesis and different aspects oftumorigenesis in RMS A robust zebrafish model of RAS-induced RMS has been established sharing twomorphologicand immunophenotypic features resembling ERMS [51] Inone case both are associated with tissue-restricted geneexpression in RMS while in another both comprise a RAS-induced gene signature Cross-species microarray compar-isons confirm that conserved genetic tumor-specific andtissue-restricted pathways such as RAS and p53 pathwaysdrive RMS growth Tg(hsp70-HRASG12V) zebrafish embryoswere generated to evaluate gene expression that mimics RASpathway activation during tumorigenesis to defineRAS targetgenes [52] Another KRASG12D-induced zebrafish embryonalRMS was utilized to assess the therapeutic effects By theblockage of two major downstream signaling pathwaysMAPKERK and AKTS6K1 the model showed that inhibi-tion of translation initiation suppresses tumor cell prolifera-tion [52]

All the above zebrafish cancer models share similar cellu-lar features and molecular pathways with human RMS espe-cially ERMS and can demonstrate a response to therapiesin a similar manner with human RMS Thus a tremendousopportunity is available to implement these animal modelsinto different steps for novel targeted therapeutic develop-ment

10 Sarcoma

63 Mouse Models in RMS Mouse models established fromin vivo studies may contribute to the understanding of thegenetic basis in tumor development and progression andmayhelp to test the efficacies of novel antineoplastic agents

The importance of PAX3PAX7-FKHR fusion proteins inthe progression of ARMS tumorigenesis is evident from bothin vitro and in vivo studies Transgenic mouse models gener-ated with the PAX-FKHR fusion genemimic the formation ofhuman ARMS [67] A mouse model was developed with het-erozygous loss of Sufu a tumor suppressor inHedgehog (Hh)signaling combined with p53 loss driven ERMS with a lowpenetrance (9) [53] These studies indicate that the modelsconsistently formingARMSnot only need the introduction ofthe PAX-FKHR fusion gene but also need to be accompaniedby other genetic events such as p53 pathway disruption [54]

Compared with ARMS ERMS models are more complexto generate because of their lower tumor penetrance andlonger latency JW41 cells from the p53c-fos double mutantmice models resemble human ERMS cells morphologicallyand express similar characteristic markers [55] The overex-pression of the Wnt2 gene identified in JW41 cells confirmedin human RMS cell lines can allow for further analysis of theWnt signaling pathway Both of these results indicate that thedownregulation of theWnt pathway contributes to resistanceto apoptosis and the inhibition of myogenic differentiation inERMS

Using Ptch1 p53 andor Rb1 conditional mouse modelsand controlling prenatal or postnatal myogenic cell of originRubin et al demonstrated that the loss of p53 in maturingmyoblasts related to tumorigenesis of ERMS [96] They alsoindicated that Rb1 alteration with other oncogenic factorswas strongly associated with an undifferentiated phenotypeacting as a modifier In addition they highlighted a sub-set of p53-deficient ERMSs arising from Myf6-expressingmyoblasts which had the same latency with Pax7-expressingmurine satellite cells in the study

7 Potential Therapeutic Targets in RMS

Detection of genomic imbalances and identification of thecrucial effective genes can contribute to identifying novelpotential biomarkers and relevant targets for clinical therapyin RMS

Survivin-responsive conditionally replicating adenovi-ruses can regulate multiple factors (Survm-CRAs) [41]Although Survm-CRA can efficiently replicate and potentlyinduce cell death in RMS cells the cytotoxic effects are morepronounced in RSC-enriched or RSC-purified cells thanin RSC-exiguous or progeny-purified cells Injections ofSurvm-CRAs into tumor nodules generated by transplantingRSC-enriched cells induce significant death in RMS cells andregression of tumor nodulesThe unique therapeutic featuresof Survm-CRA include not only its therapeutic effectivenessagainst RMS but also its increasing effectiveness against CSCsuggesting that Survm-CRA may be a promising anticanceragent [41]

Rapamycin an inhibitor of mTOR can abrogate RMStumor growth in a xenograft mouse model [97] Thetumors in the rapamycin-treatedmice group show significant

reduction of proliferation and invasiveness and inductionof apoptosis The tumor growth inhibition is simultaneousassociated with the diminution of mTOR and Hh pathwayswhich are implicated in the pathogenesis of RMSThe resultsindicate that using rapamycin either alone or in combinationwith traditional chemotherapeutic drugs may represent apotential targeted agent for therapeutic intervention Ampli-fication and mutational activation of Fibroblast Growth Fac-tor Receptor 4 (FGFR4) in RMS cells promote tumor pro-gression [98] Ponatinib is the most potent FGFR4 inhibitorto block wild-type RMS cell growth mutate FGFR4 throughincreasing apoptosis and suppress the FGFR4 downstreamtarget STAT3 Ponatinib treatment slows tumor growth inRMS mouse models expressing mutated FGFR4 [98] Pona-tinib as an FGFR4 inhibitor shows potential to act as aneffective therapeutic agent for RMS tumorsThe expression ofCXC chemokine receptor 4 (CXCR4) CXCR7 and VascularEndothelial Growth Factor (VEGF) indicates poor prognosisin a variety of malignancies including RMS cell lines Mostsamples in a large series of clinical RMS cases show asso-ciation with high expression of these antagonists regardlessof their subtypes [99] However there are significant corre-lations with high expression of CXCR4 and VEGF in bothERMS and ARMS High VEGF expression is predictive ofadverse prognostic factors in RMS as the expression of theseangiogenesis factors was compared with clinicopathologicalparameters and prognosis Considering their overexpressionin RMS these chemokine receptors and VEGF could providepotential molecular therapeutic targets in RMS Some ARMScell lines have undergone apoptosis in response to antineo-plastic drugs such as bortezomib The proapoptotic BH3only family member NOXA acts as an upregulated proteindownstream PAX3-FKHR resulting in increased cell sensi-tivity to bortezomib Apoptosis in response to bortezomibcan be reversed by shRNA knockdown of Noxa PAX3-FKHR upregulation of Noxa creates a potential therapeuticinsight into inducing apoptosis in ARMS cellsThis apoptosispathway could represent a specific targeted therapy againstPAX3-FKHR-expressing ARMS cells [100]

Proton pump activation resulting from intracellular acid-ificationmay be an energetic mechanism to escape apoptosisV-ATPase is an ATP-driven proton pump acidifying theintracellular compartment and transports protons across theplasma membrane Esomeprazole as a V-ATPase inhibitor inthe PPI administration interferes with intensive ion trans-porter activity of RMS cells and induces remarkable cytotoxi-city RMS expresses a higher level ofV-ATPase comparedwithother sarcomasTherefore RMS should be very susceptible toesomeprazole treatment and V-ATPase could be consideredas a promising selective target for treatment [101]

8 Conclusion and Future Prospects

For RMS the current challenges include how to iden-tify promising novel therapeutics and integrate them intoexisting therapy For better understanding of congenitaland epigenetic modifications in the development of RMSthis review summarizes the recent genome-wide studies onmolecular and genetic alterations to decipher the underlying

Sarcoma 11

tumorigenesis mechanisms There is a link between geneticand epigenetic alterations responsible for a variety of tumorcell growth proliferation differentiation apoptosis andtherapy-resistance mechanisms The identification of theprognostic value of the PAX-FKHR fusion status in RMSis one of the most important shifts in the lineage and therisk assessment of RMS which exerts an oncogenic effectthrough multiple pathways and is incorporated into mostrelevant studies The misbalanced expression of a number ofmiRs involved in the regulation of myogenic differentiationalso implicates the tumorsrsquo escape suppressive mechanismsMoreover we described several in vivo models of RMSto explore underlying histogenesis and different aspects oftumorigenesis Recent work on CSCs has also contributedto understanding the cell origins refining molecular char-acterization and elucidating the underlying basis of refrac-tory responses to current adjuvant treatments of subsets oftumorsThese integrative approaches can provide opportuni-ties to identify diagnostic and prognostic biomarkers appliedto the individual targeted therapy and significantly improveRMS prognosis

Abbreviations

RMS RhabdomyosarcomaARMS Alveolar rhabdomyosarcomaERMS Embryonal rhabdomyosarcomaPRMS Pleomorphic rhabdomyosarcomaSRMS Sclerosingspindle cell

rhabdomyosarcomamiRNA miR MicroRNACSC Cancer stem cellMSC Mesenchymal stem cell

Conflict of Interests

The authors declare that there is no conflict of interestsregarding the publication of this paper

Acknowledgments

This work was supported in part by grants from the Gat-tegno and Wechsler funds Dr Duan was supported in partthrough a grant from Sarcoma Foundation of America (SFA)a grant from National Cancer Institute (NCI)National Insti-tutes of Health (NIH) UO1 CA 151452 and a grant from anAcademic Enrichment Fund of MGH Orthopedic Surgery

References

[1] L Yang T Takimoto and J Fujimoto ldquoPrognostic model forpredicting overall survival in children and adolescents withrhabdomyosarcomardquo BMC Cancer vol 14 no 1 article 6542014

[2] D Egas-Bejar and W W Huh ldquoRhabdomyosarcoma in adoles-cent and young adult patients current perspectivesrdquo AdolescentHealth Medicine andTherapeutics vol 5 pp 115ndash125 2014

[3] J C Breneman E Lyden A S Pappo et al ldquoPrognosticfactors and clinical outcomes in children and adolescents withmetastatic rhabdomyosarcomamdasha report from the intergrouprhabdomyosarcoma study IVrdquo Journal of Clinical Oncology vol21 no 1 pp 78ndash84 2003

[4] V D Soleimani and M A Rudnicki ldquoNew insights into theorigin and the genetic basis of rhabdomyosarcomasrdquo CancerCell vol 19 no 2 pp 157ndash159 2011

[5] T Glass D D Cochrane S R Rassekh K Goddard and JHukin ldquoGrowing teratoma syndrome in intracranial non-ger-minomatous germ cell tumors (iNGGCTs) a risk for secondarymalignant transformationmdasha report of two casesrdquo Childrsquos Ner-vous System vol 30 no 5 pp 953ndash957 2014

[6] J Ji C Eng and K Hemminki ldquoFamilial risk for soft tissuetumors a nation-wide epidemiological study from SwedenrdquoJournal of Cancer Research and Clinical Oncology vol 134 no5 pp 617ndash624 2008

[7] C D M Fletcher J A Bridge P Hogendoorn and F MertensWHO Classification of Tumours of Soft Tissue and Bone vol 5IARC Press Paris France 4th edition 2013

[8] I Sultan I Qaddoumi S Yaser C Rodriguez-Galindo and AFerrari ldquoComparing adult and pediatric rhabdomyosarcoma inthe surveillance epidemiology and end results program 1973 to2005 an analysis of 2600 patientsrdquo Journal of Clinical Oncologyvol 27 no 20 pp 3391ndash3397 2009

[9] B Raney W Huh D Hawkins et al ldquoOutcome of patientswith localized orbital sarcoma who relapsed following treat-ment on Intergroup Rhabdomyosarcoma Study Group (IRSG)Protocols-III and -IV 1984ndash1997 a report from the ChildrenrsquosOncology Grouprdquo Pediatric Blood and Cancer vol 60 no 3 pp371ndash376 2013

[10] W Crist E A Gehan A H Ragab et al ldquoThe third intergrouprhabdomyosarcoma studyrdquo Journal of Clinical Oncology vol 13no 3 pp 610ndash630 1995

[11] C Liu D Li J Jiang et al ldquoAnalysis of molecular cytogeneticalteration in rhabdomyosarcoma by array comparative genomichybridizationrdquo PLoS ONE vol 9 no 4 Article ID e94924 2014

[12] DWilliamson E Missiaglia A de Reynies et al ldquoFusion gene-negative alveolar rhabdomyosarcoma is clinically and molec-ularly indistinguishable from embryonal rhabdomyosarcomardquoJournal of Clinical Oncology vol 28 no 13 pp 2151ndash2158 2010

[13] V Paulson G Chandler D Rakheja et al ldquoHigh-resolutionarray CGH identifies common mechanisms that drive embry-onal rhabdomyosarcoma pathogenesisrdquo Genes Chromosomesand Cancer vol 50 no 6 pp 397ndash408 2011

[14] S Park J Lee I-G Do et al ldquoAberrant CDK4 amplificationin refractory rhabdomyosarcoma as identified by genomicprofilingrdquo Scientific Reports vol 4 article 3623 2014

[15] C Liu D Li J Hu et al ldquoChromosomal and genetic imbalancesin Chinese patients with rhabdomyosarcoma detected by high-resolution array comparative genomic hybridizationrdquo Interna-tional Journal of Clinical and Experimental Pathology vol 7 no2 pp 690ndash698 2014

[16] Q-X Li C-X Liu C-P Chun et al ldquoChromosomal imbalancesrevealed in primary rhabdomyo-sarcomas by comparativegenomic hybridizationrdquo Chinese Medical Journal vol 122 no11 pp 1277ndash1282 2009

[17] E Missiaglia J Selfe M Hamdi et al ldquoGenomic imbalancesin rhabdomyosarcoma cell lines affect expression of genesfrequently altered in primary tumors an approach to identifycandidate genes involved in tumor developmentrdquo Genes Chro-mosomes and Cancer vol 48 no 6 pp 455ndash467 2009

12 Sarcoma

[18] F G Barr F Duan L M Smith et al ldquoGenomic and clinicalanalyses of 2p24 and 12q13-q14 amplification in alveolar rhab-domyosarcoma a report from the childrenrsquos oncology grouprdquoGenes Chromosomes and Cancer vol 48 no 8 pp 661ndash6722009

[19] T Gordon AMcManus J Anderson et al ldquoCytogenetic abnor-malities in 42 rhabdomyosarcoma a United Kingdom CancerCytogenetics Group StudyrdquoMedical and Pediatric Oncology vol36 no 2 pp 259ndash267 2001

[20] X Chen E Stewart A A Shelat et al ldquoTargeting oxidativestress in embryonal rhabdomyosarcomardquo Cancer Cell vol 24no 6 pp 710ndash724 2013

[21] S Lizard-Nacol F Mugneret C Volk C Turc-Carel M Favrotand T Philip ldquoTranslocation (213)(q37q14) in alveolar rhab-domyosarcoma a new caserdquo Cancer Genetics and Cytogeneticsvol 25 no 2 pp 373ndash374 1987

[22] P H B Sorensen J C Lynch S J Qualman et al ldquoPAX3-FKHR and PAX7-FKHR gene fusions are prognostic indicatorsin alveolar rhabdomyosarcoma a report from the childrenrsquosoncology grouprdquo Journal of Clinical Oncology vol 20 no 11 pp2672ndash2679 2002

[23] A D Marshall and G C Grosveld ldquoAlveolar rhabdomyo-sarcomamdashthe molecular drivers of PAX37-FOXO1-inducedtumorigenesisrdquo Skeletal Muscle vol 2 no 1 article 25 2012

[24] E Missiaglia D Williamson J Chisholm et al ldquoPAX3FOXO1fusion gene status is the key prognostic molecular markerin rhabdomyosarcoma and significantly improves current riskstratificationrdquo Journal of Clinical Oncology vol 30 no 14 pp1670ndash1677 2012

[25] J F Shern L Chen J Chmielecki et al ldquoComprehensivegenomic analysis of rhabdomyosarcoma reveals a landscape ofalterations affecting a common genetic axis in fusion-positiveand fusion-negative tumorsrdquo Cancer Discovery vol 4 no 2 pp216ndash231 2014

[26] MWachtel M Dettling E Koscielniak et al ldquoGene expressionsignatures identify rhabdomyosarcoma subtypes and detect anovel t(22)(q35p23) translocation fusing PAX3 to NCOA1rdquoCancer Research vol 64 no 16 pp 5539ndash5545 2004

[27] E Davicioni M J Anderson F G Finckenstein et al ldquoMolec-ular classification of rhabdomyosarcomamdashgenotypic and phe-notypic determinants of diagnosis a report from the ChildrenrsquosOncology Grouprdquo The American Journal of Pathology vol 174no 2 pp 550ndash564 2009

[28] M Visser C Sijmons J Bras et al ldquoAllelotype of pediatric rhab-domyosarcomardquo Oncogene vol 15 no 11 pp 1309ndash1314 1997

[29] R Gil-Benso T San-Miguel R C Callaghan et al ldquoChromo-somal and genetic changes produced in tumoral progression ofembryonal rhabdomyosarcomardquo Histopathology vol 62 no 5pp 816ndash819 2013

[30] S X Skapek J Anderson F G Barr et al ldquoPAX-FOXO1 fusionstatus drives unfavorable outcome for children with rhabdom-yosarcoma a childrenrsquos oncology group reportrdquo Pediatric Bloodand Cancer vol 60 no 9 pp 1411ndash1417 2013

[31] HHosoi N Kakazu E Konishi et al ldquoA novel PAX3 rearrange-ment in embryonal rhabdomyosarcomardquo Cancer Genetics andCytogenetics vol 189 no 2 pp 98ndash104 2009

[32] E Takaoka H Sonobe K Akimaru et al ldquoMultiple sites ofhighly amplified DNA sequences detected bymolecular cytoge-netic analysis inHS-RMS-2 a newpleomorphic rhabdomyosar-coma cell linerdquo American Journal of Cancer Research vol 2 no2 pp 141ndash152 2012

[33] K Szuhai D de Jong W Y Leung C D M Fletcher and P CWHogendoorn ldquoTransactivatingmutation of theMYOD1 geneis a frequent event in adult spindle cell rhabdomyosarcomardquoJournal of Pathology vol 232 no 3 pp 300ndash307 2014

[34] N P Agaram C-L Chen L Zhang M P Laquaglia L Wexlerand C R Antonescu ldquoRecurrent MYOD1 mutations in pedi-atric and adult sclerosing and spindle cell rhabdomyosarcomasEvidence for a common pathogenesisrdquoGenes Chromosomes andCancer vol 53 no 9 pp 779ndash787 2014

[35] H Yoshida M Miyachi K Sakamoto et al ldquoPAX3-NCOA2fusion gene has a dual role in promoting the proliferation andinhibiting the myogenic differentiation of rhabdomyosarcomacellsrdquo Oncogene vol 33 pp 5601ndash5608 2013

[36] J M Mosquera A Sboner L Zhang et al ldquoRecurrent NCOA2gene rearrangements in congenitalinfantile spindle cell rhab-domyosarcomardquo Genes Chromosomes and Cancer vol 52 no6 pp 538ndash550 2013

[37] V Thavaraj A Sethi and L S Arya ldquoIncomplete Beckwith-Wiedemann syndrome in a child with orbital rhabdomyosar-comardquo Indian Pediatrics vol 39 no 3 pp 299ndash304 2002

[38] D Walter S Satheesha P Albrecht et al ldquoCD133 positiveembryonal rhabdomyosarcoma stem-like cell population isenriched in rhabdospheresrdquo PLoS ONE vol 6 no 5 Article IDe19506 2011

[39] Y-X Ren F G Finckenstein D A Abdueva et al ldquoMousemesenchymal stem cells expressing PAX-FKHR form alveolarrhabdomyosarcomas by cooperating with secondary muta-tionsrdquo Cancer Research vol 68 no 16 pp 6587ndash6597 2008

[40] M Salerno S Avnet G Bonuccelli S Hosogi D Granchi andN Baldini ldquoImpairment of lysosomal activity as a therapeuticmodality targeting cancer stem cells of embryonal rhabdom-yosarcoma cell line RDrdquo PLoS ONE vol 9 no 10 Article IDe110340 2014

[41] K Tanoue Y Wang M Ikeda et al ldquoSurvivin-responsive con-ditionally replicating adenovirus kills rhabdomyosarcoma stemcells more efficiently than their progenyrdquo Journal of Transla-tional Medicine vol 12 no 1 article 27 2014

[42] P K Rao EMissiaglia L Shields et al ldquoDistinct roles formiR-1and miR-133a in the proliferation and differentiation of rhab-domyosarcoma cellsrdquo The FASEB Journal vol 24 no 9 pp3427ndash3437 2010

[43] D Yan X D Dong X Chen et al ldquoMicroRNA-1206 targets c-met and inhibits rhabdomyosarcomadevelopmentrdquoThe Journalof Biological Chemistry vol 284 no 43 pp 29596ndash29604 2009

[44] I Marchesi A Giordano and L Bagella ldquoRoles of enhancerof zeste homolog 2 from skeletal muscle differentiation torhabdomyosarcoma carcinogenesisrdquoCell Cycle vol 13 no 4 pp516ndash527 2014

[45] M M Sun J F Li L L Guo et al ldquoTGF-1205731 suppression ofmicroRNA-450b-5p expression a novel mechanism for block-ingmyogenic differentiation of rhabdomyosarcomardquoOncogenevol 33 no 16 pp 2075ndash2086 2014

[46] Y Diao X Guo L Jiang et al ldquoMiR-203 a tumor suppressorfrequently down-regulated by promoter hypermethylation inrhabdomyosarcomardquo The Journal of Biological Chemistry vol289 no 1 pp 529ndash539 2014

[47] H-J Huang J Liu HHua et al ldquoMiR-214 andN-ras regulatoryloop suppresses rhabdomyosarcoma cell growth and xenografttumorigenesisrdquo Oncotarget vol 5 no 8 pp 2161ndash2175 2014

Sarcoma 13

[48] A L Sarver L Li and S Subramanian ldquomicroRNA miR-183functions as an oncogene by targeting the transcription factorEGR1 and promoting tumor cell migrationrdquo Cancer Researchvol 70 no 23 pp 9570ndash9580 2010

[49] R L Galindo J A Allport and E N Olson ldquoA Drosophilamodel of the rhabdomyosarcoma initiator PAX7-FKHRrdquo Pro-ceedings of the National Academy of Sciences of the United Statesof America vol 103 no 36 pp 13439ndash13444 2006

[50] U Avirneni-Vadlamudi K A Galindo T R Endicott V Paul-son S Cameron and R L Galindo ldquoDrosophila and mammal-ian models uncover a role for the myoblast fusion gene TANC1in rhabdomyosarcomardquo The Journal of Clinical Investigationvol 122 no 1 pp 403ndash407 2012

[51] E Y Chen andDM Langenau ldquoZebrafishmodels of rhabdom-yosarcomardquoMethods in Cell Biology vol 105 pp 383ndash402 2011

[52] X Le E K Pugach S Hettmer et al ldquoA novel chemicalscreening strategy in zebrafish identifies common pathways inembryogenesis and rhabdomyosarcoma developmentrdquo Devel-opment vol 140 no 11 pp 2354ndash2364 2012

[53] Y Lee R Kawagoe K Sasai et al ldquoLoss of suppressor-of-fusedfunction promotes tumorigenesisrdquoOncogene vol 26 no 44 pp6442ndash6447 2007

[54] C Keller B R Arenkiel C M Coffin N El-Bardeesy R ADePinho and M R Capecchi ldquoAlveolar rhabdomyosarcomasin conditional Pax3Fkhr mice cooperativity of Ink4aARF andTrp53 loss of functionrdquo Genes and Development vol 18 no 21pp 2614ndash2626 2004

[55] S Singh CVinson CMGurley et al ldquoImpairedWnt signalingin embryonal rhabdomyosarcoma cells from p53c-fos doublemutant micerdquo American Journal of Pathology vol 177 no 4 pp2055ndash2066 2010

[56] G J Schaaf J M Ruijter F Van Ruissen et al ldquoFull transcrip-tome analysis of rhabdomyosarcoma normal and fetal skeletalmuscle statistical comparison of multiple SAGE librariesrdquo TheFASEB Journal vol 19 no 3 pp 404ndash406 2005

[57] D S Hawkins A A Gupta and E R Rudzinski ldquoWhat is newin the biology and treatment of pediatric rhabdomyosarcomardquoCurrent Opinion in Pediatrics vol 26 no 1 pp 50ndash56 2014

[58] M Zhang J Truscott and J Davie ldquoLoss of MEF2D expressioninhibits differentiation and contributes to oncogenesis in rhab-domyosarcoma cellsrdquoMolecular Cancer vol 12 no 1 article 1502013

[59] M Ciesla J Dulak and A Jozkowicz ldquomicroRNAs and epi-genetic mechanisms of rhabdomyosarcoma developmentrdquo TheInternational Journal of Biochemistry amp Cell Biology vol 53 pp482ndash492 2014

[60] S Ricaud B Vernus M Duclos et al ldquoInhibition of autocrinesecretion of myostatin enhances terminal differentiation inhuman rhabdomyosarcoma cellsrdquo Oncogene vol 22 no 51 pp8221ndash8232 2003

[61] C S Guo C Degnin T A Fiddler D Stauffer andM JThayerldquoRegulation of MyoD activity and muscle cell differentiation byMDM2 pRb and Sp1rdquoThe Journal of Biological Chemistry vol278 no 25 pp 22615ndash22622 2003

[62] S Rossi E Stoppani P L Puri and A Fanzani ldquoDifferenti-ation of human rhabdomyosarcoma RD cells is regulated byreciprocal functional interactions between myostatin p38 andextracellular regulated kinase signalling pathwaysrdquo EuropeanJournal of Cancer vol 47 no 7 pp 1095ndash1105 2011

[63] Z S Walters B Villarejo-Balcells D Olmos et al ldquoJARID2is a direct target of the PAX3-FOXO1 fusion protein andinhibits myogenic differentiation of rhabdomyosarcoma cellsrdquoOncogene vol 33 no 9 pp 1148ndash1157 2014

[64] T Hosoyama M I Aslam J Abraham et al ldquoIL-4R drives ded-ifferentiation mitogenesis and metastasis in rhabdomyosar-comardquo Clinical Cancer Research vol 17 no 9 pp 2757ndash27662011

[65] F S Dela Cruz ldquoCancer stem cells in pediatric sarcomasrdquoFrontiers in Oncology vol 3 article 168 2013

[66] M J Bissell and W C Hines ldquoWhy donrsquot we get more cancerA proposed role of the microenvironment in restraining cancerprogressionrdquo Nature Medicine vol 17 no 3 pp 320ndash329 2011

[67] E Charytonowicz C Cordon-Cardo I Matushansky and MZiman ldquoAlveolar rhabdomyosarcoma is the cell of origin amesenchymal stem cellrdquoCancer Letters vol 279 no 2 pp 126ndash136 2009

[68] W K Cavenee ldquoMuscling in on rhabdomyosarcomardquo NatureMedicine vol 8 no 11 pp 1200ndash1201 2002

[69] B Zhu and J K Davie ldquoNew insights into signalling-pathwayalterations in rhabdomyosarcomardquo British Journal of Cancervol 112 no 2 pp 227ndash231 2015

[70] M Zhang C M Linardic and D G Kirsch ldquoRAS and ROS inrhabdomyosarcomardquo Cancer Cell vol 24 no 6 pp 689ndash6912013

[71] S C Marianna L Ianzano and G Nicoletti ldquoTumor sup-pressor genes promote rhabdomyosarcoma progression in p53heterozygousrdquo Oncotarget vol 5 no 1 pp 108ndash119 2013

[72] SWang L Guo L Dong et al ldquoTGF-beta1 signal pathwaymaycontribute to rhabdomyosarcoma development by inhibitingdifferentiationrdquo Cancer Science vol 101 no 5 pp 1108ndash11162010

[73] J Wesche K Haglund and E M Haugsten ldquoFibroblast growthfactors and their receptors in cancerrdquo Biochemical Journal vol437 no 2 pp 199ndash213 2011

[74] M Wachtel J Rakic M Okoniewski P Bode F Niggli andB W Schafer ldquoFGFR4 signaling couples to Bim and not Bmfto discriminate subsets of alveolar rhabdomyosarcoma cellsrdquoInternational Journal of Cancer vol 135 no 7 pp 1543ndash15522014

[75] P L Puri ZWu P Zhang et al ldquoInduction of terminal differen-tiation by constitutive activation of p38 MAP kinase in humanrhabdomyosarcoma cellsrdquo Genes and Development vol 14 no5 pp 574ndash584 2000

[76] A M Tremblay E Missiaglia G G Galli et al ldquoThe Hippotransducer YAP1 transforms activated satellite cells and is apotent effector of embryonal rhabdomyosarcoma formationrdquoCancer Cell vol 26 no 2 pp 273ndash287 2014

[77] R N Judson A M Tremblay P Knopp et al ldquoThe hippo path-way member Yap plays a key role in influencing fate decisionsin muscle satellite cellsrdquo Journal of Cell Science vol 125 no 24pp 6009ndash6019 2012

[78] L E S Crose K A Galindo J G Kephart et al ldquoAlve-olar rhabdomyosarcoma-associated PAX3-FOXO1 promotestumorigenesis via Hippo pathway suppressionrdquo Journal ofClinical Investigation vol 124 no 1 pp 285ndash296 2014

[79] B Belyea J G Kephart J Blum D G Kirsch and C MLinardic ldquoEmbryonic signaling pathways and rhabdomyosar-coma contributions to cancer development and opportunitiesfor therapeutic targetingrdquo Sarcoma vol 2012 Article ID 40623913 pages 2012

14 Sarcoma

[80] J Huertas-Martinez S Rello-Varona D Herrero-Martin et alldquoCaveolin-1 is down-regulated in alveolar rhabdomyosarcomasand negatively regulates tumor growthrdquo Oncotarget vol 5 no20 pp 9744ndash9755 2014

[81] S Rossi P L Poliani CMissale EMonti andA Fanzani ldquoCav-eolins in rhabdomyosarcomardquo Journal of Cellular andMolecularMedicine vol 15 no 12 pp 2553ndash2568 2011

[82] P G Tsioli E S Patsouris C Giaginis and S E TheocharisldquoDNA repair systems in rhabdomyosarcomardquo Histology andHistopathology vol 28 no 8 pp 971ndash984 2013

[83] E Gatzidou C Michailidi S Tseleni-Balafouta and S Theo-charis ldquoAn epitome of DNA repair related genes and mecha-nisms in thyroid carcinomardquo Cancer Letters vol 290 no 2 pp139ndash147 2010

[84] R Rota R Ciarapica A Giordano L Miele and F LocatellildquoMicroRNAs in rhabdomyosarcoma pathogenetic implicationsand translational potentialityrdquoMolecular Cancer vol 10 article120 2011

[85] A Gougelet J Perez D Pissaloux et al ldquoMiRNA profiling howto bypass the current difficulties in the diagnosis and treatmentof sarcomasrdquo Sarcoma vol 2011 Article ID 460650 13 pages2011

[86] R Ciarapica G Russo F Verginelli et al ldquoDeregulated expres-sion of miR-26a and Ezh2 in rhabdomyosarcomardquo Cell Cyclevol 8 no 1 pp 172ndash175 2009

[87] L Li A L Sarver S Alamgir and S Subramanian ldquoDownreg-ulation of microRNAs miR-1 -206 and -29 stabilizes PAX3 andCCND2 expression in rhabdomyosarcomardquo Laboratory Investi-gation vol 92 no 4 pp 571ndash583 2012

[88] S-Y Ng G K Bogu B Soh and L W Stanton ldquoThe longnoncoding RNA RMST interacts with SOX2 to regulate neu-rogenesisrdquoMolecular Cell vol 51 no 3 pp 349ndash359 2013

[89] C A Lynch B Tycko T H Bestor and C P Walsh ldquoReac-tivation of a silenced H19 gene in human rhabdomyosarcomaby demethylation of DNA but not by histone hyperacetylationrdquoMolecular Cancer vol 1 article 2 2002

[90] P Onyango and A P Feinberg ldquoA nucleolar protein H19opposite tumor suppressor (HOTS) is a tumor growth inhibitorencoded by a human imprinted H19 antisense transcriptrdquoProceedings of the National Academy of Sciences of the UnitedStates of America vol 108 no 40 pp 16759ndash16764 2011

[91] C E Albacker N Y Storer E M Langdon et al ldquoThe his-tone methyltransferase SUV39H1 suppresses embryonal rhab-domyosarcoma formation in zebrafishrdquo PLoS ONE vol 8 no 5Article ID e64969 2013

[92] N Y Storer R M White A Uong et al ldquoZebrafish rhab-domyosarcoma reflects the developmental stage of oncogeneexpression during myogenesisrdquo Development vol 140 no 14pp 3040ndash3050 2013

[93] M E Hatley W Tang M R Garcia et al ldquoA mouse modelof rhabdomyosarcoma originating from the adipocyte lineagerdquoCancer Cell vol 22 no 4 pp 536ndash546 2012

[94] V Hosur A Kavirayani J Riefler et al ldquoDystrophin and dysfer-lin doublemutantmice a novelmodel for rhabdomyosarcomardquoCancer Genetics vol 205 no 5 pp 232ndash241 2012

[95] W Goessling T E North and L I Zon ldquoNew waves of discov-ery modeling cancer in zebrafishrdquo Journal of Clinical Oncologyvol 25 no 17 pp 2473ndash2479 2007

[96] B P Rubin K Nishijo H-I H Chen et al ldquoEvidence for anunanticipated relationship between undifferentiated pleomor-phic sarcoma and embryonal rhabdomyosarcomardquo Cancer Cellvol 19 no 2 pp 177ndash191 2011

[97] S Z Kaylani J Xu R K Srivastava L Kopelovich J G Presseyand M Athar ldquoRapamycin targeting mTOR and hedgehog sig-naling pathways blocks human rhabdomyosarcoma growth inxenograft murine modelrdquo Biochemical and Biophysical ResearchCommunications vol 435 no 4 pp 557ndash561 2013

[98] S Q Li A T Cheuk J F Shern et al ldquoTargeting wild-type andmutationally activated FGFR4 in rhabdomyosarcoma with theinhibitor ponatinib (AP24534)rdquoPLoSONE vol 8 no 10 ArticleID e76551 2013

[99] K Miyoshi K Kohashi F Fushimi et al ldquoClose correlationbetween CXCR4 and VEGF expression and frequent CXCR7expression in rhabdomyosarcomardquo Human Pathology vol 45no 9 pp 1900ndash1909 2014

[100] ADMarshall F Picchione R I KGeltink andGCGrosveldldquoPAX3-FOXO1 induces up-regulation of Noxa sensitizing alve-olar rhabdomyosarcoma cells to apoptosisrdquo Neoplasia vol 15no 7 pp 738ndash748 2013

[101] F Perut S Avnet C Fotia et al ldquoV-ATPase as an effective thera-peutic target for sarcomasrdquo Experimental Cell Research vol 320no 1 pp 21ndash32 2014

Submit your manuscripts athttpwwwhindawicom

Stem CellsInternational

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

MEDIATORSINFLAMMATION

of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Behavioural Neurology

EndocrinologyInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Disease Markers

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

BioMed Research International

OncologyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Oxidative Medicine and Cellular Longevity

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

PPAR Research

The Scientific World JournalHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Immunology ResearchHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal of

ObesityJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Computational and Mathematical Methods in Medicine

OphthalmologyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Diabetes ResearchJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Research and TreatmentAIDS

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Gastroenterology Research and Practice

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Parkinsonrsquos Disease

Evidence-Based Complementary and Alternative Medicine

Volume 2014Hindawi Publishing Corporationhttpwwwhindawicom

Page 5: Review Article Rhabdomyosarcoma: Advances in Molecular and

Sarcoma 5

Table 3 Studies identifying stem cells in RMS

Markersubstrate Source Stem cell gene Functional characterization Reference

CD133 Orthotopicxenograft model

OCT4 NANOG c-MYCPAX3 and SOX2

Correlating with poor overallsurvival

Walter et al2011 [38]

PAX-FKHR Bone marrow ofC57BL6 mice

Myf5 MEF2 MyoD andmyogenin

Determining the molecularmyogenic and histologicphenotype of ARMS

Ren et al 2008[39]

V-ATPase RD cell line NANOG and OCT34

Driving mechanisms of areduced sensitivity to anticancerdrugs and activities related toinvasion and metastasis

Salerno et al2014 [40]

Survm-CRAsKYM-1 cell line(FGFR3-positive)

FGFR3Therapeutic effectiveness againstall cell populations and increasedeffectiveness against CSCs

Tanoue et al2014 [41]

Migration

specification

Injured muscle

Terminal differentiation

Fusion

PAX7

Myoblasts

Myoblast

MyoD

MyoD

Myf5

PAX3

Rb

Ezh2

IGF2

HDAC4YY1

YY1

ERK

AKT

RMS

Myogenin

P38 kinase

Myostatin

Progenitor

cell

N-RAS

Myofibers

FGFR4

UBE2C

UHRF1

NOTCH2

YWHAB

Satellite cells

RUNX1 MEF2C

JDP2 NFIC

Myocytes

Restoration of proliferation blockade of

differentiation

Proliferatio

n

Cell circle arrestEarlydifferentiation

TGF-1205731

Figure 2 Myogenic pathways in the tumorigenesis of RMS In aberrant neoplastic condition progenitor cells residing in muscle result inaberrant pathways which lead to malignant transformation and fail to differentiate proliferate uncontrollably and form RMS Sharp arrows(rarr) indicate upregulationactivation and blunt arrows (perp) indicate downregulationinhibition

the maturation of myotubes [64] An IL-4R blockade mighthelp therapeutically to modulate the expression of myogenictranscription factors (MYOD or myogenin) [57 64]

32 Potential Cancer Stem Cell in RMS Cancer Stem Cells(CSCs) may contribute to inherent refractory responses tocurrent therapies andmetastasis [65]Many CSCmodels playan important role in the development of RMS [38ndash41 66](Table 3)

A study explored a potential important role for mesen-chymal stem cells (MSCs) as the cell of origin of ARMS [67]Ren et al confirmed that PAX-FKHR fusion genes commitMSCs to a myogenic lineage by inhibiting terminal differen-tiation and contributing to ARMS formation [39] Anotherstudy showed that PAX-FKHR induces skeletal myogenesisin MSCs by transactivating MYOD and myogenin and trans-forms mesenchymal progenitor cells to the skeletal muscle lin-eage leading to malignant formation resembling ARMS [39]

6 Sarcoma

METCDKN2AB

PAX37-FKHR

MDM2

RAS

Raf

GLI1

PDK1p53Rb

Hh

Myo MEF2

p21

Ptch

FGFR4

IGF1R

p16INK4Ap14ARF

p19ARF

IGF2

FGF

Figure 3 Genetic analyses of RMS have pinpointed several common alterations including inactivation of a master regulator of p53 andRb pathways CDKN2AB and activation of FGFR4 RAS and Hedgehog (Hh) signaling The modifications of these pathways influenceoncogenesis and metastatic potential

As a human hematopoietic CSC marker Walter et alproposed that upregulated CD133 in ERMS can act as aprognostic marker and might help with the development ofnovel targeted therapies for ERMS [38]

4 Signaling Pathway Alterations in RMS

Recent studies have looked into signaling pathway alterationsand their downstream effects in RMSThese new findings notonly helped to improve the understanding of this malignancybut also offered novel potential therapeutic strategies forimproved treatment for patientswithRMS [68 69] (Figure 3)

41 RAS Signaling Pathway RASmutations commonlymain-tain the protein in its GTP bound state and therefore renderit constitutively active in RMS Zhang et al identified RASfamily members NRAS KRAS and HRAS in the RASpathway as themost commonlymutated genes in ERMS [70]the mutation status of these genes is significantly associatedwith the risk of ERMS development [70] A study foundthat a majority of ERMS demonstrated activation of the RASpathway exclusively either by homozygous deletion of NF1or by point mutations in one of the RAS family members asdescribed above [13] Despite remarkable genetic and molec-ular heterogeneity most (93 4144) RMS tumors hijackeda common receptor tyrosine kinaseRASPIK3CA geneticaxis [25] It could occur via two alternative mechanismsrearrangement of a PAX gene and accumulation of mutationsthat were downstream targets of the PAX fusion protein

Skeletal muscle cells have a robust antioxidant defensesystem to protect the DNA lipids and proteins from thedeleterious effects of excess Reactive Oxygen Species (ROS)

Cancer cells also have elevated ROS due to their increasedmetabolic activity oncogenic stimulation andmitochondrialdysfunction These findings implicate RAS mutations andoxidative stress as potential therapeutic targets for high-riskERMS

42 IGF Signaling Pathway There is clear preclinical datathat supports the involvement of the Insulin-like GrowthFactor (IGF) signaling pathway in RMS tumorigenesis andprogression [69] Zhu and Davie demonstrated that IGFpromotes the proliferation of RMS cells while blocking IGFsignaling interferes with cell growth in vivoThe IGF receptorIGF-1R is highly overexpressed both on the cell surface andin the nucleus of RMS cells furthermore RMS cell lines aresensitive to IGF-1R inhibitionThe IGF-2 locus shows a loss ofimprinting in both ERMS andARMS tumors and the expres-sion of PAX3-FKHR has been demonstrated to upregulateIGF-2 and activate IGF pathway in ARMS [69] Mariannaet al found that IGF-2 and tumor suppressors p19Arf andp21Cip1 are overexpressed and prominently upregulated inRMS mouse models [71]

43 TGF-120573 Signaling Pathway The regulatory role of Trans-forming Growth Factor-120573 (TGF-120573) in RMS has been recentlystudied [72] Wang et al demonstrated that the expression ofTGF-1205731 is significantly higher in RMS than in normal skeletalmuscle The inhibition of TGF-1205731 expression by shRNA-expressing vectors reverses themalignant behavior of RMSbyinhibiting cell growth and inducingmyogenic differentiationTGF-1205731 shRNA induces myogenin expression a regulator ofmyogenic differentiation genes Myogenin acts as a target fornegative regulation of myogenesis by TGF-1205731 signals with

Sarcoma 7

a differentiation regulatory cascade These results suggestthat the TGF-1205731 signaling pathway disrupts the differentia-tion of myogenic progenitors leading to the development ofRMS

44 FGF Signaling Pathway Fibroblast Growth Factor (FGF)is crucial in embryonic development and functions to driveproliferation FGFs and their receptors (FGFRs) are essentialregulators in the processes of proliferation antiapoptosisdrug resistance and angiogenesis in RMS [69] RMS over-expresses the receptor tyrosine kinase FGFR4 which causesautophosphorylation and constitutive signaling in correlationwith poor differentiation and decreased survival [73] Recentwork has shown that FGF signaling can prevent ARMS cellsfrom apoptosis induced by targeting the IGF1-R-PI3K-mTORpathway [74]

45 ERK Signaling Pathway Previous studies have shownthat elevated myostatin expression deregulates ExtracellularRegulated Kinase (ERK) signaling and deficient activationof the p38 pathway contributes to the differentiation blockin RMS cells [62] The ERK pathway is frequently highlyactivated in RMS cells from embryonic derivation due to thepresence of activating RASmutations leading to reduced dif-ferentiation by blocking the p38 pathway [75]These findingsindicate that interventions that target themyostatinERKp38network could be an effective way to promote differentiationof RMS

46 Hippo Signaling Pathway Many upstream signal trans-duction proteins in response to cues from the cellular micro-environment regulate the core Hippo pathway [76] TheHippo pathway may limit tumorigenesis by inducing thecytosolic localization of the transcriptional cofactor Yes-Associated Protein 1 (YAP1) High YAP levels and activitiesincrease activated satellite cells and prevent differentiationby activating genes that inhibit differentiation [77] YAP1-TEAD1 was found to upregulate proproliferative and onco-genic genes and maintain the ERMS differentiation blockby interfering with MYOD1 and MEF2 prodifferentiationactivities [76] Inhibition of theHippo pathway is exhibited inRMS through downregulation of Hippo pathway tumor sup-pressors or upregulation of YAP [78] These data suggest thatHippo pathway dysfunction promotes RMS development

47 Notch Signaling Pathway and Wnt Signaling PathwayNotch pathway as an embryonic signaling pathway promotesmuscle stem cell maintenance by inhibiting myogenic earlydifferentiation and expanding pools of progenitor cells dur-ing both embryogenesis and postnatal muscle regenerationSimilar to Notch Wnt has various roles in embryonic fetaland neonatalmyogenesis during skeletalmyogenesis As bothpathways are regulators of myogenic lineage determinationand maturation RMS can arise from disordered regulationof these normal developmental pathways [79]

48 Caveolins in RMS Recent findings have shown that cav-eolins are expressed in a cell stage-dependent manner inRMS [80 81] Caveolins are scaffolding proteins that regulate

several targets and pathways (such as p53 IGF RASERK andTGF-120573myostatin signaling) associated with RMS develop-ment and therefore loss or gain of caveolin function is ableto generate multiple effects on the tumor behaviorThereforerecognizing their precise roles in RMS progression will becrucial to elaborate targeted therapies particularly as oneof the three different isoforms CAV1 could act as a potenttumor suppressor in ARMS Inhibition of CAV1 function cancontribute to aberrant cell proliferation leading to ARMSdevelopment [80]

49 DNA Repairing System Some studies indicate that RMSis characterized by germline mutations of DNA repair genes[82 83] DNA repair gene alterations in RMS occur sec-ondarily to malignant transformation The modificationsin DNA repair enzyme activity can result in resistance toadjuvant treatment in RMS tumor cells which limits theprognosis of RMS There are two pathways for repairingDNA breaks directly without affecting DNA structure andindirectly by DNA phosphodiester backbone cleavage [82](Figure 4) Direct repair includes repair during replicationand enzymatic repair Indirect repair is comprised of exci-sion repair systems including base excision repair (BER)nucleotide excision repair (NER) mismatch repair (MMR)and recombination repair (RR) [83] Defective DNA repairmechanisms result from point mutations or LOH in RMSand contribute to tumorigenesis MGMT and MMR proteinactivity and expression levels are used as predictor indices fortherapy outcome in RMS BER as the major DNA repair sys-tem against damage resulting from cellular metabolism canreverse the cytotoxic effects of alkylation agents used suchas antineoplastics subsequent tumor progression and deathimplicates an RMS mechanism of resistance to chemother-apeutic agents This may provide novel individualized ther-apeutics targeting downregulation of activated DNA repairenzymes or upregulation of DNA repair deficiencies [82]

5 MicroRNA (miRNA miR)Expression in RMS

Muscle-specific and ubiquitously expressed miRs appeardownregulated in RMS tumors and cell lines compared withthe normal counterparts These miRs often play crucial rolesas antioncogenes Inhibition of these miRs contributes toenhanced tumorigenesis through the modulation of diversemolecular pathways Upregulation of prooncogenic miRs hasbeen detected in RMS recently as well [84] (Table 4)

51 Antioncogenic miR in RMS Gain-of-function experimentshave demonstrated that reexpression of selected ldquotumor sup-pressorrdquo miRs impairs the tumorigenic behavior of RMS cells[84 85] As myo-miR family members miR-1 miR-133a andmiR-206 have been demonstrated to be downregulated inRMS and shown to have activity on mRNA expression bytargeting c-Met [42 43 86] Inhibition of thesemyo-miRNAsmay cause aberrant cell proliferation and migration in myo-genesis leading to RMS development especially for ERMSThere have been more miRs found as tumor suppressors inthis malignancy including miR-29 miR-450b-5p miR-203

8 Sarcoma

Table 4 MicroRNAs involved in myogenesis and RMS development

miRNA Expression Target Function Reference

miR-1 and miR-133a Downregulation MYH9Myogenic miRNA inhibit differentiationand promote proliferation in myogenesiscytostatic

Rao et al 2010 [42]

miR-206 Downregulation cMET Promote differentiation and proliferationin myogenesis

Yan et al 2009 [43]

miR-29 Downregulation HADC4 YY1 EZH2 Promote stabilization of RMS phenotype Marchesi et al 2014 [44]

miR-450-5p Downregulation ENOX PAX9 Promote differentiation and progression Sun et al 2014 [45]

miR-203 Downregulation JAK STAT Notch Inhibit differentiation and proliferation inmyogenesis tumor suppressor

Diao et al 2014 [46]

miR-214 Downregulation N-Ras Inhibit tumor cell growth and inducemyogenic differentiation and apoptosis

Huang et al 2014 [47]

miR-183 Upregulation EGR1 Promote migration and metastasis Sarver et al 2010 [48]

PMS2

MGMT

HR

Mismatch Recombination

NHEJ MMEJ

p53

Excision

BER NER

RMS

DNA repair

Direct repair Indirect repair

repair repairrepair

Figure 4The DNA repair systems in RMSThere are two pathways repairing the DNA lesions directly without affecting DNA structure andindirectly by DNA phosphodiester backbone cleavage The modifications in DNA repair enzymes expression or activity lead to resistanceto chemotherapy and radiation in RMS tumor cells MGMT O6-methylguanine-DNA methyltransferase BER base excision repair NERnucleotide excision repair HR homologous recombination NHEJ nonhomologous end-joining MMEJ microhomology-mediated end-joining

and miR-214 [44 46 47 84 87] They are all significantlydownregulated in RMS compared with normal skeletal mus-cles and function to inhibit tumor cell growth and inducemyogenic differentiation and apoptosis in RMS

52 Oncogenic miRNA in RMS The transcription factor EGR1is a tumor suppressor gene that is downregulated in RMS [48]miR-183 functions as an oncogene by targeting EGR1 and pro-moting tumor cell migration Either by direct anti-miR treat-ment or by indirect mechanisms that decrease transcriptmiR-183 targeted treatmentsmight provide a potential optionto RMS patients

The basic strategy of current effective miR-based treat-ment studies is either efficient reexpression of miRs or restor-ing the function of miRs to inhibit the expression of certain

protein-coding genes and promote muscle differentiationMoreover miR expression profiling in tumors and possiblytheir detection in peripheral blood during treatment may beable to predict the response to chemotherapy or radiotherapyand be useful as a prognostic signature for the development oftreatment resistanceThe prognostic value of miR expressionlevels in RMS is a powerful tool in creating a better-tailoredstrategy for particular subsets of patients

53 Long Noncoding RNAs in RMS Long noncoding RNAs(LNCRNAs) are abundant in the mammalian transcriptomeand have been shown to play a role in RMS development [8889]TheH19 gene localized within a chromosomal region onhuman chromosome 11p15 encodes an imprinted untrans-lated RNA [90] H19 opposite tumor suppressor (HOTS)

Sarcoma 9

Table 5 In vivo animal models of RMS

Model TargetModel origin Reference

Inactivation Expression

DrosophilaPAX-FKHR Bipartite Gal4-UAS

expression systemGalindo et al2006 [49]

PAX7-FOXO1 rols Chromosomal deletionDf(3L)vin5

Avirneni-Vadlamudi et al2012 [50]

Zebrafishrag2 promoter c-Myc kRASG12D

Chen andLangenau 2011[51]

MAPKERK andAKTS6K1 PD98059 TPCK Tg(hsp70-HRASG12V) Le et al 2012

[52]

Mouse

Sufu N-myc Sfrp1 Ptch2and cyclin D1 Sufu+minus Lee et al 2007

[53]

Ink4aArf and Trp53 Pax3FKHR Pax3P3Fpwt Keller et al2004 [54]

Wnt120573-cateninsignaling pathway Wnt2 p53minusminusc-fosminusminus Singh et al 2010

[55]

a tumor growth inhibitor is encoded by an imprinted H19antisense transcript Overexpression of HOTS inhibits RMStumor cell growth Silencing HOTS by RNAi was shown toincrease in vitro colony formation as well as in vivo tumorgrowth of RMS [90]

6 In Vivo Models of RMS

Several in vivo models have recently been developed inDrosophila zebrafish and mice to further understand themolecular and cellular biology of RMS tumorigenesis [49ndash55 91ndash94] (Table 5)

61 Drosophila Models in RMS Despite the evolutionarydistance a strong conservation of genes pathways andregulatory molecular networks have been demonstratedbetween flies and humans Many human disease genes haverelated sequences in Drosophila In contrast to mammalianmodels the generation of Drosophila mutants is easy cheapand fast

For a better understanding of the pathogenic conse-quences of PAX-FKHR expression Drosophila was chosenbecause the animalrsquos transparent outer cuticle allows forsimple real-time monitoring of muscle abnormalities elicitedby PAX-FKHR including subtle or focal changes [49] Themutation in the myoblast fusion gene rolling pebbles (rols)was reported to dominantly suppress PAX-FKHR1-inducedlethality by using a Drosophila model of PAX-FKHR1-mediated transformation [50] PAX-FKHR1 signaling upreg-ulates the TANC1 gene and blocks myoblasts from terminaldifferentiation However downregulating the TANC1 genecould cause RMS cells to lose their neoplastic state undergofusion and form differentiation This novel finding uncov-ered a PAX-FKHR1-TANC1 neoplasia axis collaboratively ina Drosophila model and loss-gain-of-function studies inmammalian platforms [50]

62 Zebrafish Models in RMS In contrast to other mod-els the short tumor onset in zebrafish allows for rapididentification of essential genes in various processes suchas tumor growth self-renewal and maintenance There areother advantages for using zebrafish in the study of humandisease and development including the ease and low cost ofraising large numbers of fish and the highly conserved geneticand biochemical pathways between zebrafish and mammals[95]

As zebrafish RMS is highly similar to human ERMSby using fluorescent transgenic approaches it is more con-venient to understand histogenesis and different aspects oftumorigenesis in RMS A robust zebrafish model of RAS-induced RMS has been established sharing twomorphologicand immunophenotypic features resembling ERMS [51] Inone case both are associated with tissue-restricted geneexpression in RMS while in another both comprise a RAS-induced gene signature Cross-species microarray compar-isons confirm that conserved genetic tumor-specific andtissue-restricted pathways such as RAS and p53 pathwaysdrive RMS growth Tg(hsp70-HRASG12V) zebrafish embryoswere generated to evaluate gene expression that mimics RASpathway activation during tumorigenesis to defineRAS targetgenes [52] Another KRASG12D-induced zebrafish embryonalRMS was utilized to assess the therapeutic effects By theblockage of two major downstream signaling pathwaysMAPKERK and AKTS6K1 the model showed that inhibi-tion of translation initiation suppresses tumor cell prolifera-tion [52]

All the above zebrafish cancer models share similar cellu-lar features and molecular pathways with human RMS espe-cially ERMS and can demonstrate a response to therapiesin a similar manner with human RMS Thus a tremendousopportunity is available to implement these animal modelsinto different steps for novel targeted therapeutic develop-ment

10 Sarcoma

63 Mouse Models in RMS Mouse models established fromin vivo studies may contribute to the understanding of thegenetic basis in tumor development and progression andmayhelp to test the efficacies of novel antineoplastic agents

The importance of PAX3PAX7-FKHR fusion proteins inthe progression of ARMS tumorigenesis is evident from bothin vitro and in vivo studies Transgenic mouse models gener-ated with the PAX-FKHR fusion genemimic the formation ofhuman ARMS [67] A mouse model was developed with het-erozygous loss of Sufu a tumor suppressor inHedgehog (Hh)signaling combined with p53 loss driven ERMS with a lowpenetrance (9) [53] These studies indicate that the modelsconsistently formingARMSnot only need the introduction ofthe PAX-FKHR fusion gene but also need to be accompaniedby other genetic events such as p53 pathway disruption [54]

Compared with ARMS ERMS models are more complexto generate because of their lower tumor penetrance andlonger latency JW41 cells from the p53c-fos double mutantmice models resemble human ERMS cells morphologicallyand express similar characteristic markers [55] The overex-pression of the Wnt2 gene identified in JW41 cells confirmedin human RMS cell lines can allow for further analysis of theWnt signaling pathway Both of these results indicate that thedownregulation of theWnt pathway contributes to resistanceto apoptosis and the inhibition of myogenic differentiation inERMS

Using Ptch1 p53 andor Rb1 conditional mouse modelsand controlling prenatal or postnatal myogenic cell of originRubin et al demonstrated that the loss of p53 in maturingmyoblasts related to tumorigenesis of ERMS [96] They alsoindicated that Rb1 alteration with other oncogenic factorswas strongly associated with an undifferentiated phenotypeacting as a modifier In addition they highlighted a sub-set of p53-deficient ERMSs arising from Myf6-expressingmyoblasts which had the same latency with Pax7-expressingmurine satellite cells in the study

7 Potential Therapeutic Targets in RMS

Detection of genomic imbalances and identification of thecrucial effective genes can contribute to identifying novelpotential biomarkers and relevant targets for clinical therapyin RMS

Survivin-responsive conditionally replicating adenovi-ruses can regulate multiple factors (Survm-CRAs) [41]Although Survm-CRA can efficiently replicate and potentlyinduce cell death in RMS cells the cytotoxic effects are morepronounced in RSC-enriched or RSC-purified cells thanin RSC-exiguous or progeny-purified cells Injections ofSurvm-CRAs into tumor nodules generated by transplantingRSC-enriched cells induce significant death in RMS cells andregression of tumor nodulesThe unique therapeutic featuresof Survm-CRA include not only its therapeutic effectivenessagainst RMS but also its increasing effectiveness against CSCsuggesting that Survm-CRA may be a promising anticanceragent [41]

Rapamycin an inhibitor of mTOR can abrogate RMStumor growth in a xenograft mouse model [97] Thetumors in the rapamycin-treatedmice group show significant

reduction of proliferation and invasiveness and inductionof apoptosis The tumor growth inhibition is simultaneousassociated with the diminution of mTOR and Hh pathwayswhich are implicated in the pathogenesis of RMSThe resultsindicate that using rapamycin either alone or in combinationwith traditional chemotherapeutic drugs may represent apotential targeted agent for therapeutic intervention Ampli-fication and mutational activation of Fibroblast Growth Fac-tor Receptor 4 (FGFR4) in RMS cells promote tumor pro-gression [98] Ponatinib is the most potent FGFR4 inhibitorto block wild-type RMS cell growth mutate FGFR4 throughincreasing apoptosis and suppress the FGFR4 downstreamtarget STAT3 Ponatinib treatment slows tumor growth inRMS mouse models expressing mutated FGFR4 [98] Pona-tinib as an FGFR4 inhibitor shows potential to act as aneffective therapeutic agent for RMS tumorsThe expression ofCXC chemokine receptor 4 (CXCR4) CXCR7 and VascularEndothelial Growth Factor (VEGF) indicates poor prognosisin a variety of malignancies including RMS cell lines Mostsamples in a large series of clinical RMS cases show asso-ciation with high expression of these antagonists regardlessof their subtypes [99] However there are significant corre-lations with high expression of CXCR4 and VEGF in bothERMS and ARMS High VEGF expression is predictive ofadverse prognostic factors in RMS as the expression of theseangiogenesis factors was compared with clinicopathologicalparameters and prognosis Considering their overexpressionin RMS these chemokine receptors and VEGF could providepotential molecular therapeutic targets in RMS Some ARMScell lines have undergone apoptosis in response to antineo-plastic drugs such as bortezomib The proapoptotic BH3only family member NOXA acts as an upregulated proteindownstream PAX3-FKHR resulting in increased cell sensi-tivity to bortezomib Apoptosis in response to bortezomibcan be reversed by shRNA knockdown of Noxa PAX3-FKHR upregulation of Noxa creates a potential therapeuticinsight into inducing apoptosis in ARMS cellsThis apoptosispathway could represent a specific targeted therapy againstPAX3-FKHR-expressing ARMS cells [100]

Proton pump activation resulting from intracellular acid-ificationmay be an energetic mechanism to escape apoptosisV-ATPase is an ATP-driven proton pump acidifying theintracellular compartment and transports protons across theplasma membrane Esomeprazole as a V-ATPase inhibitor inthe PPI administration interferes with intensive ion trans-porter activity of RMS cells and induces remarkable cytotoxi-city RMS expresses a higher level ofV-ATPase comparedwithother sarcomasTherefore RMS should be very susceptible toesomeprazole treatment and V-ATPase could be consideredas a promising selective target for treatment [101]

8 Conclusion and Future Prospects

For RMS the current challenges include how to iden-tify promising novel therapeutics and integrate them intoexisting therapy For better understanding of congenitaland epigenetic modifications in the development of RMSthis review summarizes the recent genome-wide studies onmolecular and genetic alterations to decipher the underlying

Sarcoma 11

tumorigenesis mechanisms There is a link between geneticand epigenetic alterations responsible for a variety of tumorcell growth proliferation differentiation apoptosis andtherapy-resistance mechanisms The identification of theprognostic value of the PAX-FKHR fusion status in RMSis one of the most important shifts in the lineage and therisk assessment of RMS which exerts an oncogenic effectthrough multiple pathways and is incorporated into mostrelevant studies The misbalanced expression of a number ofmiRs involved in the regulation of myogenic differentiationalso implicates the tumorsrsquo escape suppressive mechanismsMoreover we described several in vivo models of RMSto explore underlying histogenesis and different aspects oftumorigenesis Recent work on CSCs has also contributedto understanding the cell origins refining molecular char-acterization and elucidating the underlying basis of refrac-tory responses to current adjuvant treatments of subsets oftumorsThese integrative approaches can provide opportuni-ties to identify diagnostic and prognostic biomarkers appliedto the individual targeted therapy and significantly improveRMS prognosis

Abbreviations

RMS RhabdomyosarcomaARMS Alveolar rhabdomyosarcomaERMS Embryonal rhabdomyosarcomaPRMS Pleomorphic rhabdomyosarcomaSRMS Sclerosingspindle cell

rhabdomyosarcomamiRNA miR MicroRNACSC Cancer stem cellMSC Mesenchymal stem cell

Conflict of Interests

The authors declare that there is no conflict of interestsregarding the publication of this paper

Acknowledgments

This work was supported in part by grants from the Gat-tegno and Wechsler funds Dr Duan was supported in partthrough a grant from Sarcoma Foundation of America (SFA)a grant from National Cancer Institute (NCI)National Insti-tutes of Health (NIH) UO1 CA 151452 and a grant from anAcademic Enrichment Fund of MGH Orthopedic Surgery

References

[1] L Yang T Takimoto and J Fujimoto ldquoPrognostic model forpredicting overall survival in children and adolescents withrhabdomyosarcomardquo BMC Cancer vol 14 no 1 article 6542014

[2] D Egas-Bejar and W W Huh ldquoRhabdomyosarcoma in adoles-cent and young adult patients current perspectivesrdquo AdolescentHealth Medicine andTherapeutics vol 5 pp 115ndash125 2014

[3] J C Breneman E Lyden A S Pappo et al ldquoPrognosticfactors and clinical outcomes in children and adolescents withmetastatic rhabdomyosarcomamdasha report from the intergrouprhabdomyosarcoma study IVrdquo Journal of Clinical Oncology vol21 no 1 pp 78ndash84 2003

[4] V D Soleimani and M A Rudnicki ldquoNew insights into theorigin and the genetic basis of rhabdomyosarcomasrdquo CancerCell vol 19 no 2 pp 157ndash159 2011

[5] T Glass D D Cochrane S R Rassekh K Goddard and JHukin ldquoGrowing teratoma syndrome in intracranial non-ger-minomatous germ cell tumors (iNGGCTs) a risk for secondarymalignant transformationmdasha report of two casesrdquo Childrsquos Ner-vous System vol 30 no 5 pp 953ndash957 2014

[6] J Ji C Eng and K Hemminki ldquoFamilial risk for soft tissuetumors a nation-wide epidemiological study from SwedenrdquoJournal of Cancer Research and Clinical Oncology vol 134 no5 pp 617ndash624 2008

[7] C D M Fletcher J A Bridge P Hogendoorn and F MertensWHO Classification of Tumours of Soft Tissue and Bone vol 5IARC Press Paris France 4th edition 2013

[8] I Sultan I Qaddoumi S Yaser C Rodriguez-Galindo and AFerrari ldquoComparing adult and pediatric rhabdomyosarcoma inthe surveillance epidemiology and end results program 1973 to2005 an analysis of 2600 patientsrdquo Journal of Clinical Oncologyvol 27 no 20 pp 3391ndash3397 2009

[9] B Raney W Huh D Hawkins et al ldquoOutcome of patientswith localized orbital sarcoma who relapsed following treat-ment on Intergroup Rhabdomyosarcoma Study Group (IRSG)Protocols-III and -IV 1984ndash1997 a report from the ChildrenrsquosOncology Grouprdquo Pediatric Blood and Cancer vol 60 no 3 pp371ndash376 2013

[10] W Crist E A Gehan A H Ragab et al ldquoThe third intergrouprhabdomyosarcoma studyrdquo Journal of Clinical Oncology vol 13no 3 pp 610ndash630 1995

[11] C Liu D Li J Jiang et al ldquoAnalysis of molecular cytogeneticalteration in rhabdomyosarcoma by array comparative genomichybridizationrdquo PLoS ONE vol 9 no 4 Article ID e94924 2014

[12] DWilliamson E Missiaglia A de Reynies et al ldquoFusion gene-negative alveolar rhabdomyosarcoma is clinically and molec-ularly indistinguishable from embryonal rhabdomyosarcomardquoJournal of Clinical Oncology vol 28 no 13 pp 2151ndash2158 2010

[13] V Paulson G Chandler D Rakheja et al ldquoHigh-resolutionarray CGH identifies common mechanisms that drive embry-onal rhabdomyosarcoma pathogenesisrdquo Genes Chromosomesand Cancer vol 50 no 6 pp 397ndash408 2011

[14] S Park J Lee I-G Do et al ldquoAberrant CDK4 amplificationin refractory rhabdomyosarcoma as identified by genomicprofilingrdquo Scientific Reports vol 4 article 3623 2014

[15] C Liu D Li J Hu et al ldquoChromosomal and genetic imbalancesin Chinese patients with rhabdomyosarcoma detected by high-resolution array comparative genomic hybridizationrdquo Interna-tional Journal of Clinical and Experimental Pathology vol 7 no2 pp 690ndash698 2014

[16] Q-X Li C-X Liu C-P Chun et al ldquoChromosomal imbalancesrevealed in primary rhabdomyo-sarcomas by comparativegenomic hybridizationrdquo Chinese Medical Journal vol 122 no11 pp 1277ndash1282 2009

[17] E Missiaglia J Selfe M Hamdi et al ldquoGenomic imbalancesin rhabdomyosarcoma cell lines affect expression of genesfrequently altered in primary tumors an approach to identifycandidate genes involved in tumor developmentrdquo Genes Chro-mosomes and Cancer vol 48 no 6 pp 455ndash467 2009

12 Sarcoma

[18] F G Barr F Duan L M Smith et al ldquoGenomic and clinicalanalyses of 2p24 and 12q13-q14 amplification in alveolar rhab-domyosarcoma a report from the childrenrsquos oncology grouprdquoGenes Chromosomes and Cancer vol 48 no 8 pp 661ndash6722009

[19] T Gordon AMcManus J Anderson et al ldquoCytogenetic abnor-malities in 42 rhabdomyosarcoma a United Kingdom CancerCytogenetics Group StudyrdquoMedical and Pediatric Oncology vol36 no 2 pp 259ndash267 2001

[20] X Chen E Stewart A A Shelat et al ldquoTargeting oxidativestress in embryonal rhabdomyosarcomardquo Cancer Cell vol 24no 6 pp 710ndash724 2013

[21] S Lizard-Nacol F Mugneret C Volk C Turc-Carel M Favrotand T Philip ldquoTranslocation (213)(q37q14) in alveolar rhab-domyosarcoma a new caserdquo Cancer Genetics and Cytogeneticsvol 25 no 2 pp 373ndash374 1987

[22] P H B Sorensen J C Lynch S J Qualman et al ldquoPAX3-FKHR and PAX7-FKHR gene fusions are prognostic indicatorsin alveolar rhabdomyosarcoma a report from the childrenrsquosoncology grouprdquo Journal of Clinical Oncology vol 20 no 11 pp2672ndash2679 2002

[23] A D Marshall and G C Grosveld ldquoAlveolar rhabdomyo-sarcomamdashthe molecular drivers of PAX37-FOXO1-inducedtumorigenesisrdquo Skeletal Muscle vol 2 no 1 article 25 2012

[24] E Missiaglia D Williamson J Chisholm et al ldquoPAX3FOXO1fusion gene status is the key prognostic molecular markerin rhabdomyosarcoma and significantly improves current riskstratificationrdquo Journal of Clinical Oncology vol 30 no 14 pp1670ndash1677 2012

[25] J F Shern L Chen J Chmielecki et al ldquoComprehensivegenomic analysis of rhabdomyosarcoma reveals a landscape ofalterations affecting a common genetic axis in fusion-positiveand fusion-negative tumorsrdquo Cancer Discovery vol 4 no 2 pp216ndash231 2014

[26] MWachtel M Dettling E Koscielniak et al ldquoGene expressionsignatures identify rhabdomyosarcoma subtypes and detect anovel t(22)(q35p23) translocation fusing PAX3 to NCOA1rdquoCancer Research vol 64 no 16 pp 5539ndash5545 2004

[27] E Davicioni M J Anderson F G Finckenstein et al ldquoMolec-ular classification of rhabdomyosarcomamdashgenotypic and phe-notypic determinants of diagnosis a report from the ChildrenrsquosOncology Grouprdquo The American Journal of Pathology vol 174no 2 pp 550ndash564 2009

[28] M Visser C Sijmons J Bras et al ldquoAllelotype of pediatric rhab-domyosarcomardquo Oncogene vol 15 no 11 pp 1309ndash1314 1997

[29] R Gil-Benso T San-Miguel R C Callaghan et al ldquoChromo-somal and genetic changes produced in tumoral progression ofembryonal rhabdomyosarcomardquo Histopathology vol 62 no 5pp 816ndash819 2013

[30] S X Skapek J Anderson F G Barr et al ldquoPAX-FOXO1 fusionstatus drives unfavorable outcome for children with rhabdom-yosarcoma a childrenrsquos oncology group reportrdquo Pediatric Bloodand Cancer vol 60 no 9 pp 1411ndash1417 2013

[31] HHosoi N Kakazu E Konishi et al ldquoA novel PAX3 rearrange-ment in embryonal rhabdomyosarcomardquo Cancer Genetics andCytogenetics vol 189 no 2 pp 98ndash104 2009

[32] E Takaoka H Sonobe K Akimaru et al ldquoMultiple sites ofhighly amplified DNA sequences detected bymolecular cytoge-netic analysis inHS-RMS-2 a newpleomorphic rhabdomyosar-coma cell linerdquo American Journal of Cancer Research vol 2 no2 pp 141ndash152 2012

[33] K Szuhai D de Jong W Y Leung C D M Fletcher and P CWHogendoorn ldquoTransactivatingmutation of theMYOD1 geneis a frequent event in adult spindle cell rhabdomyosarcomardquoJournal of Pathology vol 232 no 3 pp 300ndash307 2014

[34] N P Agaram C-L Chen L Zhang M P Laquaglia L Wexlerand C R Antonescu ldquoRecurrent MYOD1 mutations in pedi-atric and adult sclerosing and spindle cell rhabdomyosarcomasEvidence for a common pathogenesisrdquoGenes Chromosomes andCancer vol 53 no 9 pp 779ndash787 2014

[35] H Yoshida M Miyachi K Sakamoto et al ldquoPAX3-NCOA2fusion gene has a dual role in promoting the proliferation andinhibiting the myogenic differentiation of rhabdomyosarcomacellsrdquo Oncogene vol 33 pp 5601ndash5608 2013

[36] J M Mosquera A Sboner L Zhang et al ldquoRecurrent NCOA2gene rearrangements in congenitalinfantile spindle cell rhab-domyosarcomardquo Genes Chromosomes and Cancer vol 52 no6 pp 538ndash550 2013

[37] V Thavaraj A Sethi and L S Arya ldquoIncomplete Beckwith-Wiedemann syndrome in a child with orbital rhabdomyosar-comardquo Indian Pediatrics vol 39 no 3 pp 299ndash304 2002

[38] D Walter S Satheesha P Albrecht et al ldquoCD133 positiveembryonal rhabdomyosarcoma stem-like cell population isenriched in rhabdospheresrdquo PLoS ONE vol 6 no 5 Article IDe19506 2011

[39] Y-X Ren F G Finckenstein D A Abdueva et al ldquoMousemesenchymal stem cells expressing PAX-FKHR form alveolarrhabdomyosarcomas by cooperating with secondary muta-tionsrdquo Cancer Research vol 68 no 16 pp 6587ndash6597 2008

[40] M Salerno S Avnet G Bonuccelli S Hosogi D Granchi andN Baldini ldquoImpairment of lysosomal activity as a therapeuticmodality targeting cancer stem cells of embryonal rhabdom-yosarcoma cell line RDrdquo PLoS ONE vol 9 no 10 Article IDe110340 2014

[41] K Tanoue Y Wang M Ikeda et al ldquoSurvivin-responsive con-ditionally replicating adenovirus kills rhabdomyosarcoma stemcells more efficiently than their progenyrdquo Journal of Transla-tional Medicine vol 12 no 1 article 27 2014

[42] P K Rao EMissiaglia L Shields et al ldquoDistinct roles formiR-1and miR-133a in the proliferation and differentiation of rhab-domyosarcoma cellsrdquo The FASEB Journal vol 24 no 9 pp3427ndash3437 2010

[43] D Yan X D Dong X Chen et al ldquoMicroRNA-1206 targets c-met and inhibits rhabdomyosarcomadevelopmentrdquoThe Journalof Biological Chemistry vol 284 no 43 pp 29596ndash29604 2009

[44] I Marchesi A Giordano and L Bagella ldquoRoles of enhancerof zeste homolog 2 from skeletal muscle differentiation torhabdomyosarcoma carcinogenesisrdquoCell Cycle vol 13 no 4 pp516ndash527 2014

[45] M M Sun J F Li L L Guo et al ldquoTGF-1205731 suppression ofmicroRNA-450b-5p expression a novel mechanism for block-ingmyogenic differentiation of rhabdomyosarcomardquoOncogenevol 33 no 16 pp 2075ndash2086 2014

[46] Y Diao X Guo L Jiang et al ldquoMiR-203 a tumor suppressorfrequently down-regulated by promoter hypermethylation inrhabdomyosarcomardquo The Journal of Biological Chemistry vol289 no 1 pp 529ndash539 2014

[47] H-J Huang J Liu HHua et al ldquoMiR-214 andN-ras regulatoryloop suppresses rhabdomyosarcoma cell growth and xenografttumorigenesisrdquo Oncotarget vol 5 no 8 pp 2161ndash2175 2014

Sarcoma 13

[48] A L Sarver L Li and S Subramanian ldquomicroRNA miR-183functions as an oncogene by targeting the transcription factorEGR1 and promoting tumor cell migrationrdquo Cancer Researchvol 70 no 23 pp 9570ndash9580 2010

[49] R L Galindo J A Allport and E N Olson ldquoA Drosophilamodel of the rhabdomyosarcoma initiator PAX7-FKHRrdquo Pro-ceedings of the National Academy of Sciences of the United Statesof America vol 103 no 36 pp 13439ndash13444 2006

[50] U Avirneni-Vadlamudi K A Galindo T R Endicott V Paul-son S Cameron and R L Galindo ldquoDrosophila and mammal-ian models uncover a role for the myoblast fusion gene TANC1in rhabdomyosarcomardquo The Journal of Clinical Investigationvol 122 no 1 pp 403ndash407 2012

[51] E Y Chen andDM Langenau ldquoZebrafishmodels of rhabdom-yosarcomardquoMethods in Cell Biology vol 105 pp 383ndash402 2011

[52] X Le E K Pugach S Hettmer et al ldquoA novel chemicalscreening strategy in zebrafish identifies common pathways inembryogenesis and rhabdomyosarcoma developmentrdquo Devel-opment vol 140 no 11 pp 2354ndash2364 2012

[53] Y Lee R Kawagoe K Sasai et al ldquoLoss of suppressor-of-fusedfunction promotes tumorigenesisrdquoOncogene vol 26 no 44 pp6442ndash6447 2007

[54] C Keller B R Arenkiel C M Coffin N El-Bardeesy R ADePinho and M R Capecchi ldquoAlveolar rhabdomyosarcomasin conditional Pax3Fkhr mice cooperativity of Ink4aARF andTrp53 loss of functionrdquo Genes and Development vol 18 no 21pp 2614ndash2626 2004

[55] S Singh CVinson CMGurley et al ldquoImpairedWnt signalingin embryonal rhabdomyosarcoma cells from p53c-fos doublemutant micerdquo American Journal of Pathology vol 177 no 4 pp2055ndash2066 2010

[56] G J Schaaf J M Ruijter F Van Ruissen et al ldquoFull transcrip-tome analysis of rhabdomyosarcoma normal and fetal skeletalmuscle statistical comparison of multiple SAGE librariesrdquo TheFASEB Journal vol 19 no 3 pp 404ndash406 2005

[57] D S Hawkins A A Gupta and E R Rudzinski ldquoWhat is newin the biology and treatment of pediatric rhabdomyosarcomardquoCurrent Opinion in Pediatrics vol 26 no 1 pp 50ndash56 2014

[58] M Zhang J Truscott and J Davie ldquoLoss of MEF2D expressioninhibits differentiation and contributes to oncogenesis in rhab-domyosarcoma cellsrdquoMolecular Cancer vol 12 no 1 article 1502013

[59] M Ciesla J Dulak and A Jozkowicz ldquomicroRNAs and epi-genetic mechanisms of rhabdomyosarcoma developmentrdquo TheInternational Journal of Biochemistry amp Cell Biology vol 53 pp482ndash492 2014

[60] S Ricaud B Vernus M Duclos et al ldquoInhibition of autocrinesecretion of myostatin enhances terminal differentiation inhuman rhabdomyosarcoma cellsrdquo Oncogene vol 22 no 51 pp8221ndash8232 2003

[61] C S Guo C Degnin T A Fiddler D Stauffer andM JThayerldquoRegulation of MyoD activity and muscle cell differentiation byMDM2 pRb and Sp1rdquoThe Journal of Biological Chemistry vol278 no 25 pp 22615ndash22622 2003

[62] S Rossi E Stoppani P L Puri and A Fanzani ldquoDifferenti-ation of human rhabdomyosarcoma RD cells is regulated byreciprocal functional interactions between myostatin p38 andextracellular regulated kinase signalling pathwaysrdquo EuropeanJournal of Cancer vol 47 no 7 pp 1095ndash1105 2011

[63] Z S Walters B Villarejo-Balcells D Olmos et al ldquoJARID2is a direct target of the PAX3-FOXO1 fusion protein andinhibits myogenic differentiation of rhabdomyosarcoma cellsrdquoOncogene vol 33 no 9 pp 1148ndash1157 2014

[64] T Hosoyama M I Aslam J Abraham et al ldquoIL-4R drives ded-ifferentiation mitogenesis and metastasis in rhabdomyosar-comardquo Clinical Cancer Research vol 17 no 9 pp 2757ndash27662011

[65] F S Dela Cruz ldquoCancer stem cells in pediatric sarcomasrdquoFrontiers in Oncology vol 3 article 168 2013

[66] M J Bissell and W C Hines ldquoWhy donrsquot we get more cancerA proposed role of the microenvironment in restraining cancerprogressionrdquo Nature Medicine vol 17 no 3 pp 320ndash329 2011

[67] E Charytonowicz C Cordon-Cardo I Matushansky and MZiman ldquoAlveolar rhabdomyosarcoma is the cell of origin amesenchymal stem cellrdquoCancer Letters vol 279 no 2 pp 126ndash136 2009

[68] W K Cavenee ldquoMuscling in on rhabdomyosarcomardquo NatureMedicine vol 8 no 11 pp 1200ndash1201 2002

[69] B Zhu and J K Davie ldquoNew insights into signalling-pathwayalterations in rhabdomyosarcomardquo British Journal of Cancervol 112 no 2 pp 227ndash231 2015

[70] M Zhang C M Linardic and D G Kirsch ldquoRAS and ROS inrhabdomyosarcomardquo Cancer Cell vol 24 no 6 pp 689ndash6912013

[71] S C Marianna L Ianzano and G Nicoletti ldquoTumor sup-pressor genes promote rhabdomyosarcoma progression in p53heterozygousrdquo Oncotarget vol 5 no 1 pp 108ndash119 2013

[72] SWang L Guo L Dong et al ldquoTGF-beta1 signal pathwaymaycontribute to rhabdomyosarcoma development by inhibitingdifferentiationrdquo Cancer Science vol 101 no 5 pp 1108ndash11162010

[73] J Wesche K Haglund and E M Haugsten ldquoFibroblast growthfactors and their receptors in cancerrdquo Biochemical Journal vol437 no 2 pp 199ndash213 2011

[74] M Wachtel J Rakic M Okoniewski P Bode F Niggli andB W Schafer ldquoFGFR4 signaling couples to Bim and not Bmfto discriminate subsets of alveolar rhabdomyosarcoma cellsrdquoInternational Journal of Cancer vol 135 no 7 pp 1543ndash15522014

[75] P L Puri ZWu P Zhang et al ldquoInduction of terminal differen-tiation by constitutive activation of p38 MAP kinase in humanrhabdomyosarcoma cellsrdquo Genes and Development vol 14 no5 pp 574ndash584 2000

[76] A M Tremblay E Missiaglia G G Galli et al ldquoThe Hippotransducer YAP1 transforms activated satellite cells and is apotent effector of embryonal rhabdomyosarcoma formationrdquoCancer Cell vol 26 no 2 pp 273ndash287 2014

[77] R N Judson A M Tremblay P Knopp et al ldquoThe hippo path-way member Yap plays a key role in influencing fate decisionsin muscle satellite cellsrdquo Journal of Cell Science vol 125 no 24pp 6009ndash6019 2012

[78] L E S Crose K A Galindo J G Kephart et al ldquoAlve-olar rhabdomyosarcoma-associated PAX3-FOXO1 promotestumorigenesis via Hippo pathway suppressionrdquo Journal ofClinical Investigation vol 124 no 1 pp 285ndash296 2014

[79] B Belyea J G Kephart J Blum D G Kirsch and C MLinardic ldquoEmbryonic signaling pathways and rhabdomyosar-coma contributions to cancer development and opportunitiesfor therapeutic targetingrdquo Sarcoma vol 2012 Article ID 40623913 pages 2012

14 Sarcoma

[80] J Huertas-Martinez S Rello-Varona D Herrero-Martin et alldquoCaveolin-1 is down-regulated in alveolar rhabdomyosarcomasand negatively regulates tumor growthrdquo Oncotarget vol 5 no20 pp 9744ndash9755 2014

[81] S Rossi P L Poliani CMissale EMonti andA Fanzani ldquoCav-eolins in rhabdomyosarcomardquo Journal of Cellular andMolecularMedicine vol 15 no 12 pp 2553ndash2568 2011

[82] P G Tsioli E S Patsouris C Giaginis and S E TheocharisldquoDNA repair systems in rhabdomyosarcomardquo Histology andHistopathology vol 28 no 8 pp 971ndash984 2013

[83] E Gatzidou C Michailidi S Tseleni-Balafouta and S Theo-charis ldquoAn epitome of DNA repair related genes and mecha-nisms in thyroid carcinomardquo Cancer Letters vol 290 no 2 pp139ndash147 2010

[84] R Rota R Ciarapica A Giordano L Miele and F LocatellildquoMicroRNAs in rhabdomyosarcoma pathogenetic implicationsand translational potentialityrdquoMolecular Cancer vol 10 article120 2011

[85] A Gougelet J Perez D Pissaloux et al ldquoMiRNA profiling howto bypass the current difficulties in the diagnosis and treatmentof sarcomasrdquo Sarcoma vol 2011 Article ID 460650 13 pages2011

[86] R Ciarapica G Russo F Verginelli et al ldquoDeregulated expres-sion of miR-26a and Ezh2 in rhabdomyosarcomardquo Cell Cyclevol 8 no 1 pp 172ndash175 2009

[87] L Li A L Sarver S Alamgir and S Subramanian ldquoDownreg-ulation of microRNAs miR-1 -206 and -29 stabilizes PAX3 andCCND2 expression in rhabdomyosarcomardquo Laboratory Investi-gation vol 92 no 4 pp 571ndash583 2012

[88] S-Y Ng G K Bogu B Soh and L W Stanton ldquoThe longnoncoding RNA RMST interacts with SOX2 to regulate neu-rogenesisrdquoMolecular Cell vol 51 no 3 pp 349ndash359 2013

[89] C A Lynch B Tycko T H Bestor and C P Walsh ldquoReac-tivation of a silenced H19 gene in human rhabdomyosarcomaby demethylation of DNA but not by histone hyperacetylationrdquoMolecular Cancer vol 1 article 2 2002

[90] P Onyango and A P Feinberg ldquoA nucleolar protein H19opposite tumor suppressor (HOTS) is a tumor growth inhibitorencoded by a human imprinted H19 antisense transcriptrdquoProceedings of the National Academy of Sciences of the UnitedStates of America vol 108 no 40 pp 16759ndash16764 2011

[91] C E Albacker N Y Storer E M Langdon et al ldquoThe his-tone methyltransferase SUV39H1 suppresses embryonal rhab-domyosarcoma formation in zebrafishrdquo PLoS ONE vol 8 no 5Article ID e64969 2013

[92] N Y Storer R M White A Uong et al ldquoZebrafish rhab-domyosarcoma reflects the developmental stage of oncogeneexpression during myogenesisrdquo Development vol 140 no 14pp 3040ndash3050 2013

[93] M E Hatley W Tang M R Garcia et al ldquoA mouse modelof rhabdomyosarcoma originating from the adipocyte lineagerdquoCancer Cell vol 22 no 4 pp 536ndash546 2012

[94] V Hosur A Kavirayani J Riefler et al ldquoDystrophin and dysfer-lin doublemutantmice a novelmodel for rhabdomyosarcomardquoCancer Genetics vol 205 no 5 pp 232ndash241 2012

[95] W Goessling T E North and L I Zon ldquoNew waves of discov-ery modeling cancer in zebrafishrdquo Journal of Clinical Oncologyvol 25 no 17 pp 2473ndash2479 2007

[96] B P Rubin K Nishijo H-I H Chen et al ldquoEvidence for anunanticipated relationship between undifferentiated pleomor-phic sarcoma and embryonal rhabdomyosarcomardquo Cancer Cellvol 19 no 2 pp 177ndash191 2011

[97] S Z Kaylani J Xu R K Srivastava L Kopelovich J G Presseyand M Athar ldquoRapamycin targeting mTOR and hedgehog sig-naling pathways blocks human rhabdomyosarcoma growth inxenograft murine modelrdquo Biochemical and Biophysical ResearchCommunications vol 435 no 4 pp 557ndash561 2013

[98] S Q Li A T Cheuk J F Shern et al ldquoTargeting wild-type andmutationally activated FGFR4 in rhabdomyosarcoma with theinhibitor ponatinib (AP24534)rdquoPLoSONE vol 8 no 10 ArticleID e76551 2013

[99] K Miyoshi K Kohashi F Fushimi et al ldquoClose correlationbetween CXCR4 and VEGF expression and frequent CXCR7expression in rhabdomyosarcomardquo Human Pathology vol 45no 9 pp 1900ndash1909 2014

[100] ADMarshall F Picchione R I KGeltink andGCGrosveldldquoPAX3-FOXO1 induces up-regulation of Noxa sensitizing alve-olar rhabdomyosarcoma cells to apoptosisrdquo Neoplasia vol 15no 7 pp 738ndash748 2013

[101] F Perut S Avnet C Fotia et al ldquoV-ATPase as an effective thera-peutic target for sarcomasrdquo Experimental Cell Research vol 320no 1 pp 21ndash32 2014

Submit your manuscripts athttpwwwhindawicom

Stem CellsInternational

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

MEDIATORSINFLAMMATION

of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Behavioural Neurology

EndocrinologyInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Disease Markers

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

BioMed Research International

OncologyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Oxidative Medicine and Cellular Longevity

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

PPAR Research

The Scientific World JournalHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Immunology ResearchHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal of

ObesityJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Computational and Mathematical Methods in Medicine

OphthalmologyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Diabetes ResearchJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Research and TreatmentAIDS

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Gastroenterology Research and Practice

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Parkinsonrsquos Disease

Evidence-Based Complementary and Alternative Medicine

Volume 2014Hindawi Publishing Corporationhttpwwwhindawicom

Page 6: Review Article Rhabdomyosarcoma: Advances in Molecular and

6 Sarcoma

METCDKN2AB

PAX37-FKHR

MDM2

RAS

Raf

GLI1

PDK1p53Rb

Hh

Myo MEF2

p21

Ptch

FGFR4

IGF1R

p16INK4Ap14ARF

p19ARF

IGF2

FGF

Figure 3 Genetic analyses of RMS have pinpointed several common alterations including inactivation of a master regulator of p53 andRb pathways CDKN2AB and activation of FGFR4 RAS and Hedgehog (Hh) signaling The modifications of these pathways influenceoncogenesis and metastatic potential

As a human hematopoietic CSC marker Walter et alproposed that upregulated CD133 in ERMS can act as aprognostic marker and might help with the development ofnovel targeted therapies for ERMS [38]

4 Signaling Pathway Alterations in RMS

Recent studies have looked into signaling pathway alterationsand their downstream effects in RMSThese new findings notonly helped to improve the understanding of this malignancybut also offered novel potential therapeutic strategies forimproved treatment for patientswithRMS [68 69] (Figure 3)

41 RAS Signaling Pathway RASmutations commonlymain-tain the protein in its GTP bound state and therefore renderit constitutively active in RMS Zhang et al identified RASfamily members NRAS KRAS and HRAS in the RASpathway as themost commonlymutated genes in ERMS [70]the mutation status of these genes is significantly associatedwith the risk of ERMS development [70] A study foundthat a majority of ERMS demonstrated activation of the RASpathway exclusively either by homozygous deletion of NF1or by point mutations in one of the RAS family members asdescribed above [13] Despite remarkable genetic and molec-ular heterogeneity most (93 4144) RMS tumors hijackeda common receptor tyrosine kinaseRASPIK3CA geneticaxis [25] It could occur via two alternative mechanismsrearrangement of a PAX gene and accumulation of mutationsthat were downstream targets of the PAX fusion protein

Skeletal muscle cells have a robust antioxidant defensesystem to protect the DNA lipids and proteins from thedeleterious effects of excess Reactive Oxygen Species (ROS)

Cancer cells also have elevated ROS due to their increasedmetabolic activity oncogenic stimulation andmitochondrialdysfunction These findings implicate RAS mutations andoxidative stress as potential therapeutic targets for high-riskERMS

42 IGF Signaling Pathway There is clear preclinical datathat supports the involvement of the Insulin-like GrowthFactor (IGF) signaling pathway in RMS tumorigenesis andprogression [69] Zhu and Davie demonstrated that IGFpromotes the proliferation of RMS cells while blocking IGFsignaling interferes with cell growth in vivoThe IGF receptorIGF-1R is highly overexpressed both on the cell surface andin the nucleus of RMS cells furthermore RMS cell lines aresensitive to IGF-1R inhibitionThe IGF-2 locus shows a loss ofimprinting in both ERMS andARMS tumors and the expres-sion of PAX3-FKHR has been demonstrated to upregulateIGF-2 and activate IGF pathway in ARMS [69] Mariannaet al found that IGF-2 and tumor suppressors p19Arf andp21Cip1 are overexpressed and prominently upregulated inRMS mouse models [71]

43 TGF-120573 Signaling Pathway The regulatory role of Trans-forming Growth Factor-120573 (TGF-120573) in RMS has been recentlystudied [72] Wang et al demonstrated that the expression ofTGF-1205731 is significantly higher in RMS than in normal skeletalmuscle The inhibition of TGF-1205731 expression by shRNA-expressing vectors reverses themalignant behavior of RMSbyinhibiting cell growth and inducingmyogenic differentiationTGF-1205731 shRNA induces myogenin expression a regulator ofmyogenic differentiation genes Myogenin acts as a target fornegative regulation of myogenesis by TGF-1205731 signals with

Sarcoma 7

a differentiation regulatory cascade These results suggestthat the TGF-1205731 signaling pathway disrupts the differentia-tion of myogenic progenitors leading to the development ofRMS

44 FGF Signaling Pathway Fibroblast Growth Factor (FGF)is crucial in embryonic development and functions to driveproliferation FGFs and their receptors (FGFRs) are essentialregulators in the processes of proliferation antiapoptosisdrug resistance and angiogenesis in RMS [69] RMS over-expresses the receptor tyrosine kinase FGFR4 which causesautophosphorylation and constitutive signaling in correlationwith poor differentiation and decreased survival [73] Recentwork has shown that FGF signaling can prevent ARMS cellsfrom apoptosis induced by targeting the IGF1-R-PI3K-mTORpathway [74]

45 ERK Signaling Pathway Previous studies have shownthat elevated myostatin expression deregulates ExtracellularRegulated Kinase (ERK) signaling and deficient activationof the p38 pathway contributes to the differentiation blockin RMS cells [62] The ERK pathway is frequently highlyactivated in RMS cells from embryonic derivation due to thepresence of activating RASmutations leading to reduced dif-ferentiation by blocking the p38 pathway [75]These findingsindicate that interventions that target themyostatinERKp38network could be an effective way to promote differentiationof RMS

46 Hippo Signaling Pathway Many upstream signal trans-duction proteins in response to cues from the cellular micro-environment regulate the core Hippo pathway [76] TheHippo pathway may limit tumorigenesis by inducing thecytosolic localization of the transcriptional cofactor Yes-Associated Protein 1 (YAP1) High YAP levels and activitiesincrease activated satellite cells and prevent differentiationby activating genes that inhibit differentiation [77] YAP1-TEAD1 was found to upregulate proproliferative and onco-genic genes and maintain the ERMS differentiation blockby interfering with MYOD1 and MEF2 prodifferentiationactivities [76] Inhibition of theHippo pathway is exhibited inRMS through downregulation of Hippo pathway tumor sup-pressors or upregulation of YAP [78] These data suggest thatHippo pathway dysfunction promotes RMS development

47 Notch Signaling Pathway and Wnt Signaling PathwayNotch pathway as an embryonic signaling pathway promotesmuscle stem cell maintenance by inhibiting myogenic earlydifferentiation and expanding pools of progenitor cells dur-ing both embryogenesis and postnatal muscle regenerationSimilar to Notch Wnt has various roles in embryonic fetaland neonatalmyogenesis during skeletalmyogenesis As bothpathways are regulators of myogenic lineage determinationand maturation RMS can arise from disordered regulationof these normal developmental pathways [79]

48 Caveolins in RMS Recent findings have shown that cav-eolins are expressed in a cell stage-dependent manner inRMS [80 81] Caveolins are scaffolding proteins that regulate

several targets and pathways (such as p53 IGF RASERK andTGF-120573myostatin signaling) associated with RMS develop-ment and therefore loss or gain of caveolin function is ableto generate multiple effects on the tumor behaviorThereforerecognizing their precise roles in RMS progression will becrucial to elaborate targeted therapies particularly as oneof the three different isoforms CAV1 could act as a potenttumor suppressor in ARMS Inhibition of CAV1 function cancontribute to aberrant cell proliferation leading to ARMSdevelopment [80]

49 DNA Repairing System Some studies indicate that RMSis characterized by germline mutations of DNA repair genes[82 83] DNA repair gene alterations in RMS occur sec-ondarily to malignant transformation The modificationsin DNA repair enzyme activity can result in resistance toadjuvant treatment in RMS tumor cells which limits theprognosis of RMS There are two pathways for repairingDNA breaks directly without affecting DNA structure andindirectly by DNA phosphodiester backbone cleavage [82](Figure 4) Direct repair includes repair during replicationand enzymatic repair Indirect repair is comprised of exci-sion repair systems including base excision repair (BER)nucleotide excision repair (NER) mismatch repair (MMR)and recombination repair (RR) [83] Defective DNA repairmechanisms result from point mutations or LOH in RMSand contribute to tumorigenesis MGMT and MMR proteinactivity and expression levels are used as predictor indices fortherapy outcome in RMS BER as the major DNA repair sys-tem against damage resulting from cellular metabolism canreverse the cytotoxic effects of alkylation agents used suchas antineoplastics subsequent tumor progression and deathimplicates an RMS mechanism of resistance to chemother-apeutic agents This may provide novel individualized ther-apeutics targeting downregulation of activated DNA repairenzymes or upregulation of DNA repair deficiencies [82]

5 MicroRNA (miRNA miR)Expression in RMS

Muscle-specific and ubiquitously expressed miRs appeardownregulated in RMS tumors and cell lines compared withthe normal counterparts These miRs often play crucial rolesas antioncogenes Inhibition of these miRs contributes toenhanced tumorigenesis through the modulation of diversemolecular pathways Upregulation of prooncogenic miRs hasbeen detected in RMS recently as well [84] (Table 4)

51 Antioncogenic miR in RMS Gain-of-function experimentshave demonstrated that reexpression of selected ldquotumor sup-pressorrdquo miRs impairs the tumorigenic behavior of RMS cells[84 85] As myo-miR family members miR-1 miR-133a andmiR-206 have been demonstrated to be downregulated inRMS and shown to have activity on mRNA expression bytargeting c-Met [42 43 86] Inhibition of thesemyo-miRNAsmay cause aberrant cell proliferation and migration in myo-genesis leading to RMS development especially for ERMSThere have been more miRs found as tumor suppressors inthis malignancy including miR-29 miR-450b-5p miR-203

8 Sarcoma

Table 4 MicroRNAs involved in myogenesis and RMS development

miRNA Expression Target Function Reference

miR-1 and miR-133a Downregulation MYH9Myogenic miRNA inhibit differentiationand promote proliferation in myogenesiscytostatic

Rao et al 2010 [42]

miR-206 Downregulation cMET Promote differentiation and proliferationin myogenesis

Yan et al 2009 [43]

miR-29 Downregulation HADC4 YY1 EZH2 Promote stabilization of RMS phenotype Marchesi et al 2014 [44]

miR-450-5p Downregulation ENOX PAX9 Promote differentiation and progression Sun et al 2014 [45]

miR-203 Downregulation JAK STAT Notch Inhibit differentiation and proliferation inmyogenesis tumor suppressor

Diao et al 2014 [46]

miR-214 Downregulation N-Ras Inhibit tumor cell growth and inducemyogenic differentiation and apoptosis

Huang et al 2014 [47]

miR-183 Upregulation EGR1 Promote migration and metastasis Sarver et al 2010 [48]

PMS2

MGMT

HR

Mismatch Recombination

NHEJ MMEJ

p53

Excision

BER NER

RMS

DNA repair

Direct repair Indirect repair

repair repairrepair

Figure 4The DNA repair systems in RMSThere are two pathways repairing the DNA lesions directly without affecting DNA structure andindirectly by DNA phosphodiester backbone cleavage The modifications in DNA repair enzymes expression or activity lead to resistanceto chemotherapy and radiation in RMS tumor cells MGMT O6-methylguanine-DNA methyltransferase BER base excision repair NERnucleotide excision repair HR homologous recombination NHEJ nonhomologous end-joining MMEJ microhomology-mediated end-joining

and miR-214 [44 46 47 84 87] They are all significantlydownregulated in RMS compared with normal skeletal mus-cles and function to inhibit tumor cell growth and inducemyogenic differentiation and apoptosis in RMS

52 Oncogenic miRNA in RMS The transcription factor EGR1is a tumor suppressor gene that is downregulated in RMS [48]miR-183 functions as an oncogene by targeting EGR1 and pro-moting tumor cell migration Either by direct anti-miR treat-ment or by indirect mechanisms that decrease transcriptmiR-183 targeted treatmentsmight provide a potential optionto RMS patients

The basic strategy of current effective miR-based treat-ment studies is either efficient reexpression of miRs or restor-ing the function of miRs to inhibit the expression of certain

protein-coding genes and promote muscle differentiationMoreover miR expression profiling in tumors and possiblytheir detection in peripheral blood during treatment may beable to predict the response to chemotherapy or radiotherapyand be useful as a prognostic signature for the development oftreatment resistanceThe prognostic value of miR expressionlevels in RMS is a powerful tool in creating a better-tailoredstrategy for particular subsets of patients

53 Long Noncoding RNAs in RMS Long noncoding RNAs(LNCRNAs) are abundant in the mammalian transcriptomeand have been shown to play a role in RMS development [8889]TheH19 gene localized within a chromosomal region onhuman chromosome 11p15 encodes an imprinted untrans-lated RNA [90] H19 opposite tumor suppressor (HOTS)

Sarcoma 9

Table 5 In vivo animal models of RMS

Model TargetModel origin Reference

Inactivation Expression

DrosophilaPAX-FKHR Bipartite Gal4-UAS

expression systemGalindo et al2006 [49]

PAX7-FOXO1 rols Chromosomal deletionDf(3L)vin5

Avirneni-Vadlamudi et al2012 [50]

Zebrafishrag2 promoter c-Myc kRASG12D

Chen andLangenau 2011[51]

MAPKERK andAKTS6K1 PD98059 TPCK Tg(hsp70-HRASG12V) Le et al 2012

[52]

Mouse

Sufu N-myc Sfrp1 Ptch2and cyclin D1 Sufu+minus Lee et al 2007

[53]

Ink4aArf and Trp53 Pax3FKHR Pax3P3Fpwt Keller et al2004 [54]

Wnt120573-cateninsignaling pathway Wnt2 p53minusminusc-fosminusminus Singh et al 2010

[55]

a tumor growth inhibitor is encoded by an imprinted H19antisense transcript Overexpression of HOTS inhibits RMStumor cell growth Silencing HOTS by RNAi was shown toincrease in vitro colony formation as well as in vivo tumorgrowth of RMS [90]

6 In Vivo Models of RMS

Several in vivo models have recently been developed inDrosophila zebrafish and mice to further understand themolecular and cellular biology of RMS tumorigenesis [49ndash55 91ndash94] (Table 5)

61 Drosophila Models in RMS Despite the evolutionarydistance a strong conservation of genes pathways andregulatory molecular networks have been demonstratedbetween flies and humans Many human disease genes haverelated sequences in Drosophila In contrast to mammalianmodels the generation of Drosophila mutants is easy cheapand fast

For a better understanding of the pathogenic conse-quences of PAX-FKHR expression Drosophila was chosenbecause the animalrsquos transparent outer cuticle allows forsimple real-time monitoring of muscle abnormalities elicitedby PAX-FKHR including subtle or focal changes [49] Themutation in the myoblast fusion gene rolling pebbles (rols)was reported to dominantly suppress PAX-FKHR1-inducedlethality by using a Drosophila model of PAX-FKHR1-mediated transformation [50] PAX-FKHR1 signaling upreg-ulates the TANC1 gene and blocks myoblasts from terminaldifferentiation However downregulating the TANC1 genecould cause RMS cells to lose their neoplastic state undergofusion and form differentiation This novel finding uncov-ered a PAX-FKHR1-TANC1 neoplasia axis collaboratively ina Drosophila model and loss-gain-of-function studies inmammalian platforms [50]

62 Zebrafish Models in RMS In contrast to other mod-els the short tumor onset in zebrafish allows for rapididentification of essential genes in various processes suchas tumor growth self-renewal and maintenance There areother advantages for using zebrafish in the study of humandisease and development including the ease and low cost ofraising large numbers of fish and the highly conserved geneticand biochemical pathways between zebrafish and mammals[95]

As zebrafish RMS is highly similar to human ERMSby using fluorescent transgenic approaches it is more con-venient to understand histogenesis and different aspects oftumorigenesis in RMS A robust zebrafish model of RAS-induced RMS has been established sharing twomorphologicand immunophenotypic features resembling ERMS [51] Inone case both are associated with tissue-restricted geneexpression in RMS while in another both comprise a RAS-induced gene signature Cross-species microarray compar-isons confirm that conserved genetic tumor-specific andtissue-restricted pathways such as RAS and p53 pathwaysdrive RMS growth Tg(hsp70-HRASG12V) zebrafish embryoswere generated to evaluate gene expression that mimics RASpathway activation during tumorigenesis to defineRAS targetgenes [52] Another KRASG12D-induced zebrafish embryonalRMS was utilized to assess the therapeutic effects By theblockage of two major downstream signaling pathwaysMAPKERK and AKTS6K1 the model showed that inhibi-tion of translation initiation suppresses tumor cell prolifera-tion [52]

All the above zebrafish cancer models share similar cellu-lar features and molecular pathways with human RMS espe-cially ERMS and can demonstrate a response to therapiesin a similar manner with human RMS Thus a tremendousopportunity is available to implement these animal modelsinto different steps for novel targeted therapeutic develop-ment

10 Sarcoma

63 Mouse Models in RMS Mouse models established fromin vivo studies may contribute to the understanding of thegenetic basis in tumor development and progression andmayhelp to test the efficacies of novel antineoplastic agents

The importance of PAX3PAX7-FKHR fusion proteins inthe progression of ARMS tumorigenesis is evident from bothin vitro and in vivo studies Transgenic mouse models gener-ated with the PAX-FKHR fusion genemimic the formation ofhuman ARMS [67] A mouse model was developed with het-erozygous loss of Sufu a tumor suppressor inHedgehog (Hh)signaling combined with p53 loss driven ERMS with a lowpenetrance (9) [53] These studies indicate that the modelsconsistently formingARMSnot only need the introduction ofthe PAX-FKHR fusion gene but also need to be accompaniedby other genetic events such as p53 pathway disruption [54]

Compared with ARMS ERMS models are more complexto generate because of their lower tumor penetrance andlonger latency JW41 cells from the p53c-fos double mutantmice models resemble human ERMS cells morphologicallyand express similar characteristic markers [55] The overex-pression of the Wnt2 gene identified in JW41 cells confirmedin human RMS cell lines can allow for further analysis of theWnt signaling pathway Both of these results indicate that thedownregulation of theWnt pathway contributes to resistanceto apoptosis and the inhibition of myogenic differentiation inERMS

Using Ptch1 p53 andor Rb1 conditional mouse modelsand controlling prenatal or postnatal myogenic cell of originRubin et al demonstrated that the loss of p53 in maturingmyoblasts related to tumorigenesis of ERMS [96] They alsoindicated that Rb1 alteration with other oncogenic factorswas strongly associated with an undifferentiated phenotypeacting as a modifier In addition they highlighted a sub-set of p53-deficient ERMSs arising from Myf6-expressingmyoblasts which had the same latency with Pax7-expressingmurine satellite cells in the study

7 Potential Therapeutic Targets in RMS

Detection of genomic imbalances and identification of thecrucial effective genes can contribute to identifying novelpotential biomarkers and relevant targets for clinical therapyin RMS

Survivin-responsive conditionally replicating adenovi-ruses can regulate multiple factors (Survm-CRAs) [41]Although Survm-CRA can efficiently replicate and potentlyinduce cell death in RMS cells the cytotoxic effects are morepronounced in RSC-enriched or RSC-purified cells thanin RSC-exiguous or progeny-purified cells Injections ofSurvm-CRAs into tumor nodules generated by transplantingRSC-enriched cells induce significant death in RMS cells andregression of tumor nodulesThe unique therapeutic featuresof Survm-CRA include not only its therapeutic effectivenessagainst RMS but also its increasing effectiveness against CSCsuggesting that Survm-CRA may be a promising anticanceragent [41]

Rapamycin an inhibitor of mTOR can abrogate RMStumor growth in a xenograft mouse model [97] Thetumors in the rapamycin-treatedmice group show significant

reduction of proliferation and invasiveness and inductionof apoptosis The tumor growth inhibition is simultaneousassociated with the diminution of mTOR and Hh pathwayswhich are implicated in the pathogenesis of RMSThe resultsindicate that using rapamycin either alone or in combinationwith traditional chemotherapeutic drugs may represent apotential targeted agent for therapeutic intervention Ampli-fication and mutational activation of Fibroblast Growth Fac-tor Receptor 4 (FGFR4) in RMS cells promote tumor pro-gression [98] Ponatinib is the most potent FGFR4 inhibitorto block wild-type RMS cell growth mutate FGFR4 throughincreasing apoptosis and suppress the FGFR4 downstreamtarget STAT3 Ponatinib treatment slows tumor growth inRMS mouse models expressing mutated FGFR4 [98] Pona-tinib as an FGFR4 inhibitor shows potential to act as aneffective therapeutic agent for RMS tumorsThe expression ofCXC chemokine receptor 4 (CXCR4) CXCR7 and VascularEndothelial Growth Factor (VEGF) indicates poor prognosisin a variety of malignancies including RMS cell lines Mostsamples in a large series of clinical RMS cases show asso-ciation with high expression of these antagonists regardlessof their subtypes [99] However there are significant corre-lations with high expression of CXCR4 and VEGF in bothERMS and ARMS High VEGF expression is predictive ofadverse prognostic factors in RMS as the expression of theseangiogenesis factors was compared with clinicopathologicalparameters and prognosis Considering their overexpressionin RMS these chemokine receptors and VEGF could providepotential molecular therapeutic targets in RMS Some ARMScell lines have undergone apoptosis in response to antineo-plastic drugs such as bortezomib The proapoptotic BH3only family member NOXA acts as an upregulated proteindownstream PAX3-FKHR resulting in increased cell sensi-tivity to bortezomib Apoptosis in response to bortezomibcan be reversed by shRNA knockdown of Noxa PAX3-FKHR upregulation of Noxa creates a potential therapeuticinsight into inducing apoptosis in ARMS cellsThis apoptosispathway could represent a specific targeted therapy againstPAX3-FKHR-expressing ARMS cells [100]

Proton pump activation resulting from intracellular acid-ificationmay be an energetic mechanism to escape apoptosisV-ATPase is an ATP-driven proton pump acidifying theintracellular compartment and transports protons across theplasma membrane Esomeprazole as a V-ATPase inhibitor inthe PPI administration interferes with intensive ion trans-porter activity of RMS cells and induces remarkable cytotoxi-city RMS expresses a higher level ofV-ATPase comparedwithother sarcomasTherefore RMS should be very susceptible toesomeprazole treatment and V-ATPase could be consideredas a promising selective target for treatment [101]

8 Conclusion and Future Prospects

For RMS the current challenges include how to iden-tify promising novel therapeutics and integrate them intoexisting therapy For better understanding of congenitaland epigenetic modifications in the development of RMSthis review summarizes the recent genome-wide studies onmolecular and genetic alterations to decipher the underlying

Sarcoma 11

tumorigenesis mechanisms There is a link between geneticand epigenetic alterations responsible for a variety of tumorcell growth proliferation differentiation apoptosis andtherapy-resistance mechanisms The identification of theprognostic value of the PAX-FKHR fusion status in RMSis one of the most important shifts in the lineage and therisk assessment of RMS which exerts an oncogenic effectthrough multiple pathways and is incorporated into mostrelevant studies The misbalanced expression of a number ofmiRs involved in the regulation of myogenic differentiationalso implicates the tumorsrsquo escape suppressive mechanismsMoreover we described several in vivo models of RMSto explore underlying histogenesis and different aspects oftumorigenesis Recent work on CSCs has also contributedto understanding the cell origins refining molecular char-acterization and elucidating the underlying basis of refrac-tory responses to current adjuvant treatments of subsets oftumorsThese integrative approaches can provide opportuni-ties to identify diagnostic and prognostic biomarkers appliedto the individual targeted therapy and significantly improveRMS prognosis

Abbreviations

RMS RhabdomyosarcomaARMS Alveolar rhabdomyosarcomaERMS Embryonal rhabdomyosarcomaPRMS Pleomorphic rhabdomyosarcomaSRMS Sclerosingspindle cell

rhabdomyosarcomamiRNA miR MicroRNACSC Cancer stem cellMSC Mesenchymal stem cell

Conflict of Interests

The authors declare that there is no conflict of interestsregarding the publication of this paper

Acknowledgments

This work was supported in part by grants from the Gat-tegno and Wechsler funds Dr Duan was supported in partthrough a grant from Sarcoma Foundation of America (SFA)a grant from National Cancer Institute (NCI)National Insti-tutes of Health (NIH) UO1 CA 151452 and a grant from anAcademic Enrichment Fund of MGH Orthopedic Surgery

References

[1] L Yang T Takimoto and J Fujimoto ldquoPrognostic model forpredicting overall survival in children and adolescents withrhabdomyosarcomardquo BMC Cancer vol 14 no 1 article 6542014

[2] D Egas-Bejar and W W Huh ldquoRhabdomyosarcoma in adoles-cent and young adult patients current perspectivesrdquo AdolescentHealth Medicine andTherapeutics vol 5 pp 115ndash125 2014

[3] J C Breneman E Lyden A S Pappo et al ldquoPrognosticfactors and clinical outcomes in children and adolescents withmetastatic rhabdomyosarcomamdasha report from the intergrouprhabdomyosarcoma study IVrdquo Journal of Clinical Oncology vol21 no 1 pp 78ndash84 2003

[4] V D Soleimani and M A Rudnicki ldquoNew insights into theorigin and the genetic basis of rhabdomyosarcomasrdquo CancerCell vol 19 no 2 pp 157ndash159 2011

[5] T Glass D D Cochrane S R Rassekh K Goddard and JHukin ldquoGrowing teratoma syndrome in intracranial non-ger-minomatous germ cell tumors (iNGGCTs) a risk for secondarymalignant transformationmdasha report of two casesrdquo Childrsquos Ner-vous System vol 30 no 5 pp 953ndash957 2014

[6] J Ji C Eng and K Hemminki ldquoFamilial risk for soft tissuetumors a nation-wide epidemiological study from SwedenrdquoJournal of Cancer Research and Clinical Oncology vol 134 no5 pp 617ndash624 2008

[7] C D M Fletcher J A Bridge P Hogendoorn and F MertensWHO Classification of Tumours of Soft Tissue and Bone vol 5IARC Press Paris France 4th edition 2013

[8] I Sultan I Qaddoumi S Yaser C Rodriguez-Galindo and AFerrari ldquoComparing adult and pediatric rhabdomyosarcoma inthe surveillance epidemiology and end results program 1973 to2005 an analysis of 2600 patientsrdquo Journal of Clinical Oncologyvol 27 no 20 pp 3391ndash3397 2009

[9] B Raney W Huh D Hawkins et al ldquoOutcome of patientswith localized orbital sarcoma who relapsed following treat-ment on Intergroup Rhabdomyosarcoma Study Group (IRSG)Protocols-III and -IV 1984ndash1997 a report from the ChildrenrsquosOncology Grouprdquo Pediatric Blood and Cancer vol 60 no 3 pp371ndash376 2013

[10] W Crist E A Gehan A H Ragab et al ldquoThe third intergrouprhabdomyosarcoma studyrdquo Journal of Clinical Oncology vol 13no 3 pp 610ndash630 1995

[11] C Liu D Li J Jiang et al ldquoAnalysis of molecular cytogeneticalteration in rhabdomyosarcoma by array comparative genomichybridizationrdquo PLoS ONE vol 9 no 4 Article ID e94924 2014

[12] DWilliamson E Missiaglia A de Reynies et al ldquoFusion gene-negative alveolar rhabdomyosarcoma is clinically and molec-ularly indistinguishable from embryonal rhabdomyosarcomardquoJournal of Clinical Oncology vol 28 no 13 pp 2151ndash2158 2010

[13] V Paulson G Chandler D Rakheja et al ldquoHigh-resolutionarray CGH identifies common mechanisms that drive embry-onal rhabdomyosarcoma pathogenesisrdquo Genes Chromosomesand Cancer vol 50 no 6 pp 397ndash408 2011

[14] S Park J Lee I-G Do et al ldquoAberrant CDK4 amplificationin refractory rhabdomyosarcoma as identified by genomicprofilingrdquo Scientific Reports vol 4 article 3623 2014

[15] C Liu D Li J Hu et al ldquoChromosomal and genetic imbalancesin Chinese patients with rhabdomyosarcoma detected by high-resolution array comparative genomic hybridizationrdquo Interna-tional Journal of Clinical and Experimental Pathology vol 7 no2 pp 690ndash698 2014

[16] Q-X Li C-X Liu C-P Chun et al ldquoChromosomal imbalancesrevealed in primary rhabdomyo-sarcomas by comparativegenomic hybridizationrdquo Chinese Medical Journal vol 122 no11 pp 1277ndash1282 2009

[17] E Missiaglia J Selfe M Hamdi et al ldquoGenomic imbalancesin rhabdomyosarcoma cell lines affect expression of genesfrequently altered in primary tumors an approach to identifycandidate genes involved in tumor developmentrdquo Genes Chro-mosomes and Cancer vol 48 no 6 pp 455ndash467 2009

12 Sarcoma

[18] F G Barr F Duan L M Smith et al ldquoGenomic and clinicalanalyses of 2p24 and 12q13-q14 amplification in alveolar rhab-domyosarcoma a report from the childrenrsquos oncology grouprdquoGenes Chromosomes and Cancer vol 48 no 8 pp 661ndash6722009

[19] T Gordon AMcManus J Anderson et al ldquoCytogenetic abnor-malities in 42 rhabdomyosarcoma a United Kingdom CancerCytogenetics Group StudyrdquoMedical and Pediatric Oncology vol36 no 2 pp 259ndash267 2001

[20] X Chen E Stewart A A Shelat et al ldquoTargeting oxidativestress in embryonal rhabdomyosarcomardquo Cancer Cell vol 24no 6 pp 710ndash724 2013

[21] S Lizard-Nacol F Mugneret C Volk C Turc-Carel M Favrotand T Philip ldquoTranslocation (213)(q37q14) in alveolar rhab-domyosarcoma a new caserdquo Cancer Genetics and Cytogeneticsvol 25 no 2 pp 373ndash374 1987

[22] P H B Sorensen J C Lynch S J Qualman et al ldquoPAX3-FKHR and PAX7-FKHR gene fusions are prognostic indicatorsin alveolar rhabdomyosarcoma a report from the childrenrsquosoncology grouprdquo Journal of Clinical Oncology vol 20 no 11 pp2672ndash2679 2002

[23] A D Marshall and G C Grosveld ldquoAlveolar rhabdomyo-sarcomamdashthe molecular drivers of PAX37-FOXO1-inducedtumorigenesisrdquo Skeletal Muscle vol 2 no 1 article 25 2012

[24] E Missiaglia D Williamson J Chisholm et al ldquoPAX3FOXO1fusion gene status is the key prognostic molecular markerin rhabdomyosarcoma and significantly improves current riskstratificationrdquo Journal of Clinical Oncology vol 30 no 14 pp1670ndash1677 2012

[25] J F Shern L Chen J Chmielecki et al ldquoComprehensivegenomic analysis of rhabdomyosarcoma reveals a landscape ofalterations affecting a common genetic axis in fusion-positiveand fusion-negative tumorsrdquo Cancer Discovery vol 4 no 2 pp216ndash231 2014

[26] MWachtel M Dettling E Koscielniak et al ldquoGene expressionsignatures identify rhabdomyosarcoma subtypes and detect anovel t(22)(q35p23) translocation fusing PAX3 to NCOA1rdquoCancer Research vol 64 no 16 pp 5539ndash5545 2004

[27] E Davicioni M J Anderson F G Finckenstein et al ldquoMolec-ular classification of rhabdomyosarcomamdashgenotypic and phe-notypic determinants of diagnosis a report from the ChildrenrsquosOncology Grouprdquo The American Journal of Pathology vol 174no 2 pp 550ndash564 2009

[28] M Visser C Sijmons J Bras et al ldquoAllelotype of pediatric rhab-domyosarcomardquo Oncogene vol 15 no 11 pp 1309ndash1314 1997

[29] R Gil-Benso T San-Miguel R C Callaghan et al ldquoChromo-somal and genetic changes produced in tumoral progression ofembryonal rhabdomyosarcomardquo Histopathology vol 62 no 5pp 816ndash819 2013

[30] S X Skapek J Anderson F G Barr et al ldquoPAX-FOXO1 fusionstatus drives unfavorable outcome for children with rhabdom-yosarcoma a childrenrsquos oncology group reportrdquo Pediatric Bloodand Cancer vol 60 no 9 pp 1411ndash1417 2013

[31] HHosoi N Kakazu E Konishi et al ldquoA novel PAX3 rearrange-ment in embryonal rhabdomyosarcomardquo Cancer Genetics andCytogenetics vol 189 no 2 pp 98ndash104 2009

[32] E Takaoka H Sonobe K Akimaru et al ldquoMultiple sites ofhighly amplified DNA sequences detected bymolecular cytoge-netic analysis inHS-RMS-2 a newpleomorphic rhabdomyosar-coma cell linerdquo American Journal of Cancer Research vol 2 no2 pp 141ndash152 2012

[33] K Szuhai D de Jong W Y Leung C D M Fletcher and P CWHogendoorn ldquoTransactivatingmutation of theMYOD1 geneis a frequent event in adult spindle cell rhabdomyosarcomardquoJournal of Pathology vol 232 no 3 pp 300ndash307 2014

[34] N P Agaram C-L Chen L Zhang M P Laquaglia L Wexlerand C R Antonescu ldquoRecurrent MYOD1 mutations in pedi-atric and adult sclerosing and spindle cell rhabdomyosarcomasEvidence for a common pathogenesisrdquoGenes Chromosomes andCancer vol 53 no 9 pp 779ndash787 2014

[35] H Yoshida M Miyachi K Sakamoto et al ldquoPAX3-NCOA2fusion gene has a dual role in promoting the proliferation andinhibiting the myogenic differentiation of rhabdomyosarcomacellsrdquo Oncogene vol 33 pp 5601ndash5608 2013

[36] J M Mosquera A Sboner L Zhang et al ldquoRecurrent NCOA2gene rearrangements in congenitalinfantile spindle cell rhab-domyosarcomardquo Genes Chromosomes and Cancer vol 52 no6 pp 538ndash550 2013

[37] V Thavaraj A Sethi and L S Arya ldquoIncomplete Beckwith-Wiedemann syndrome in a child with orbital rhabdomyosar-comardquo Indian Pediatrics vol 39 no 3 pp 299ndash304 2002

[38] D Walter S Satheesha P Albrecht et al ldquoCD133 positiveembryonal rhabdomyosarcoma stem-like cell population isenriched in rhabdospheresrdquo PLoS ONE vol 6 no 5 Article IDe19506 2011

[39] Y-X Ren F G Finckenstein D A Abdueva et al ldquoMousemesenchymal stem cells expressing PAX-FKHR form alveolarrhabdomyosarcomas by cooperating with secondary muta-tionsrdquo Cancer Research vol 68 no 16 pp 6587ndash6597 2008

[40] M Salerno S Avnet G Bonuccelli S Hosogi D Granchi andN Baldini ldquoImpairment of lysosomal activity as a therapeuticmodality targeting cancer stem cells of embryonal rhabdom-yosarcoma cell line RDrdquo PLoS ONE vol 9 no 10 Article IDe110340 2014

[41] K Tanoue Y Wang M Ikeda et al ldquoSurvivin-responsive con-ditionally replicating adenovirus kills rhabdomyosarcoma stemcells more efficiently than their progenyrdquo Journal of Transla-tional Medicine vol 12 no 1 article 27 2014

[42] P K Rao EMissiaglia L Shields et al ldquoDistinct roles formiR-1and miR-133a in the proliferation and differentiation of rhab-domyosarcoma cellsrdquo The FASEB Journal vol 24 no 9 pp3427ndash3437 2010

[43] D Yan X D Dong X Chen et al ldquoMicroRNA-1206 targets c-met and inhibits rhabdomyosarcomadevelopmentrdquoThe Journalof Biological Chemistry vol 284 no 43 pp 29596ndash29604 2009

[44] I Marchesi A Giordano and L Bagella ldquoRoles of enhancerof zeste homolog 2 from skeletal muscle differentiation torhabdomyosarcoma carcinogenesisrdquoCell Cycle vol 13 no 4 pp516ndash527 2014

[45] M M Sun J F Li L L Guo et al ldquoTGF-1205731 suppression ofmicroRNA-450b-5p expression a novel mechanism for block-ingmyogenic differentiation of rhabdomyosarcomardquoOncogenevol 33 no 16 pp 2075ndash2086 2014

[46] Y Diao X Guo L Jiang et al ldquoMiR-203 a tumor suppressorfrequently down-regulated by promoter hypermethylation inrhabdomyosarcomardquo The Journal of Biological Chemistry vol289 no 1 pp 529ndash539 2014

[47] H-J Huang J Liu HHua et al ldquoMiR-214 andN-ras regulatoryloop suppresses rhabdomyosarcoma cell growth and xenografttumorigenesisrdquo Oncotarget vol 5 no 8 pp 2161ndash2175 2014

Sarcoma 13

[48] A L Sarver L Li and S Subramanian ldquomicroRNA miR-183functions as an oncogene by targeting the transcription factorEGR1 and promoting tumor cell migrationrdquo Cancer Researchvol 70 no 23 pp 9570ndash9580 2010

[49] R L Galindo J A Allport and E N Olson ldquoA Drosophilamodel of the rhabdomyosarcoma initiator PAX7-FKHRrdquo Pro-ceedings of the National Academy of Sciences of the United Statesof America vol 103 no 36 pp 13439ndash13444 2006

[50] U Avirneni-Vadlamudi K A Galindo T R Endicott V Paul-son S Cameron and R L Galindo ldquoDrosophila and mammal-ian models uncover a role for the myoblast fusion gene TANC1in rhabdomyosarcomardquo The Journal of Clinical Investigationvol 122 no 1 pp 403ndash407 2012

[51] E Y Chen andDM Langenau ldquoZebrafishmodels of rhabdom-yosarcomardquoMethods in Cell Biology vol 105 pp 383ndash402 2011

[52] X Le E K Pugach S Hettmer et al ldquoA novel chemicalscreening strategy in zebrafish identifies common pathways inembryogenesis and rhabdomyosarcoma developmentrdquo Devel-opment vol 140 no 11 pp 2354ndash2364 2012

[53] Y Lee R Kawagoe K Sasai et al ldquoLoss of suppressor-of-fusedfunction promotes tumorigenesisrdquoOncogene vol 26 no 44 pp6442ndash6447 2007

[54] C Keller B R Arenkiel C M Coffin N El-Bardeesy R ADePinho and M R Capecchi ldquoAlveolar rhabdomyosarcomasin conditional Pax3Fkhr mice cooperativity of Ink4aARF andTrp53 loss of functionrdquo Genes and Development vol 18 no 21pp 2614ndash2626 2004

[55] S Singh CVinson CMGurley et al ldquoImpairedWnt signalingin embryonal rhabdomyosarcoma cells from p53c-fos doublemutant micerdquo American Journal of Pathology vol 177 no 4 pp2055ndash2066 2010

[56] G J Schaaf J M Ruijter F Van Ruissen et al ldquoFull transcrip-tome analysis of rhabdomyosarcoma normal and fetal skeletalmuscle statistical comparison of multiple SAGE librariesrdquo TheFASEB Journal vol 19 no 3 pp 404ndash406 2005

[57] D S Hawkins A A Gupta and E R Rudzinski ldquoWhat is newin the biology and treatment of pediatric rhabdomyosarcomardquoCurrent Opinion in Pediatrics vol 26 no 1 pp 50ndash56 2014

[58] M Zhang J Truscott and J Davie ldquoLoss of MEF2D expressioninhibits differentiation and contributes to oncogenesis in rhab-domyosarcoma cellsrdquoMolecular Cancer vol 12 no 1 article 1502013

[59] M Ciesla J Dulak and A Jozkowicz ldquomicroRNAs and epi-genetic mechanisms of rhabdomyosarcoma developmentrdquo TheInternational Journal of Biochemistry amp Cell Biology vol 53 pp482ndash492 2014

[60] S Ricaud B Vernus M Duclos et al ldquoInhibition of autocrinesecretion of myostatin enhances terminal differentiation inhuman rhabdomyosarcoma cellsrdquo Oncogene vol 22 no 51 pp8221ndash8232 2003

[61] C S Guo C Degnin T A Fiddler D Stauffer andM JThayerldquoRegulation of MyoD activity and muscle cell differentiation byMDM2 pRb and Sp1rdquoThe Journal of Biological Chemistry vol278 no 25 pp 22615ndash22622 2003

[62] S Rossi E Stoppani P L Puri and A Fanzani ldquoDifferenti-ation of human rhabdomyosarcoma RD cells is regulated byreciprocal functional interactions between myostatin p38 andextracellular regulated kinase signalling pathwaysrdquo EuropeanJournal of Cancer vol 47 no 7 pp 1095ndash1105 2011

[63] Z S Walters B Villarejo-Balcells D Olmos et al ldquoJARID2is a direct target of the PAX3-FOXO1 fusion protein andinhibits myogenic differentiation of rhabdomyosarcoma cellsrdquoOncogene vol 33 no 9 pp 1148ndash1157 2014

[64] T Hosoyama M I Aslam J Abraham et al ldquoIL-4R drives ded-ifferentiation mitogenesis and metastasis in rhabdomyosar-comardquo Clinical Cancer Research vol 17 no 9 pp 2757ndash27662011

[65] F S Dela Cruz ldquoCancer stem cells in pediatric sarcomasrdquoFrontiers in Oncology vol 3 article 168 2013

[66] M J Bissell and W C Hines ldquoWhy donrsquot we get more cancerA proposed role of the microenvironment in restraining cancerprogressionrdquo Nature Medicine vol 17 no 3 pp 320ndash329 2011

[67] E Charytonowicz C Cordon-Cardo I Matushansky and MZiman ldquoAlveolar rhabdomyosarcoma is the cell of origin amesenchymal stem cellrdquoCancer Letters vol 279 no 2 pp 126ndash136 2009

[68] W K Cavenee ldquoMuscling in on rhabdomyosarcomardquo NatureMedicine vol 8 no 11 pp 1200ndash1201 2002

[69] B Zhu and J K Davie ldquoNew insights into signalling-pathwayalterations in rhabdomyosarcomardquo British Journal of Cancervol 112 no 2 pp 227ndash231 2015

[70] M Zhang C M Linardic and D G Kirsch ldquoRAS and ROS inrhabdomyosarcomardquo Cancer Cell vol 24 no 6 pp 689ndash6912013

[71] S C Marianna L Ianzano and G Nicoletti ldquoTumor sup-pressor genes promote rhabdomyosarcoma progression in p53heterozygousrdquo Oncotarget vol 5 no 1 pp 108ndash119 2013

[72] SWang L Guo L Dong et al ldquoTGF-beta1 signal pathwaymaycontribute to rhabdomyosarcoma development by inhibitingdifferentiationrdquo Cancer Science vol 101 no 5 pp 1108ndash11162010

[73] J Wesche K Haglund and E M Haugsten ldquoFibroblast growthfactors and their receptors in cancerrdquo Biochemical Journal vol437 no 2 pp 199ndash213 2011

[74] M Wachtel J Rakic M Okoniewski P Bode F Niggli andB W Schafer ldquoFGFR4 signaling couples to Bim and not Bmfto discriminate subsets of alveolar rhabdomyosarcoma cellsrdquoInternational Journal of Cancer vol 135 no 7 pp 1543ndash15522014

[75] P L Puri ZWu P Zhang et al ldquoInduction of terminal differen-tiation by constitutive activation of p38 MAP kinase in humanrhabdomyosarcoma cellsrdquo Genes and Development vol 14 no5 pp 574ndash584 2000

[76] A M Tremblay E Missiaglia G G Galli et al ldquoThe Hippotransducer YAP1 transforms activated satellite cells and is apotent effector of embryonal rhabdomyosarcoma formationrdquoCancer Cell vol 26 no 2 pp 273ndash287 2014

[77] R N Judson A M Tremblay P Knopp et al ldquoThe hippo path-way member Yap plays a key role in influencing fate decisionsin muscle satellite cellsrdquo Journal of Cell Science vol 125 no 24pp 6009ndash6019 2012

[78] L E S Crose K A Galindo J G Kephart et al ldquoAlve-olar rhabdomyosarcoma-associated PAX3-FOXO1 promotestumorigenesis via Hippo pathway suppressionrdquo Journal ofClinical Investigation vol 124 no 1 pp 285ndash296 2014

[79] B Belyea J G Kephart J Blum D G Kirsch and C MLinardic ldquoEmbryonic signaling pathways and rhabdomyosar-coma contributions to cancer development and opportunitiesfor therapeutic targetingrdquo Sarcoma vol 2012 Article ID 40623913 pages 2012

14 Sarcoma

[80] J Huertas-Martinez S Rello-Varona D Herrero-Martin et alldquoCaveolin-1 is down-regulated in alveolar rhabdomyosarcomasand negatively regulates tumor growthrdquo Oncotarget vol 5 no20 pp 9744ndash9755 2014

[81] S Rossi P L Poliani CMissale EMonti andA Fanzani ldquoCav-eolins in rhabdomyosarcomardquo Journal of Cellular andMolecularMedicine vol 15 no 12 pp 2553ndash2568 2011

[82] P G Tsioli E S Patsouris C Giaginis and S E TheocharisldquoDNA repair systems in rhabdomyosarcomardquo Histology andHistopathology vol 28 no 8 pp 971ndash984 2013

[83] E Gatzidou C Michailidi S Tseleni-Balafouta and S Theo-charis ldquoAn epitome of DNA repair related genes and mecha-nisms in thyroid carcinomardquo Cancer Letters vol 290 no 2 pp139ndash147 2010

[84] R Rota R Ciarapica A Giordano L Miele and F LocatellildquoMicroRNAs in rhabdomyosarcoma pathogenetic implicationsand translational potentialityrdquoMolecular Cancer vol 10 article120 2011

[85] A Gougelet J Perez D Pissaloux et al ldquoMiRNA profiling howto bypass the current difficulties in the diagnosis and treatmentof sarcomasrdquo Sarcoma vol 2011 Article ID 460650 13 pages2011

[86] R Ciarapica G Russo F Verginelli et al ldquoDeregulated expres-sion of miR-26a and Ezh2 in rhabdomyosarcomardquo Cell Cyclevol 8 no 1 pp 172ndash175 2009

[87] L Li A L Sarver S Alamgir and S Subramanian ldquoDownreg-ulation of microRNAs miR-1 -206 and -29 stabilizes PAX3 andCCND2 expression in rhabdomyosarcomardquo Laboratory Investi-gation vol 92 no 4 pp 571ndash583 2012

[88] S-Y Ng G K Bogu B Soh and L W Stanton ldquoThe longnoncoding RNA RMST interacts with SOX2 to regulate neu-rogenesisrdquoMolecular Cell vol 51 no 3 pp 349ndash359 2013

[89] C A Lynch B Tycko T H Bestor and C P Walsh ldquoReac-tivation of a silenced H19 gene in human rhabdomyosarcomaby demethylation of DNA but not by histone hyperacetylationrdquoMolecular Cancer vol 1 article 2 2002

[90] P Onyango and A P Feinberg ldquoA nucleolar protein H19opposite tumor suppressor (HOTS) is a tumor growth inhibitorencoded by a human imprinted H19 antisense transcriptrdquoProceedings of the National Academy of Sciences of the UnitedStates of America vol 108 no 40 pp 16759ndash16764 2011

[91] C E Albacker N Y Storer E M Langdon et al ldquoThe his-tone methyltransferase SUV39H1 suppresses embryonal rhab-domyosarcoma formation in zebrafishrdquo PLoS ONE vol 8 no 5Article ID e64969 2013

[92] N Y Storer R M White A Uong et al ldquoZebrafish rhab-domyosarcoma reflects the developmental stage of oncogeneexpression during myogenesisrdquo Development vol 140 no 14pp 3040ndash3050 2013

[93] M E Hatley W Tang M R Garcia et al ldquoA mouse modelof rhabdomyosarcoma originating from the adipocyte lineagerdquoCancer Cell vol 22 no 4 pp 536ndash546 2012

[94] V Hosur A Kavirayani J Riefler et al ldquoDystrophin and dysfer-lin doublemutantmice a novelmodel for rhabdomyosarcomardquoCancer Genetics vol 205 no 5 pp 232ndash241 2012

[95] W Goessling T E North and L I Zon ldquoNew waves of discov-ery modeling cancer in zebrafishrdquo Journal of Clinical Oncologyvol 25 no 17 pp 2473ndash2479 2007

[96] B P Rubin K Nishijo H-I H Chen et al ldquoEvidence for anunanticipated relationship between undifferentiated pleomor-phic sarcoma and embryonal rhabdomyosarcomardquo Cancer Cellvol 19 no 2 pp 177ndash191 2011

[97] S Z Kaylani J Xu R K Srivastava L Kopelovich J G Presseyand M Athar ldquoRapamycin targeting mTOR and hedgehog sig-naling pathways blocks human rhabdomyosarcoma growth inxenograft murine modelrdquo Biochemical and Biophysical ResearchCommunications vol 435 no 4 pp 557ndash561 2013

[98] S Q Li A T Cheuk J F Shern et al ldquoTargeting wild-type andmutationally activated FGFR4 in rhabdomyosarcoma with theinhibitor ponatinib (AP24534)rdquoPLoSONE vol 8 no 10 ArticleID e76551 2013

[99] K Miyoshi K Kohashi F Fushimi et al ldquoClose correlationbetween CXCR4 and VEGF expression and frequent CXCR7expression in rhabdomyosarcomardquo Human Pathology vol 45no 9 pp 1900ndash1909 2014

[100] ADMarshall F Picchione R I KGeltink andGCGrosveldldquoPAX3-FOXO1 induces up-regulation of Noxa sensitizing alve-olar rhabdomyosarcoma cells to apoptosisrdquo Neoplasia vol 15no 7 pp 738ndash748 2013

[101] F Perut S Avnet C Fotia et al ldquoV-ATPase as an effective thera-peutic target for sarcomasrdquo Experimental Cell Research vol 320no 1 pp 21ndash32 2014

Submit your manuscripts athttpwwwhindawicom

Stem CellsInternational

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

MEDIATORSINFLAMMATION

of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Behavioural Neurology

EndocrinologyInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Disease Markers

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

BioMed Research International

OncologyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Oxidative Medicine and Cellular Longevity

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

PPAR Research

The Scientific World JournalHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Immunology ResearchHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal of

ObesityJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Computational and Mathematical Methods in Medicine

OphthalmologyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Diabetes ResearchJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Research and TreatmentAIDS

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Gastroenterology Research and Practice

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Parkinsonrsquos Disease

Evidence-Based Complementary and Alternative Medicine

Volume 2014Hindawi Publishing Corporationhttpwwwhindawicom

Page 7: Review Article Rhabdomyosarcoma: Advances in Molecular and

Sarcoma 7

a differentiation regulatory cascade These results suggestthat the TGF-1205731 signaling pathway disrupts the differentia-tion of myogenic progenitors leading to the development ofRMS

44 FGF Signaling Pathway Fibroblast Growth Factor (FGF)is crucial in embryonic development and functions to driveproliferation FGFs and their receptors (FGFRs) are essentialregulators in the processes of proliferation antiapoptosisdrug resistance and angiogenesis in RMS [69] RMS over-expresses the receptor tyrosine kinase FGFR4 which causesautophosphorylation and constitutive signaling in correlationwith poor differentiation and decreased survival [73] Recentwork has shown that FGF signaling can prevent ARMS cellsfrom apoptosis induced by targeting the IGF1-R-PI3K-mTORpathway [74]

45 ERK Signaling Pathway Previous studies have shownthat elevated myostatin expression deregulates ExtracellularRegulated Kinase (ERK) signaling and deficient activationof the p38 pathway contributes to the differentiation blockin RMS cells [62] The ERK pathway is frequently highlyactivated in RMS cells from embryonic derivation due to thepresence of activating RASmutations leading to reduced dif-ferentiation by blocking the p38 pathway [75]These findingsindicate that interventions that target themyostatinERKp38network could be an effective way to promote differentiationof RMS

46 Hippo Signaling Pathway Many upstream signal trans-duction proteins in response to cues from the cellular micro-environment regulate the core Hippo pathway [76] TheHippo pathway may limit tumorigenesis by inducing thecytosolic localization of the transcriptional cofactor Yes-Associated Protein 1 (YAP1) High YAP levels and activitiesincrease activated satellite cells and prevent differentiationby activating genes that inhibit differentiation [77] YAP1-TEAD1 was found to upregulate proproliferative and onco-genic genes and maintain the ERMS differentiation blockby interfering with MYOD1 and MEF2 prodifferentiationactivities [76] Inhibition of theHippo pathway is exhibited inRMS through downregulation of Hippo pathway tumor sup-pressors or upregulation of YAP [78] These data suggest thatHippo pathway dysfunction promotes RMS development

47 Notch Signaling Pathway and Wnt Signaling PathwayNotch pathway as an embryonic signaling pathway promotesmuscle stem cell maintenance by inhibiting myogenic earlydifferentiation and expanding pools of progenitor cells dur-ing both embryogenesis and postnatal muscle regenerationSimilar to Notch Wnt has various roles in embryonic fetaland neonatalmyogenesis during skeletalmyogenesis As bothpathways are regulators of myogenic lineage determinationand maturation RMS can arise from disordered regulationof these normal developmental pathways [79]

48 Caveolins in RMS Recent findings have shown that cav-eolins are expressed in a cell stage-dependent manner inRMS [80 81] Caveolins are scaffolding proteins that regulate

several targets and pathways (such as p53 IGF RASERK andTGF-120573myostatin signaling) associated with RMS develop-ment and therefore loss or gain of caveolin function is ableto generate multiple effects on the tumor behaviorThereforerecognizing their precise roles in RMS progression will becrucial to elaborate targeted therapies particularly as oneof the three different isoforms CAV1 could act as a potenttumor suppressor in ARMS Inhibition of CAV1 function cancontribute to aberrant cell proliferation leading to ARMSdevelopment [80]

49 DNA Repairing System Some studies indicate that RMSis characterized by germline mutations of DNA repair genes[82 83] DNA repair gene alterations in RMS occur sec-ondarily to malignant transformation The modificationsin DNA repair enzyme activity can result in resistance toadjuvant treatment in RMS tumor cells which limits theprognosis of RMS There are two pathways for repairingDNA breaks directly without affecting DNA structure andindirectly by DNA phosphodiester backbone cleavage [82](Figure 4) Direct repair includes repair during replicationand enzymatic repair Indirect repair is comprised of exci-sion repair systems including base excision repair (BER)nucleotide excision repair (NER) mismatch repair (MMR)and recombination repair (RR) [83] Defective DNA repairmechanisms result from point mutations or LOH in RMSand contribute to tumorigenesis MGMT and MMR proteinactivity and expression levels are used as predictor indices fortherapy outcome in RMS BER as the major DNA repair sys-tem against damage resulting from cellular metabolism canreverse the cytotoxic effects of alkylation agents used suchas antineoplastics subsequent tumor progression and deathimplicates an RMS mechanism of resistance to chemother-apeutic agents This may provide novel individualized ther-apeutics targeting downregulation of activated DNA repairenzymes or upregulation of DNA repair deficiencies [82]

5 MicroRNA (miRNA miR)Expression in RMS

Muscle-specific and ubiquitously expressed miRs appeardownregulated in RMS tumors and cell lines compared withthe normal counterparts These miRs often play crucial rolesas antioncogenes Inhibition of these miRs contributes toenhanced tumorigenesis through the modulation of diversemolecular pathways Upregulation of prooncogenic miRs hasbeen detected in RMS recently as well [84] (Table 4)

51 Antioncogenic miR in RMS Gain-of-function experimentshave demonstrated that reexpression of selected ldquotumor sup-pressorrdquo miRs impairs the tumorigenic behavior of RMS cells[84 85] As myo-miR family members miR-1 miR-133a andmiR-206 have been demonstrated to be downregulated inRMS and shown to have activity on mRNA expression bytargeting c-Met [42 43 86] Inhibition of thesemyo-miRNAsmay cause aberrant cell proliferation and migration in myo-genesis leading to RMS development especially for ERMSThere have been more miRs found as tumor suppressors inthis malignancy including miR-29 miR-450b-5p miR-203

8 Sarcoma

Table 4 MicroRNAs involved in myogenesis and RMS development

miRNA Expression Target Function Reference

miR-1 and miR-133a Downregulation MYH9Myogenic miRNA inhibit differentiationand promote proliferation in myogenesiscytostatic

Rao et al 2010 [42]

miR-206 Downregulation cMET Promote differentiation and proliferationin myogenesis

Yan et al 2009 [43]

miR-29 Downregulation HADC4 YY1 EZH2 Promote stabilization of RMS phenotype Marchesi et al 2014 [44]

miR-450-5p Downregulation ENOX PAX9 Promote differentiation and progression Sun et al 2014 [45]

miR-203 Downregulation JAK STAT Notch Inhibit differentiation and proliferation inmyogenesis tumor suppressor

Diao et al 2014 [46]

miR-214 Downregulation N-Ras Inhibit tumor cell growth and inducemyogenic differentiation and apoptosis

Huang et al 2014 [47]

miR-183 Upregulation EGR1 Promote migration and metastasis Sarver et al 2010 [48]

PMS2

MGMT

HR

Mismatch Recombination

NHEJ MMEJ

p53

Excision

BER NER

RMS

DNA repair

Direct repair Indirect repair

repair repairrepair

Figure 4The DNA repair systems in RMSThere are two pathways repairing the DNA lesions directly without affecting DNA structure andindirectly by DNA phosphodiester backbone cleavage The modifications in DNA repair enzymes expression or activity lead to resistanceto chemotherapy and radiation in RMS tumor cells MGMT O6-methylguanine-DNA methyltransferase BER base excision repair NERnucleotide excision repair HR homologous recombination NHEJ nonhomologous end-joining MMEJ microhomology-mediated end-joining

and miR-214 [44 46 47 84 87] They are all significantlydownregulated in RMS compared with normal skeletal mus-cles and function to inhibit tumor cell growth and inducemyogenic differentiation and apoptosis in RMS

52 Oncogenic miRNA in RMS The transcription factor EGR1is a tumor suppressor gene that is downregulated in RMS [48]miR-183 functions as an oncogene by targeting EGR1 and pro-moting tumor cell migration Either by direct anti-miR treat-ment or by indirect mechanisms that decrease transcriptmiR-183 targeted treatmentsmight provide a potential optionto RMS patients

The basic strategy of current effective miR-based treat-ment studies is either efficient reexpression of miRs or restor-ing the function of miRs to inhibit the expression of certain

protein-coding genes and promote muscle differentiationMoreover miR expression profiling in tumors and possiblytheir detection in peripheral blood during treatment may beable to predict the response to chemotherapy or radiotherapyand be useful as a prognostic signature for the development oftreatment resistanceThe prognostic value of miR expressionlevels in RMS is a powerful tool in creating a better-tailoredstrategy for particular subsets of patients

53 Long Noncoding RNAs in RMS Long noncoding RNAs(LNCRNAs) are abundant in the mammalian transcriptomeand have been shown to play a role in RMS development [8889]TheH19 gene localized within a chromosomal region onhuman chromosome 11p15 encodes an imprinted untrans-lated RNA [90] H19 opposite tumor suppressor (HOTS)

Sarcoma 9

Table 5 In vivo animal models of RMS

Model TargetModel origin Reference

Inactivation Expression

DrosophilaPAX-FKHR Bipartite Gal4-UAS

expression systemGalindo et al2006 [49]

PAX7-FOXO1 rols Chromosomal deletionDf(3L)vin5

Avirneni-Vadlamudi et al2012 [50]

Zebrafishrag2 promoter c-Myc kRASG12D

Chen andLangenau 2011[51]

MAPKERK andAKTS6K1 PD98059 TPCK Tg(hsp70-HRASG12V) Le et al 2012

[52]

Mouse

Sufu N-myc Sfrp1 Ptch2and cyclin D1 Sufu+minus Lee et al 2007

[53]

Ink4aArf and Trp53 Pax3FKHR Pax3P3Fpwt Keller et al2004 [54]

Wnt120573-cateninsignaling pathway Wnt2 p53minusminusc-fosminusminus Singh et al 2010

[55]

a tumor growth inhibitor is encoded by an imprinted H19antisense transcript Overexpression of HOTS inhibits RMStumor cell growth Silencing HOTS by RNAi was shown toincrease in vitro colony formation as well as in vivo tumorgrowth of RMS [90]

6 In Vivo Models of RMS

Several in vivo models have recently been developed inDrosophila zebrafish and mice to further understand themolecular and cellular biology of RMS tumorigenesis [49ndash55 91ndash94] (Table 5)

61 Drosophila Models in RMS Despite the evolutionarydistance a strong conservation of genes pathways andregulatory molecular networks have been demonstratedbetween flies and humans Many human disease genes haverelated sequences in Drosophila In contrast to mammalianmodels the generation of Drosophila mutants is easy cheapand fast

For a better understanding of the pathogenic conse-quences of PAX-FKHR expression Drosophila was chosenbecause the animalrsquos transparent outer cuticle allows forsimple real-time monitoring of muscle abnormalities elicitedby PAX-FKHR including subtle or focal changes [49] Themutation in the myoblast fusion gene rolling pebbles (rols)was reported to dominantly suppress PAX-FKHR1-inducedlethality by using a Drosophila model of PAX-FKHR1-mediated transformation [50] PAX-FKHR1 signaling upreg-ulates the TANC1 gene and blocks myoblasts from terminaldifferentiation However downregulating the TANC1 genecould cause RMS cells to lose their neoplastic state undergofusion and form differentiation This novel finding uncov-ered a PAX-FKHR1-TANC1 neoplasia axis collaboratively ina Drosophila model and loss-gain-of-function studies inmammalian platforms [50]

62 Zebrafish Models in RMS In contrast to other mod-els the short tumor onset in zebrafish allows for rapididentification of essential genes in various processes suchas tumor growth self-renewal and maintenance There areother advantages for using zebrafish in the study of humandisease and development including the ease and low cost ofraising large numbers of fish and the highly conserved geneticand biochemical pathways between zebrafish and mammals[95]

As zebrafish RMS is highly similar to human ERMSby using fluorescent transgenic approaches it is more con-venient to understand histogenesis and different aspects oftumorigenesis in RMS A robust zebrafish model of RAS-induced RMS has been established sharing twomorphologicand immunophenotypic features resembling ERMS [51] Inone case both are associated with tissue-restricted geneexpression in RMS while in another both comprise a RAS-induced gene signature Cross-species microarray compar-isons confirm that conserved genetic tumor-specific andtissue-restricted pathways such as RAS and p53 pathwaysdrive RMS growth Tg(hsp70-HRASG12V) zebrafish embryoswere generated to evaluate gene expression that mimics RASpathway activation during tumorigenesis to defineRAS targetgenes [52] Another KRASG12D-induced zebrafish embryonalRMS was utilized to assess the therapeutic effects By theblockage of two major downstream signaling pathwaysMAPKERK and AKTS6K1 the model showed that inhibi-tion of translation initiation suppresses tumor cell prolifera-tion [52]

All the above zebrafish cancer models share similar cellu-lar features and molecular pathways with human RMS espe-cially ERMS and can demonstrate a response to therapiesin a similar manner with human RMS Thus a tremendousopportunity is available to implement these animal modelsinto different steps for novel targeted therapeutic develop-ment

10 Sarcoma

63 Mouse Models in RMS Mouse models established fromin vivo studies may contribute to the understanding of thegenetic basis in tumor development and progression andmayhelp to test the efficacies of novel antineoplastic agents

The importance of PAX3PAX7-FKHR fusion proteins inthe progression of ARMS tumorigenesis is evident from bothin vitro and in vivo studies Transgenic mouse models gener-ated with the PAX-FKHR fusion genemimic the formation ofhuman ARMS [67] A mouse model was developed with het-erozygous loss of Sufu a tumor suppressor inHedgehog (Hh)signaling combined with p53 loss driven ERMS with a lowpenetrance (9) [53] These studies indicate that the modelsconsistently formingARMSnot only need the introduction ofthe PAX-FKHR fusion gene but also need to be accompaniedby other genetic events such as p53 pathway disruption [54]

Compared with ARMS ERMS models are more complexto generate because of their lower tumor penetrance andlonger latency JW41 cells from the p53c-fos double mutantmice models resemble human ERMS cells morphologicallyand express similar characteristic markers [55] The overex-pression of the Wnt2 gene identified in JW41 cells confirmedin human RMS cell lines can allow for further analysis of theWnt signaling pathway Both of these results indicate that thedownregulation of theWnt pathway contributes to resistanceto apoptosis and the inhibition of myogenic differentiation inERMS

Using Ptch1 p53 andor Rb1 conditional mouse modelsand controlling prenatal or postnatal myogenic cell of originRubin et al demonstrated that the loss of p53 in maturingmyoblasts related to tumorigenesis of ERMS [96] They alsoindicated that Rb1 alteration with other oncogenic factorswas strongly associated with an undifferentiated phenotypeacting as a modifier In addition they highlighted a sub-set of p53-deficient ERMSs arising from Myf6-expressingmyoblasts which had the same latency with Pax7-expressingmurine satellite cells in the study

7 Potential Therapeutic Targets in RMS

Detection of genomic imbalances and identification of thecrucial effective genes can contribute to identifying novelpotential biomarkers and relevant targets for clinical therapyin RMS

Survivin-responsive conditionally replicating adenovi-ruses can regulate multiple factors (Survm-CRAs) [41]Although Survm-CRA can efficiently replicate and potentlyinduce cell death in RMS cells the cytotoxic effects are morepronounced in RSC-enriched or RSC-purified cells thanin RSC-exiguous or progeny-purified cells Injections ofSurvm-CRAs into tumor nodules generated by transplantingRSC-enriched cells induce significant death in RMS cells andregression of tumor nodulesThe unique therapeutic featuresof Survm-CRA include not only its therapeutic effectivenessagainst RMS but also its increasing effectiveness against CSCsuggesting that Survm-CRA may be a promising anticanceragent [41]

Rapamycin an inhibitor of mTOR can abrogate RMStumor growth in a xenograft mouse model [97] Thetumors in the rapamycin-treatedmice group show significant

reduction of proliferation and invasiveness and inductionof apoptosis The tumor growth inhibition is simultaneousassociated with the diminution of mTOR and Hh pathwayswhich are implicated in the pathogenesis of RMSThe resultsindicate that using rapamycin either alone or in combinationwith traditional chemotherapeutic drugs may represent apotential targeted agent for therapeutic intervention Ampli-fication and mutational activation of Fibroblast Growth Fac-tor Receptor 4 (FGFR4) in RMS cells promote tumor pro-gression [98] Ponatinib is the most potent FGFR4 inhibitorto block wild-type RMS cell growth mutate FGFR4 throughincreasing apoptosis and suppress the FGFR4 downstreamtarget STAT3 Ponatinib treatment slows tumor growth inRMS mouse models expressing mutated FGFR4 [98] Pona-tinib as an FGFR4 inhibitor shows potential to act as aneffective therapeutic agent for RMS tumorsThe expression ofCXC chemokine receptor 4 (CXCR4) CXCR7 and VascularEndothelial Growth Factor (VEGF) indicates poor prognosisin a variety of malignancies including RMS cell lines Mostsamples in a large series of clinical RMS cases show asso-ciation with high expression of these antagonists regardlessof their subtypes [99] However there are significant corre-lations with high expression of CXCR4 and VEGF in bothERMS and ARMS High VEGF expression is predictive ofadverse prognostic factors in RMS as the expression of theseangiogenesis factors was compared with clinicopathologicalparameters and prognosis Considering their overexpressionin RMS these chemokine receptors and VEGF could providepotential molecular therapeutic targets in RMS Some ARMScell lines have undergone apoptosis in response to antineo-plastic drugs such as bortezomib The proapoptotic BH3only family member NOXA acts as an upregulated proteindownstream PAX3-FKHR resulting in increased cell sensi-tivity to bortezomib Apoptosis in response to bortezomibcan be reversed by shRNA knockdown of Noxa PAX3-FKHR upregulation of Noxa creates a potential therapeuticinsight into inducing apoptosis in ARMS cellsThis apoptosispathway could represent a specific targeted therapy againstPAX3-FKHR-expressing ARMS cells [100]

Proton pump activation resulting from intracellular acid-ificationmay be an energetic mechanism to escape apoptosisV-ATPase is an ATP-driven proton pump acidifying theintracellular compartment and transports protons across theplasma membrane Esomeprazole as a V-ATPase inhibitor inthe PPI administration interferes with intensive ion trans-porter activity of RMS cells and induces remarkable cytotoxi-city RMS expresses a higher level ofV-ATPase comparedwithother sarcomasTherefore RMS should be very susceptible toesomeprazole treatment and V-ATPase could be consideredas a promising selective target for treatment [101]

8 Conclusion and Future Prospects

For RMS the current challenges include how to iden-tify promising novel therapeutics and integrate them intoexisting therapy For better understanding of congenitaland epigenetic modifications in the development of RMSthis review summarizes the recent genome-wide studies onmolecular and genetic alterations to decipher the underlying

Sarcoma 11

tumorigenesis mechanisms There is a link between geneticand epigenetic alterations responsible for a variety of tumorcell growth proliferation differentiation apoptosis andtherapy-resistance mechanisms The identification of theprognostic value of the PAX-FKHR fusion status in RMSis one of the most important shifts in the lineage and therisk assessment of RMS which exerts an oncogenic effectthrough multiple pathways and is incorporated into mostrelevant studies The misbalanced expression of a number ofmiRs involved in the regulation of myogenic differentiationalso implicates the tumorsrsquo escape suppressive mechanismsMoreover we described several in vivo models of RMSto explore underlying histogenesis and different aspects oftumorigenesis Recent work on CSCs has also contributedto understanding the cell origins refining molecular char-acterization and elucidating the underlying basis of refrac-tory responses to current adjuvant treatments of subsets oftumorsThese integrative approaches can provide opportuni-ties to identify diagnostic and prognostic biomarkers appliedto the individual targeted therapy and significantly improveRMS prognosis

Abbreviations

RMS RhabdomyosarcomaARMS Alveolar rhabdomyosarcomaERMS Embryonal rhabdomyosarcomaPRMS Pleomorphic rhabdomyosarcomaSRMS Sclerosingspindle cell

rhabdomyosarcomamiRNA miR MicroRNACSC Cancer stem cellMSC Mesenchymal stem cell

Conflict of Interests

The authors declare that there is no conflict of interestsregarding the publication of this paper

Acknowledgments

This work was supported in part by grants from the Gat-tegno and Wechsler funds Dr Duan was supported in partthrough a grant from Sarcoma Foundation of America (SFA)a grant from National Cancer Institute (NCI)National Insti-tutes of Health (NIH) UO1 CA 151452 and a grant from anAcademic Enrichment Fund of MGH Orthopedic Surgery

References

[1] L Yang T Takimoto and J Fujimoto ldquoPrognostic model forpredicting overall survival in children and adolescents withrhabdomyosarcomardquo BMC Cancer vol 14 no 1 article 6542014

[2] D Egas-Bejar and W W Huh ldquoRhabdomyosarcoma in adoles-cent and young adult patients current perspectivesrdquo AdolescentHealth Medicine andTherapeutics vol 5 pp 115ndash125 2014

[3] J C Breneman E Lyden A S Pappo et al ldquoPrognosticfactors and clinical outcomes in children and adolescents withmetastatic rhabdomyosarcomamdasha report from the intergrouprhabdomyosarcoma study IVrdquo Journal of Clinical Oncology vol21 no 1 pp 78ndash84 2003

[4] V D Soleimani and M A Rudnicki ldquoNew insights into theorigin and the genetic basis of rhabdomyosarcomasrdquo CancerCell vol 19 no 2 pp 157ndash159 2011

[5] T Glass D D Cochrane S R Rassekh K Goddard and JHukin ldquoGrowing teratoma syndrome in intracranial non-ger-minomatous germ cell tumors (iNGGCTs) a risk for secondarymalignant transformationmdasha report of two casesrdquo Childrsquos Ner-vous System vol 30 no 5 pp 953ndash957 2014

[6] J Ji C Eng and K Hemminki ldquoFamilial risk for soft tissuetumors a nation-wide epidemiological study from SwedenrdquoJournal of Cancer Research and Clinical Oncology vol 134 no5 pp 617ndash624 2008

[7] C D M Fletcher J A Bridge P Hogendoorn and F MertensWHO Classification of Tumours of Soft Tissue and Bone vol 5IARC Press Paris France 4th edition 2013

[8] I Sultan I Qaddoumi S Yaser C Rodriguez-Galindo and AFerrari ldquoComparing adult and pediatric rhabdomyosarcoma inthe surveillance epidemiology and end results program 1973 to2005 an analysis of 2600 patientsrdquo Journal of Clinical Oncologyvol 27 no 20 pp 3391ndash3397 2009

[9] B Raney W Huh D Hawkins et al ldquoOutcome of patientswith localized orbital sarcoma who relapsed following treat-ment on Intergroup Rhabdomyosarcoma Study Group (IRSG)Protocols-III and -IV 1984ndash1997 a report from the ChildrenrsquosOncology Grouprdquo Pediatric Blood and Cancer vol 60 no 3 pp371ndash376 2013

[10] W Crist E A Gehan A H Ragab et al ldquoThe third intergrouprhabdomyosarcoma studyrdquo Journal of Clinical Oncology vol 13no 3 pp 610ndash630 1995

[11] C Liu D Li J Jiang et al ldquoAnalysis of molecular cytogeneticalteration in rhabdomyosarcoma by array comparative genomichybridizationrdquo PLoS ONE vol 9 no 4 Article ID e94924 2014

[12] DWilliamson E Missiaglia A de Reynies et al ldquoFusion gene-negative alveolar rhabdomyosarcoma is clinically and molec-ularly indistinguishable from embryonal rhabdomyosarcomardquoJournal of Clinical Oncology vol 28 no 13 pp 2151ndash2158 2010

[13] V Paulson G Chandler D Rakheja et al ldquoHigh-resolutionarray CGH identifies common mechanisms that drive embry-onal rhabdomyosarcoma pathogenesisrdquo Genes Chromosomesand Cancer vol 50 no 6 pp 397ndash408 2011

[14] S Park J Lee I-G Do et al ldquoAberrant CDK4 amplificationin refractory rhabdomyosarcoma as identified by genomicprofilingrdquo Scientific Reports vol 4 article 3623 2014

[15] C Liu D Li J Hu et al ldquoChromosomal and genetic imbalancesin Chinese patients with rhabdomyosarcoma detected by high-resolution array comparative genomic hybridizationrdquo Interna-tional Journal of Clinical and Experimental Pathology vol 7 no2 pp 690ndash698 2014

[16] Q-X Li C-X Liu C-P Chun et al ldquoChromosomal imbalancesrevealed in primary rhabdomyo-sarcomas by comparativegenomic hybridizationrdquo Chinese Medical Journal vol 122 no11 pp 1277ndash1282 2009

[17] E Missiaglia J Selfe M Hamdi et al ldquoGenomic imbalancesin rhabdomyosarcoma cell lines affect expression of genesfrequently altered in primary tumors an approach to identifycandidate genes involved in tumor developmentrdquo Genes Chro-mosomes and Cancer vol 48 no 6 pp 455ndash467 2009

12 Sarcoma

[18] F G Barr F Duan L M Smith et al ldquoGenomic and clinicalanalyses of 2p24 and 12q13-q14 amplification in alveolar rhab-domyosarcoma a report from the childrenrsquos oncology grouprdquoGenes Chromosomes and Cancer vol 48 no 8 pp 661ndash6722009

[19] T Gordon AMcManus J Anderson et al ldquoCytogenetic abnor-malities in 42 rhabdomyosarcoma a United Kingdom CancerCytogenetics Group StudyrdquoMedical and Pediatric Oncology vol36 no 2 pp 259ndash267 2001

[20] X Chen E Stewart A A Shelat et al ldquoTargeting oxidativestress in embryonal rhabdomyosarcomardquo Cancer Cell vol 24no 6 pp 710ndash724 2013

[21] S Lizard-Nacol F Mugneret C Volk C Turc-Carel M Favrotand T Philip ldquoTranslocation (213)(q37q14) in alveolar rhab-domyosarcoma a new caserdquo Cancer Genetics and Cytogeneticsvol 25 no 2 pp 373ndash374 1987

[22] P H B Sorensen J C Lynch S J Qualman et al ldquoPAX3-FKHR and PAX7-FKHR gene fusions are prognostic indicatorsin alveolar rhabdomyosarcoma a report from the childrenrsquosoncology grouprdquo Journal of Clinical Oncology vol 20 no 11 pp2672ndash2679 2002

[23] A D Marshall and G C Grosveld ldquoAlveolar rhabdomyo-sarcomamdashthe molecular drivers of PAX37-FOXO1-inducedtumorigenesisrdquo Skeletal Muscle vol 2 no 1 article 25 2012

[24] E Missiaglia D Williamson J Chisholm et al ldquoPAX3FOXO1fusion gene status is the key prognostic molecular markerin rhabdomyosarcoma and significantly improves current riskstratificationrdquo Journal of Clinical Oncology vol 30 no 14 pp1670ndash1677 2012

[25] J F Shern L Chen J Chmielecki et al ldquoComprehensivegenomic analysis of rhabdomyosarcoma reveals a landscape ofalterations affecting a common genetic axis in fusion-positiveand fusion-negative tumorsrdquo Cancer Discovery vol 4 no 2 pp216ndash231 2014

[26] MWachtel M Dettling E Koscielniak et al ldquoGene expressionsignatures identify rhabdomyosarcoma subtypes and detect anovel t(22)(q35p23) translocation fusing PAX3 to NCOA1rdquoCancer Research vol 64 no 16 pp 5539ndash5545 2004

[27] E Davicioni M J Anderson F G Finckenstein et al ldquoMolec-ular classification of rhabdomyosarcomamdashgenotypic and phe-notypic determinants of diagnosis a report from the ChildrenrsquosOncology Grouprdquo The American Journal of Pathology vol 174no 2 pp 550ndash564 2009

[28] M Visser C Sijmons J Bras et al ldquoAllelotype of pediatric rhab-domyosarcomardquo Oncogene vol 15 no 11 pp 1309ndash1314 1997

[29] R Gil-Benso T San-Miguel R C Callaghan et al ldquoChromo-somal and genetic changes produced in tumoral progression ofembryonal rhabdomyosarcomardquo Histopathology vol 62 no 5pp 816ndash819 2013

[30] S X Skapek J Anderson F G Barr et al ldquoPAX-FOXO1 fusionstatus drives unfavorable outcome for children with rhabdom-yosarcoma a childrenrsquos oncology group reportrdquo Pediatric Bloodand Cancer vol 60 no 9 pp 1411ndash1417 2013

[31] HHosoi N Kakazu E Konishi et al ldquoA novel PAX3 rearrange-ment in embryonal rhabdomyosarcomardquo Cancer Genetics andCytogenetics vol 189 no 2 pp 98ndash104 2009

[32] E Takaoka H Sonobe K Akimaru et al ldquoMultiple sites ofhighly amplified DNA sequences detected bymolecular cytoge-netic analysis inHS-RMS-2 a newpleomorphic rhabdomyosar-coma cell linerdquo American Journal of Cancer Research vol 2 no2 pp 141ndash152 2012

[33] K Szuhai D de Jong W Y Leung C D M Fletcher and P CWHogendoorn ldquoTransactivatingmutation of theMYOD1 geneis a frequent event in adult spindle cell rhabdomyosarcomardquoJournal of Pathology vol 232 no 3 pp 300ndash307 2014

[34] N P Agaram C-L Chen L Zhang M P Laquaglia L Wexlerand C R Antonescu ldquoRecurrent MYOD1 mutations in pedi-atric and adult sclerosing and spindle cell rhabdomyosarcomasEvidence for a common pathogenesisrdquoGenes Chromosomes andCancer vol 53 no 9 pp 779ndash787 2014

[35] H Yoshida M Miyachi K Sakamoto et al ldquoPAX3-NCOA2fusion gene has a dual role in promoting the proliferation andinhibiting the myogenic differentiation of rhabdomyosarcomacellsrdquo Oncogene vol 33 pp 5601ndash5608 2013

[36] J M Mosquera A Sboner L Zhang et al ldquoRecurrent NCOA2gene rearrangements in congenitalinfantile spindle cell rhab-domyosarcomardquo Genes Chromosomes and Cancer vol 52 no6 pp 538ndash550 2013

[37] V Thavaraj A Sethi and L S Arya ldquoIncomplete Beckwith-Wiedemann syndrome in a child with orbital rhabdomyosar-comardquo Indian Pediatrics vol 39 no 3 pp 299ndash304 2002

[38] D Walter S Satheesha P Albrecht et al ldquoCD133 positiveembryonal rhabdomyosarcoma stem-like cell population isenriched in rhabdospheresrdquo PLoS ONE vol 6 no 5 Article IDe19506 2011

[39] Y-X Ren F G Finckenstein D A Abdueva et al ldquoMousemesenchymal stem cells expressing PAX-FKHR form alveolarrhabdomyosarcomas by cooperating with secondary muta-tionsrdquo Cancer Research vol 68 no 16 pp 6587ndash6597 2008

[40] M Salerno S Avnet G Bonuccelli S Hosogi D Granchi andN Baldini ldquoImpairment of lysosomal activity as a therapeuticmodality targeting cancer stem cells of embryonal rhabdom-yosarcoma cell line RDrdquo PLoS ONE vol 9 no 10 Article IDe110340 2014

[41] K Tanoue Y Wang M Ikeda et al ldquoSurvivin-responsive con-ditionally replicating adenovirus kills rhabdomyosarcoma stemcells more efficiently than their progenyrdquo Journal of Transla-tional Medicine vol 12 no 1 article 27 2014

[42] P K Rao EMissiaglia L Shields et al ldquoDistinct roles formiR-1and miR-133a in the proliferation and differentiation of rhab-domyosarcoma cellsrdquo The FASEB Journal vol 24 no 9 pp3427ndash3437 2010

[43] D Yan X D Dong X Chen et al ldquoMicroRNA-1206 targets c-met and inhibits rhabdomyosarcomadevelopmentrdquoThe Journalof Biological Chemistry vol 284 no 43 pp 29596ndash29604 2009

[44] I Marchesi A Giordano and L Bagella ldquoRoles of enhancerof zeste homolog 2 from skeletal muscle differentiation torhabdomyosarcoma carcinogenesisrdquoCell Cycle vol 13 no 4 pp516ndash527 2014

[45] M M Sun J F Li L L Guo et al ldquoTGF-1205731 suppression ofmicroRNA-450b-5p expression a novel mechanism for block-ingmyogenic differentiation of rhabdomyosarcomardquoOncogenevol 33 no 16 pp 2075ndash2086 2014

[46] Y Diao X Guo L Jiang et al ldquoMiR-203 a tumor suppressorfrequently down-regulated by promoter hypermethylation inrhabdomyosarcomardquo The Journal of Biological Chemistry vol289 no 1 pp 529ndash539 2014

[47] H-J Huang J Liu HHua et al ldquoMiR-214 andN-ras regulatoryloop suppresses rhabdomyosarcoma cell growth and xenografttumorigenesisrdquo Oncotarget vol 5 no 8 pp 2161ndash2175 2014

Sarcoma 13

[48] A L Sarver L Li and S Subramanian ldquomicroRNA miR-183functions as an oncogene by targeting the transcription factorEGR1 and promoting tumor cell migrationrdquo Cancer Researchvol 70 no 23 pp 9570ndash9580 2010

[49] R L Galindo J A Allport and E N Olson ldquoA Drosophilamodel of the rhabdomyosarcoma initiator PAX7-FKHRrdquo Pro-ceedings of the National Academy of Sciences of the United Statesof America vol 103 no 36 pp 13439ndash13444 2006

[50] U Avirneni-Vadlamudi K A Galindo T R Endicott V Paul-son S Cameron and R L Galindo ldquoDrosophila and mammal-ian models uncover a role for the myoblast fusion gene TANC1in rhabdomyosarcomardquo The Journal of Clinical Investigationvol 122 no 1 pp 403ndash407 2012

[51] E Y Chen andDM Langenau ldquoZebrafishmodels of rhabdom-yosarcomardquoMethods in Cell Biology vol 105 pp 383ndash402 2011

[52] X Le E K Pugach S Hettmer et al ldquoA novel chemicalscreening strategy in zebrafish identifies common pathways inembryogenesis and rhabdomyosarcoma developmentrdquo Devel-opment vol 140 no 11 pp 2354ndash2364 2012

[53] Y Lee R Kawagoe K Sasai et al ldquoLoss of suppressor-of-fusedfunction promotes tumorigenesisrdquoOncogene vol 26 no 44 pp6442ndash6447 2007

[54] C Keller B R Arenkiel C M Coffin N El-Bardeesy R ADePinho and M R Capecchi ldquoAlveolar rhabdomyosarcomasin conditional Pax3Fkhr mice cooperativity of Ink4aARF andTrp53 loss of functionrdquo Genes and Development vol 18 no 21pp 2614ndash2626 2004

[55] S Singh CVinson CMGurley et al ldquoImpairedWnt signalingin embryonal rhabdomyosarcoma cells from p53c-fos doublemutant micerdquo American Journal of Pathology vol 177 no 4 pp2055ndash2066 2010

[56] G J Schaaf J M Ruijter F Van Ruissen et al ldquoFull transcrip-tome analysis of rhabdomyosarcoma normal and fetal skeletalmuscle statistical comparison of multiple SAGE librariesrdquo TheFASEB Journal vol 19 no 3 pp 404ndash406 2005

[57] D S Hawkins A A Gupta and E R Rudzinski ldquoWhat is newin the biology and treatment of pediatric rhabdomyosarcomardquoCurrent Opinion in Pediatrics vol 26 no 1 pp 50ndash56 2014

[58] M Zhang J Truscott and J Davie ldquoLoss of MEF2D expressioninhibits differentiation and contributes to oncogenesis in rhab-domyosarcoma cellsrdquoMolecular Cancer vol 12 no 1 article 1502013

[59] M Ciesla J Dulak and A Jozkowicz ldquomicroRNAs and epi-genetic mechanisms of rhabdomyosarcoma developmentrdquo TheInternational Journal of Biochemistry amp Cell Biology vol 53 pp482ndash492 2014

[60] S Ricaud B Vernus M Duclos et al ldquoInhibition of autocrinesecretion of myostatin enhances terminal differentiation inhuman rhabdomyosarcoma cellsrdquo Oncogene vol 22 no 51 pp8221ndash8232 2003

[61] C S Guo C Degnin T A Fiddler D Stauffer andM JThayerldquoRegulation of MyoD activity and muscle cell differentiation byMDM2 pRb and Sp1rdquoThe Journal of Biological Chemistry vol278 no 25 pp 22615ndash22622 2003

[62] S Rossi E Stoppani P L Puri and A Fanzani ldquoDifferenti-ation of human rhabdomyosarcoma RD cells is regulated byreciprocal functional interactions between myostatin p38 andextracellular regulated kinase signalling pathwaysrdquo EuropeanJournal of Cancer vol 47 no 7 pp 1095ndash1105 2011

[63] Z S Walters B Villarejo-Balcells D Olmos et al ldquoJARID2is a direct target of the PAX3-FOXO1 fusion protein andinhibits myogenic differentiation of rhabdomyosarcoma cellsrdquoOncogene vol 33 no 9 pp 1148ndash1157 2014

[64] T Hosoyama M I Aslam J Abraham et al ldquoIL-4R drives ded-ifferentiation mitogenesis and metastasis in rhabdomyosar-comardquo Clinical Cancer Research vol 17 no 9 pp 2757ndash27662011

[65] F S Dela Cruz ldquoCancer stem cells in pediatric sarcomasrdquoFrontiers in Oncology vol 3 article 168 2013

[66] M J Bissell and W C Hines ldquoWhy donrsquot we get more cancerA proposed role of the microenvironment in restraining cancerprogressionrdquo Nature Medicine vol 17 no 3 pp 320ndash329 2011

[67] E Charytonowicz C Cordon-Cardo I Matushansky and MZiman ldquoAlveolar rhabdomyosarcoma is the cell of origin amesenchymal stem cellrdquoCancer Letters vol 279 no 2 pp 126ndash136 2009

[68] W K Cavenee ldquoMuscling in on rhabdomyosarcomardquo NatureMedicine vol 8 no 11 pp 1200ndash1201 2002

[69] B Zhu and J K Davie ldquoNew insights into signalling-pathwayalterations in rhabdomyosarcomardquo British Journal of Cancervol 112 no 2 pp 227ndash231 2015

[70] M Zhang C M Linardic and D G Kirsch ldquoRAS and ROS inrhabdomyosarcomardquo Cancer Cell vol 24 no 6 pp 689ndash6912013

[71] S C Marianna L Ianzano and G Nicoletti ldquoTumor sup-pressor genes promote rhabdomyosarcoma progression in p53heterozygousrdquo Oncotarget vol 5 no 1 pp 108ndash119 2013

[72] SWang L Guo L Dong et al ldquoTGF-beta1 signal pathwaymaycontribute to rhabdomyosarcoma development by inhibitingdifferentiationrdquo Cancer Science vol 101 no 5 pp 1108ndash11162010

[73] J Wesche K Haglund and E M Haugsten ldquoFibroblast growthfactors and their receptors in cancerrdquo Biochemical Journal vol437 no 2 pp 199ndash213 2011

[74] M Wachtel J Rakic M Okoniewski P Bode F Niggli andB W Schafer ldquoFGFR4 signaling couples to Bim and not Bmfto discriminate subsets of alveolar rhabdomyosarcoma cellsrdquoInternational Journal of Cancer vol 135 no 7 pp 1543ndash15522014

[75] P L Puri ZWu P Zhang et al ldquoInduction of terminal differen-tiation by constitutive activation of p38 MAP kinase in humanrhabdomyosarcoma cellsrdquo Genes and Development vol 14 no5 pp 574ndash584 2000

[76] A M Tremblay E Missiaglia G G Galli et al ldquoThe Hippotransducer YAP1 transforms activated satellite cells and is apotent effector of embryonal rhabdomyosarcoma formationrdquoCancer Cell vol 26 no 2 pp 273ndash287 2014

[77] R N Judson A M Tremblay P Knopp et al ldquoThe hippo path-way member Yap plays a key role in influencing fate decisionsin muscle satellite cellsrdquo Journal of Cell Science vol 125 no 24pp 6009ndash6019 2012

[78] L E S Crose K A Galindo J G Kephart et al ldquoAlve-olar rhabdomyosarcoma-associated PAX3-FOXO1 promotestumorigenesis via Hippo pathway suppressionrdquo Journal ofClinical Investigation vol 124 no 1 pp 285ndash296 2014

[79] B Belyea J G Kephart J Blum D G Kirsch and C MLinardic ldquoEmbryonic signaling pathways and rhabdomyosar-coma contributions to cancer development and opportunitiesfor therapeutic targetingrdquo Sarcoma vol 2012 Article ID 40623913 pages 2012

14 Sarcoma

[80] J Huertas-Martinez S Rello-Varona D Herrero-Martin et alldquoCaveolin-1 is down-regulated in alveolar rhabdomyosarcomasand negatively regulates tumor growthrdquo Oncotarget vol 5 no20 pp 9744ndash9755 2014

[81] S Rossi P L Poliani CMissale EMonti andA Fanzani ldquoCav-eolins in rhabdomyosarcomardquo Journal of Cellular andMolecularMedicine vol 15 no 12 pp 2553ndash2568 2011

[82] P G Tsioli E S Patsouris C Giaginis and S E TheocharisldquoDNA repair systems in rhabdomyosarcomardquo Histology andHistopathology vol 28 no 8 pp 971ndash984 2013

[83] E Gatzidou C Michailidi S Tseleni-Balafouta and S Theo-charis ldquoAn epitome of DNA repair related genes and mecha-nisms in thyroid carcinomardquo Cancer Letters vol 290 no 2 pp139ndash147 2010

[84] R Rota R Ciarapica A Giordano L Miele and F LocatellildquoMicroRNAs in rhabdomyosarcoma pathogenetic implicationsand translational potentialityrdquoMolecular Cancer vol 10 article120 2011

[85] A Gougelet J Perez D Pissaloux et al ldquoMiRNA profiling howto bypass the current difficulties in the diagnosis and treatmentof sarcomasrdquo Sarcoma vol 2011 Article ID 460650 13 pages2011

[86] R Ciarapica G Russo F Verginelli et al ldquoDeregulated expres-sion of miR-26a and Ezh2 in rhabdomyosarcomardquo Cell Cyclevol 8 no 1 pp 172ndash175 2009

[87] L Li A L Sarver S Alamgir and S Subramanian ldquoDownreg-ulation of microRNAs miR-1 -206 and -29 stabilizes PAX3 andCCND2 expression in rhabdomyosarcomardquo Laboratory Investi-gation vol 92 no 4 pp 571ndash583 2012

[88] S-Y Ng G K Bogu B Soh and L W Stanton ldquoThe longnoncoding RNA RMST interacts with SOX2 to regulate neu-rogenesisrdquoMolecular Cell vol 51 no 3 pp 349ndash359 2013

[89] C A Lynch B Tycko T H Bestor and C P Walsh ldquoReac-tivation of a silenced H19 gene in human rhabdomyosarcomaby demethylation of DNA but not by histone hyperacetylationrdquoMolecular Cancer vol 1 article 2 2002

[90] P Onyango and A P Feinberg ldquoA nucleolar protein H19opposite tumor suppressor (HOTS) is a tumor growth inhibitorencoded by a human imprinted H19 antisense transcriptrdquoProceedings of the National Academy of Sciences of the UnitedStates of America vol 108 no 40 pp 16759ndash16764 2011

[91] C E Albacker N Y Storer E M Langdon et al ldquoThe his-tone methyltransferase SUV39H1 suppresses embryonal rhab-domyosarcoma formation in zebrafishrdquo PLoS ONE vol 8 no 5Article ID e64969 2013

[92] N Y Storer R M White A Uong et al ldquoZebrafish rhab-domyosarcoma reflects the developmental stage of oncogeneexpression during myogenesisrdquo Development vol 140 no 14pp 3040ndash3050 2013

[93] M E Hatley W Tang M R Garcia et al ldquoA mouse modelof rhabdomyosarcoma originating from the adipocyte lineagerdquoCancer Cell vol 22 no 4 pp 536ndash546 2012

[94] V Hosur A Kavirayani J Riefler et al ldquoDystrophin and dysfer-lin doublemutantmice a novelmodel for rhabdomyosarcomardquoCancer Genetics vol 205 no 5 pp 232ndash241 2012

[95] W Goessling T E North and L I Zon ldquoNew waves of discov-ery modeling cancer in zebrafishrdquo Journal of Clinical Oncologyvol 25 no 17 pp 2473ndash2479 2007

[96] B P Rubin K Nishijo H-I H Chen et al ldquoEvidence for anunanticipated relationship between undifferentiated pleomor-phic sarcoma and embryonal rhabdomyosarcomardquo Cancer Cellvol 19 no 2 pp 177ndash191 2011

[97] S Z Kaylani J Xu R K Srivastava L Kopelovich J G Presseyand M Athar ldquoRapamycin targeting mTOR and hedgehog sig-naling pathways blocks human rhabdomyosarcoma growth inxenograft murine modelrdquo Biochemical and Biophysical ResearchCommunications vol 435 no 4 pp 557ndash561 2013

[98] S Q Li A T Cheuk J F Shern et al ldquoTargeting wild-type andmutationally activated FGFR4 in rhabdomyosarcoma with theinhibitor ponatinib (AP24534)rdquoPLoSONE vol 8 no 10 ArticleID e76551 2013

[99] K Miyoshi K Kohashi F Fushimi et al ldquoClose correlationbetween CXCR4 and VEGF expression and frequent CXCR7expression in rhabdomyosarcomardquo Human Pathology vol 45no 9 pp 1900ndash1909 2014

[100] ADMarshall F Picchione R I KGeltink andGCGrosveldldquoPAX3-FOXO1 induces up-regulation of Noxa sensitizing alve-olar rhabdomyosarcoma cells to apoptosisrdquo Neoplasia vol 15no 7 pp 738ndash748 2013

[101] F Perut S Avnet C Fotia et al ldquoV-ATPase as an effective thera-peutic target for sarcomasrdquo Experimental Cell Research vol 320no 1 pp 21ndash32 2014

Submit your manuscripts athttpwwwhindawicom

Stem CellsInternational

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

MEDIATORSINFLAMMATION

of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Behavioural Neurology

EndocrinologyInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Disease Markers

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

BioMed Research International

OncologyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Oxidative Medicine and Cellular Longevity

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

PPAR Research

The Scientific World JournalHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Immunology ResearchHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal of

ObesityJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Computational and Mathematical Methods in Medicine

OphthalmologyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Diabetes ResearchJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Research and TreatmentAIDS

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Gastroenterology Research and Practice

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Parkinsonrsquos Disease

Evidence-Based Complementary and Alternative Medicine

Volume 2014Hindawi Publishing Corporationhttpwwwhindawicom

Page 8: Review Article Rhabdomyosarcoma: Advances in Molecular and

8 Sarcoma

Table 4 MicroRNAs involved in myogenesis and RMS development

miRNA Expression Target Function Reference

miR-1 and miR-133a Downregulation MYH9Myogenic miRNA inhibit differentiationand promote proliferation in myogenesiscytostatic

Rao et al 2010 [42]

miR-206 Downregulation cMET Promote differentiation and proliferationin myogenesis

Yan et al 2009 [43]

miR-29 Downregulation HADC4 YY1 EZH2 Promote stabilization of RMS phenotype Marchesi et al 2014 [44]

miR-450-5p Downregulation ENOX PAX9 Promote differentiation and progression Sun et al 2014 [45]

miR-203 Downregulation JAK STAT Notch Inhibit differentiation and proliferation inmyogenesis tumor suppressor

Diao et al 2014 [46]

miR-214 Downregulation N-Ras Inhibit tumor cell growth and inducemyogenic differentiation and apoptosis

Huang et al 2014 [47]

miR-183 Upregulation EGR1 Promote migration and metastasis Sarver et al 2010 [48]

PMS2

MGMT

HR

Mismatch Recombination

NHEJ MMEJ

p53

Excision

BER NER

RMS

DNA repair

Direct repair Indirect repair

repair repairrepair

Figure 4The DNA repair systems in RMSThere are two pathways repairing the DNA lesions directly without affecting DNA structure andindirectly by DNA phosphodiester backbone cleavage The modifications in DNA repair enzymes expression or activity lead to resistanceto chemotherapy and radiation in RMS tumor cells MGMT O6-methylguanine-DNA methyltransferase BER base excision repair NERnucleotide excision repair HR homologous recombination NHEJ nonhomologous end-joining MMEJ microhomology-mediated end-joining

and miR-214 [44 46 47 84 87] They are all significantlydownregulated in RMS compared with normal skeletal mus-cles and function to inhibit tumor cell growth and inducemyogenic differentiation and apoptosis in RMS

52 Oncogenic miRNA in RMS The transcription factor EGR1is a tumor suppressor gene that is downregulated in RMS [48]miR-183 functions as an oncogene by targeting EGR1 and pro-moting tumor cell migration Either by direct anti-miR treat-ment or by indirect mechanisms that decrease transcriptmiR-183 targeted treatmentsmight provide a potential optionto RMS patients

The basic strategy of current effective miR-based treat-ment studies is either efficient reexpression of miRs or restor-ing the function of miRs to inhibit the expression of certain

protein-coding genes and promote muscle differentiationMoreover miR expression profiling in tumors and possiblytheir detection in peripheral blood during treatment may beable to predict the response to chemotherapy or radiotherapyand be useful as a prognostic signature for the development oftreatment resistanceThe prognostic value of miR expressionlevels in RMS is a powerful tool in creating a better-tailoredstrategy for particular subsets of patients

53 Long Noncoding RNAs in RMS Long noncoding RNAs(LNCRNAs) are abundant in the mammalian transcriptomeand have been shown to play a role in RMS development [8889]TheH19 gene localized within a chromosomal region onhuman chromosome 11p15 encodes an imprinted untrans-lated RNA [90] H19 opposite tumor suppressor (HOTS)

Sarcoma 9

Table 5 In vivo animal models of RMS

Model TargetModel origin Reference

Inactivation Expression

DrosophilaPAX-FKHR Bipartite Gal4-UAS

expression systemGalindo et al2006 [49]

PAX7-FOXO1 rols Chromosomal deletionDf(3L)vin5

Avirneni-Vadlamudi et al2012 [50]

Zebrafishrag2 promoter c-Myc kRASG12D

Chen andLangenau 2011[51]

MAPKERK andAKTS6K1 PD98059 TPCK Tg(hsp70-HRASG12V) Le et al 2012

[52]

Mouse

Sufu N-myc Sfrp1 Ptch2and cyclin D1 Sufu+minus Lee et al 2007

[53]

Ink4aArf and Trp53 Pax3FKHR Pax3P3Fpwt Keller et al2004 [54]

Wnt120573-cateninsignaling pathway Wnt2 p53minusminusc-fosminusminus Singh et al 2010

[55]

a tumor growth inhibitor is encoded by an imprinted H19antisense transcript Overexpression of HOTS inhibits RMStumor cell growth Silencing HOTS by RNAi was shown toincrease in vitro colony formation as well as in vivo tumorgrowth of RMS [90]

6 In Vivo Models of RMS

Several in vivo models have recently been developed inDrosophila zebrafish and mice to further understand themolecular and cellular biology of RMS tumorigenesis [49ndash55 91ndash94] (Table 5)

61 Drosophila Models in RMS Despite the evolutionarydistance a strong conservation of genes pathways andregulatory molecular networks have been demonstratedbetween flies and humans Many human disease genes haverelated sequences in Drosophila In contrast to mammalianmodels the generation of Drosophila mutants is easy cheapand fast

For a better understanding of the pathogenic conse-quences of PAX-FKHR expression Drosophila was chosenbecause the animalrsquos transparent outer cuticle allows forsimple real-time monitoring of muscle abnormalities elicitedby PAX-FKHR including subtle or focal changes [49] Themutation in the myoblast fusion gene rolling pebbles (rols)was reported to dominantly suppress PAX-FKHR1-inducedlethality by using a Drosophila model of PAX-FKHR1-mediated transformation [50] PAX-FKHR1 signaling upreg-ulates the TANC1 gene and blocks myoblasts from terminaldifferentiation However downregulating the TANC1 genecould cause RMS cells to lose their neoplastic state undergofusion and form differentiation This novel finding uncov-ered a PAX-FKHR1-TANC1 neoplasia axis collaboratively ina Drosophila model and loss-gain-of-function studies inmammalian platforms [50]

62 Zebrafish Models in RMS In contrast to other mod-els the short tumor onset in zebrafish allows for rapididentification of essential genes in various processes suchas tumor growth self-renewal and maintenance There areother advantages for using zebrafish in the study of humandisease and development including the ease and low cost ofraising large numbers of fish and the highly conserved geneticand biochemical pathways between zebrafish and mammals[95]

As zebrafish RMS is highly similar to human ERMSby using fluorescent transgenic approaches it is more con-venient to understand histogenesis and different aspects oftumorigenesis in RMS A robust zebrafish model of RAS-induced RMS has been established sharing twomorphologicand immunophenotypic features resembling ERMS [51] Inone case both are associated with tissue-restricted geneexpression in RMS while in another both comprise a RAS-induced gene signature Cross-species microarray compar-isons confirm that conserved genetic tumor-specific andtissue-restricted pathways such as RAS and p53 pathwaysdrive RMS growth Tg(hsp70-HRASG12V) zebrafish embryoswere generated to evaluate gene expression that mimics RASpathway activation during tumorigenesis to defineRAS targetgenes [52] Another KRASG12D-induced zebrafish embryonalRMS was utilized to assess the therapeutic effects By theblockage of two major downstream signaling pathwaysMAPKERK and AKTS6K1 the model showed that inhibi-tion of translation initiation suppresses tumor cell prolifera-tion [52]

All the above zebrafish cancer models share similar cellu-lar features and molecular pathways with human RMS espe-cially ERMS and can demonstrate a response to therapiesin a similar manner with human RMS Thus a tremendousopportunity is available to implement these animal modelsinto different steps for novel targeted therapeutic develop-ment

10 Sarcoma

63 Mouse Models in RMS Mouse models established fromin vivo studies may contribute to the understanding of thegenetic basis in tumor development and progression andmayhelp to test the efficacies of novel antineoplastic agents

The importance of PAX3PAX7-FKHR fusion proteins inthe progression of ARMS tumorigenesis is evident from bothin vitro and in vivo studies Transgenic mouse models gener-ated with the PAX-FKHR fusion genemimic the formation ofhuman ARMS [67] A mouse model was developed with het-erozygous loss of Sufu a tumor suppressor inHedgehog (Hh)signaling combined with p53 loss driven ERMS with a lowpenetrance (9) [53] These studies indicate that the modelsconsistently formingARMSnot only need the introduction ofthe PAX-FKHR fusion gene but also need to be accompaniedby other genetic events such as p53 pathway disruption [54]

Compared with ARMS ERMS models are more complexto generate because of their lower tumor penetrance andlonger latency JW41 cells from the p53c-fos double mutantmice models resemble human ERMS cells morphologicallyand express similar characteristic markers [55] The overex-pression of the Wnt2 gene identified in JW41 cells confirmedin human RMS cell lines can allow for further analysis of theWnt signaling pathway Both of these results indicate that thedownregulation of theWnt pathway contributes to resistanceto apoptosis and the inhibition of myogenic differentiation inERMS

Using Ptch1 p53 andor Rb1 conditional mouse modelsand controlling prenatal or postnatal myogenic cell of originRubin et al demonstrated that the loss of p53 in maturingmyoblasts related to tumorigenesis of ERMS [96] They alsoindicated that Rb1 alteration with other oncogenic factorswas strongly associated with an undifferentiated phenotypeacting as a modifier In addition they highlighted a sub-set of p53-deficient ERMSs arising from Myf6-expressingmyoblasts which had the same latency with Pax7-expressingmurine satellite cells in the study

7 Potential Therapeutic Targets in RMS

Detection of genomic imbalances and identification of thecrucial effective genes can contribute to identifying novelpotential biomarkers and relevant targets for clinical therapyin RMS

Survivin-responsive conditionally replicating adenovi-ruses can regulate multiple factors (Survm-CRAs) [41]Although Survm-CRA can efficiently replicate and potentlyinduce cell death in RMS cells the cytotoxic effects are morepronounced in RSC-enriched or RSC-purified cells thanin RSC-exiguous or progeny-purified cells Injections ofSurvm-CRAs into tumor nodules generated by transplantingRSC-enriched cells induce significant death in RMS cells andregression of tumor nodulesThe unique therapeutic featuresof Survm-CRA include not only its therapeutic effectivenessagainst RMS but also its increasing effectiveness against CSCsuggesting that Survm-CRA may be a promising anticanceragent [41]

Rapamycin an inhibitor of mTOR can abrogate RMStumor growth in a xenograft mouse model [97] Thetumors in the rapamycin-treatedmice group show significant

reduction of proliferation and invasiveness and inductionof apoptosis The tumor growth inhibition is simultaneousassociated with the diminution of mTOR and Hh pathwayswhich are implicated in the pathogenesis of RMSThe resultsindicate that using rapamycin either alone or in combinationwith traditional chemotherapeutic drugs may represent apotential targeted agent for therapeutic intervention Ampli-fication and mutational activation of Fibroblast Growth Fac-tor Receptor 4 (FGFR4) in RMS cells promote tumor pro-gression [98] Ponatinib is the most potent FGFR4 inhibitorto block wild-type RMS cell growth mutate FGFR4 throughincreasing apoptosis and suppress the FGFR4 downstreamtarget STAT3 Ponatinib treatment slows tumor growth inRMS mouse models expressing mutated FGFR4 [98] Pona-tinib as an FGFR4 inhibitor shows potential to act as aneffective therapeutic agent for RMS tumorsThe expression ofCXC chemokine receptor 4 (CXCR4) CXCR7 and VascularEndothelial Growth Factor (VEGF) indicates poor prognosisin a variety of malignancies including RMS cell lines Mostsamples in a large series of clinical RMS cases show asso-ciation with high expression of these antagonists regardlessof their subtypes [99] However there are significant corre-lations with high expression of CXCR4 and VEGF in bothERMS and ARMS High VEGF expression is predictive ofadverse prognostic factors in RMS as the expression of theseangiogenesis factors was compared with clinicopathologicalparameters and prognosis Considering their overexpressionin RMS these chemokine receptors and VEGF could providepotential molecular therapeutic targets in RMS Some ARMScell lines have undergone apoptosis in response to antineo-plastic drugs such as bortezomib The proapoptotic BH3only family member NOXA acts as an upregulated proteindownstream PAX3-FKHR resulting in increased cell sensi-tivity to bortezomib Apoptosis in response to bortezomibcan be reversed by shRNA knockdown of Noxa PAX3-FKHR upregulation of Noxa creates a potential therapeuticinsight into inducing apoptosis in ARMS cellsThis apoptosispathway could represent a specific targeted therapy againstPAX3-FKHR-expressing ARMS cells [100]

Proton pump activation resulting from intracellular acid-ificationmay be an energetic mechanism to escape apoptosisV-ATPase is an ATP-driven proton pump acidifying theintracellular compartment and transports protons across theplasma membrane Esomeprazole as a V-ATPase inhibitor inthe PPI administration interferes with intensive ion trans-porter activity of RMS cells and induces remarkable cytotoxi-city RMS expresses a higher level ofV-ATPase comparedwithother sarcomasTherefore RMS should be very susceptible toesomeprazole treatment and V-ATPase could be consideredas a promising selective target for treatment [101]

8 Conclusion and Future Prospects

For RMS the current challenges include how to iden-tify promising novel therapeutics and integrate them intoexisting therapy For better understanding of congenitaland epigenetic modifications in the development of RMSthis review summarizes the recent genome-wide studies onmolecular and genetic alterations to decipher the underlying

Sarcoma 11

tumorigenesis mechanisms There is a link between geneticand epigenetic alterations responsible for a variety of tumorcell growth proliferation differentiation apoptosis andtherapy-resistance mechanisms The identification of theprognostic value of the PAX-FKHR fusion status in RMSis one of the most important shifts in the lineage and therisk assessment of RMS which exerts an oncogenic effectthrough multiple pathways and is incorporated into mostrelevant studies The misbalanced expression of a number ofmiRs involved in the regulation of myogenic differentiationalso implicates the tumorsrsquo escape suppressive mechanismsMoreover we described several in vivo models of RMSto explore underlying histogenesis and different aspects oftumorigenesis Recent work on CSCs has also contributedto understanding the cell origins refining molecular char-acterization and elucidating the underlying basis of refrac-tory responses to current adjuvant treatments of subsets oftumorsThese integrative approaches can provide opportuni-ties to identify diagnostic and prognostic biomarkers appliedto the individual targeted therapy and significantly improveRMS prognosis

Abbreviations

RMS RhabdomyosarcomaARMS Alveolar rhabdomyosarcomaERMS Embryonal rhabdomyosarcomaPRMS Pleomorphic rhabdomyosarcomaSRMS Sclerosingspindle cell

rhabdomyosarcomamiRNA miR MicroRNACSC Cancer stem cellMSC Mesenchymal stem cell

Conflict of Interests

The authors declare that there is no conflict of interestsregarding the publication of this paper

Acknowledgments

This work was supported in part by grants from the Gat-tegno and Wechsler funds Dr Duan was supported in partthrough a grant from Sarcoma Foundation of America (SFA)a grant from National Cancer Institute (NCI)National Insti-tutes of Health (NIH) UO1 CA 151452 and a grant from anAcademic Enrichment Fund of MGH Orthopedic Surgery

References

[1] L Yang T Takimoto and J Fujimoto ldquoPrognostic model forpredicting overall survival in children and adolescents withrhabdomyosarcomardquo BMC Cancer vol 14 no 1 article 6542014

[2] D Egas-Bejar and W W Huh ldquoRhabdomyosarcoma in adoles-cent and young adult patients current perspectivesrdquo AdolescentHealth Medicine andTherapeutics vol 5 pp 115ndash125 2014

[3] J C Breneman E Lyden A S Pappo et al ldquoPrognosticfactors and clinical outcomes in children and adolescents withmetastatic rhabdomyosarcomamdasha report from the intergrouprhabdomyosarcoma study IVrdquo Journal of Clinical Oncology vol21 no 1 pp 78ndash84 2003

[4] V D Soleimani and M A Rudnicki ldquoNew insights into theorigin and the genetic basis of rhabdomyosarcomasrdquo CancerCell vol 19 no 2 pp 157ndash159 2011

[5] T Glass D D Cochrane S R Rassekh K Goddard and JHukin ldquoGrowing teratoma syndrome in intracranial non-ger-minomatous germ cell tumors (iNGGCTs) a risk for secondarymalignant transformationmdasha report of two casesrdquo Childrsquos Ner-vous System vol 30 no 5 pp 953ndash957 2014

[6] J Ji C Eng and K Hemminki ldquoFamilial risk for soft tissuetumors a nation-wide epidemiological study from SwedenrdquoJournal of Cancer Research and Clinical Oncology vol 134 no5 pp 617ndash624 2008

[7] C D M Fletcher J A Bridge P Hogendoorn and F MertensWHO Classification of Tumours of Soft Tissue and Bone vol 5IARC Press Paris France 4th edition 2013

[8] I Sultan I Qaddoumi S Yaser C Rodriguez-Galindo and AFerrari ldquoComparing adult and pediatric rhabdomyosarcoma inthe surveillance epidemiology and end results program 1973 to2005 an analysis of 2600 patientsrdquo Journal of Clinical Oncologyvol 27 no 20 pp 3391ndash3397 2009

[9] B Raney W Huh D Hawkins et al ldquoOutcome of patientswith localized orbital sarcoma who relapsed following treat-ment on Intergroup Rhabdomyosarcoma Study Group (IRSG)Protocols-III and -IV 1984ndash1997 a report from the ChildrenrsquosOncology Grouprdquo Pediatric Blood and Cancer vol 60 no 3 pp371ndash376 2013

[10] W Crist E A Gehan A H Ragab et al ldquoThe third intergrouprhabdomyosarcoma studyrdquo Journal of Clinical Oncology vol 13no 3 pp 610ndash630 1995

[11] C Liu D Li J Jiang et al ldquoAnalysis of molecular cytogeneticalteration in rhabdomyosarcoma by array comparative genomichybridizationrdquo PLoS ONE vol 9 no 4 Article ID e94924 2014

[12] DWilliamson E Missiaglia A de Reynies et al ldquoFusion gene-negative alveolar rhabdomyosarcoma is clinically and molec-ularly indistinguishable from embryonal rhabdomyosarcomardquoJournal of Clinical Oncology vol 28 no 13 pp 2151ndash2158 2010

[13] V Paulson G Chandler D Rakheja et al ldquoHigh-resolutionarray CGH identifies common mechanisms that drive embry-onal rhabdomyosarcoma pathogenesisrdquo Genes Chromosomesand Cancer vol 50 no 6 pp 397ndash408 2011

[14] S Park J Lee I-G Do et al ldquoAberrant CDK4 amplificationin refractory rhabdomyosarcoma as identified by genomicprofilingrdquo Scientific Reports vol 4 article 3623 2014

[15] C Liu D Li J Hu et al ldquoChromosomal and genetic imbalancesin Chinese patients with rhabdomyosarcoma detected by high-resolution array comparative genomic hybridizationrdquo Interna-tional Journal of Clinical and Experimental Pathology vol 7 no2 pp 690ndash698 2014

[16] Q-X Li C-X Liu C-P Chun et al ldquoChromosomal imbalancesrevealed in primary rhabdomyo-sarcomas by comparativegenomic hybridizationrdquo Chinese Medical Journal vol 122 no11 pp 1277ndash1282 2009

[17] E Missiaglia J Selfe M Hamdi et al ldquoGenomic imbalancesin rhabdomyosarcoma cell lines affect expression of genesfrequently altered in primary tumors an approach to identifycandidate genes involved in tumor developmentrdquo Genes Chro-mosomes and Cancer vol 48 no 6 pp 455ndash467 2009

12 Sarcoma

[18] F G Barr F Duan L M Smith et al ldquoGenomic and clinicalanalyses of 2p24 and 12q13-q14 amplification in alveolar rhab-domyosarcoma a report from the childrenrsquos oncology grouprdquoGenes Chromosomes and Cancer vol 48 no 8 pp 661ndash6722009

[19] T Gordon AMcManus J Anderson et al ldquoCytogenetic abnor-malities in 42 rhabdomyosarcoma a United Kingdom CancerCytogenetics Group StudyrdquoMedical and Pediatric Oncology vol36 no 2 pp 259ndash267 2001

[20] X Chen E Stewart A A Shelat et al ldquoTargeting oxidativestress in embryonal rhabdomyosarcomardquo Cancer Cell vol 24no 6 pp 710ndash724 2013

[21] S Lizard-Nacol F Mugneret C Volk C Turc-Carel M Favrotand T Philip ldquoTranslocation (213)(q37q14) in alveolar rhab-domyosarcoma a new caserdquo Cancer Genetics and Cytogeneticsvol 25 no 2 pp 373ndash374 1987

[22] P H B Sorensen J C Lynch S J Qualman et al ldquoPAX3-FKHR and PAX7-FKHR gene fusions are prognostic indicatorsin alveolar rhabdomyosarcoma a report from the childrenrsquosoncology grouprdquo Journal of Clinical Oncology vol 20 no 11 pp2672ndash2679 2002

[23] A D Marshall and G C Grosveld ldquoAlveolar rhabdomyo-sarcomamdashthe molecular drivers of PAX37-FOXO1-inducedtumorigenesisrdquo Skeletal Muscle vol 2 no 1 article 25 2012

[24] E Missiaglia D Williamson J Chisholm et al ldquoPAX3FOXO1fusion gene status is the key prognostic molecular markerin rhabdomyosarcoma and significantly improves current riskstratificationrdquo Journal of Clinical Oncology vol 30 no 14 pp1670ndash1677 2012

[25] J F Shern L Chen J Chmielecki et al ldquoComprehensivegenomic analysis of rhabdomyosarcoma reveals a landscape ofalterations affecting a common genetic axis in fusion-positiveand fusion-negative tumorsrdquo Cancer Discovery vol 4 no 2 pp216ndash231 2014

[26] MWachtel M Dettling E Koscielniak et al ldquoGene expressionsignatures identify rhabdomyosarcoma subtypes and detect anovel t(22)(q35p23) translocation fusing PAX3 to NCOA1rdquoCancer Research vol 64 no 16 pp 5539ndash5545 2004

[27] E Davicioni M J Anderson F G Finckenstein et al ldquoMolec-ular classification of rhabdomyosarcomamdashgenotypic and phe-notypic determinants of diagnosis a report from the ChildrenrsquosOncology Grouprdquo The American Journal of Pathology vol 174no 2 pp 550ndash564 2009

[28] M Visser C Sijmons J Bras et al ldquoAllelotype of pediatric rhab-domyosarcomardquo Oncogene vol 15 no 11 pp 1309ndash1314 1997

[29] R Gil-Benso T San-Miguel R C Callaghan et al ldquoChromo-somal and genetic changes produced in tumoral progression ofembryonal rhabdomyosarcomardquo Histopathology vol 62 no 5pp 816ndash819 2013

[30] S X Skapek J Anderson F G Barr et al ldquoPAX-FOXO1 fusionstatus drives unfavorable outcome for children with rhabdom-yosarcoma a childrenrsquos oncology group reportrdquo Pediatric Bloodand Cancer vol 60 no 9 pp 1411ndash1417 2013

[31] HHosoi N Kakazu E Konishi et al ldquoA novel PAX3 rearrange-ment in embryonal rhabdomyosarcomardquo Cancer Genetics andCytogenetics vol 189 no 2 pp 98ndash104 2009

[32] E Takaoka H Sonobe K Akimaru et al ldquoMultiple sites ofhighly amplified DNA sequences detected bymolecular cytoge-netic analysis inHS-RMS-2 a newpleomorphic rhabdomyosar-coma cell linerdquo American Journal of Cancer Research vol 2 no2 pp 141ndash152 2012

[33] K Szuhai D de Jong W Y Leung C D M Fletcher and P CWHogendoorn ldquoTransactivatingmutation of theMYOD1 geneis a frequent event in adult spindle cell rhabdomyosarcomardquoJournal of Pathology vol 232 no 3 pp 300ndash307 2014

[34] N P Agaram C-L Chen L Zhang M P Laquaglia L Wexlerand C R Antonescu ldquoRecurrent MYOD1 mutations in pedi-atric and adult sclerosing and spindle cell rhabdomyosarcomasEvidence for a common pathogenesisrdquoGenes Chromosomes andCancer vol 53 no 9 pp 779ndash787 2014

[35] H Yoshida M Miyachi K Sakamoto et al ldquoPAX3-NCOA2fusion gene has a dual role in promoting the proliferation andinhibiting the myogenic differentiation of rhabdomyosarcomacellsrdquo Oncogene vol 33 pp 5601ndash5608 2013

[36] J M Mosquera A Sboner L Zhang et al ldquoRecurrent NCOA2gene rearrangements in congenitalinfantile spindle cell rhab-domyosarcomardquo Genes Chromosomes and Cancer vol 52 no6 pp 538ndash550 2013

[37] V Thavaraj A Sethi and L S Arya ldquoIncomplete Beckwith-Wiedemann syndrome in a child with orbital rhabdomyosar-comardquo Indian Pediatrics vol 39 no 3 pp 299ndash304 2002

[38] D Walter S Satheesha P Albrecht et al ldquoCD133 positiveembryonal rhabdomyosarcoma stem-like cell population isenriched in rhabdospheresrdquo PLoS ONE vol 6 no 5 Article IDe19506 2011

[39] Y-X Ren F G Finckenstein D A Abdueva et al ldquoMousemesenchymal stem cells expressing PAX-FKHR form alveolarrhabdomyosarcomas by cooperating with secondary muta-tionsrdquo Cancer Research vol 68 no 16 pp 6587ndash6597 2008

[40] M Salerno S Avnet G Bonuccelli S Hosogi D Granchi andN Baldini ldquoImpairment of lysosomal activity as a therapeuticmodality targeting cancer stem cells of embryonal rhabdom-yosarcoma cell line RDrdquo PLoS ONE vol 9 no 10 Article IDe110340 2014

[41] K Tanoue Y Wang M Ikeda et al ldquoSurvivin-responsive con-ditionally replicating adenovirus kills rhabdomyosarcoma stemcells more efficiently than their progenyrdquo Journal of Transla-tional Medicine vol 12 no 1 article 27 2014

[42] P K Rao EMissiaglia L Shields et al ldquoDistinct roles formiR-1and miR-133a in the proliferation and differentiation of rhab-domyosarcoma cellsrdquo The FASEB Journal vol 24 no 9 pp3427ndash3437 2010

[43] D Yan X D Dong X Chen et al ldquoMicroRNA-1206 targets c-met and inhibits rhabdomyosarcomadevelopmentrdquoThe Journalof Biological Chemistry vol 284 no 43 pp 29596ndash29604 2009

[44] I Marchesi A Giordano and L Bagella ldquoRoles of enhancerof zeste homolog 2 from skeletal muscle differentiation torhabdomyosarcoma carcinogenesisrdquoCell Cycle vol 13 no 4 pp516ndash527 2014

[45] M M Sun J F Li L L Guo et al ldquoTGF-1205731 suppression ofmicroRNA-450b-5p expression a novel mechanism for block-ingmyogenic differentiation of rhabdomyosarcomardquoOncogenevol 33 no 16 pp 2075ndash2086 2014

[46] Y Diao X Guo L Jiang et al ldquoMiR-203 a tumor suppressorfrequently down-regulated by promoter hypermethylation inrhabdomyosarcomardquo The Journal of Biological Chemistry vol289 no 1 pp 529ndash539 2014

[47] H-J Huang J Liu HHua et al ldquoMiR-214 andN-ras regulatoryloop suppresses rhabdomyosarcoma cell growth and xenografttumorigenesisrdquo Oncotarget vol 5 no 8 pp 2161ndash2175 2014

Sarcoma 13

[48] A L Sarver L Li and S Subramanian ldquomicroRNA miR-183functions as an oncogene by targeting the transcription factorEGR1 and promoting tumor cell migrationrdquo Cancer Researchvol 70 no 23 pp 9570ndash9580 2010

[49] R L Galindo J A Allport and E N Olson ldquoA Drosophilamodel of the rhabdomyosarcoma initiator PAX7-FKHRrdquo Pro-ceedings of the National Academy of Sciences of the United Statesof America vol 103 no 36 pp 13439ndash13444 2006

[50] U Avirneni-Vadlamudi K A Galindo T R Endicott V Paul-son S Cameron and R L Galindo ldquoDrosophila and mammal-ian models uncover a role for the myoblast fusion gene TANC1in rhabdomyosarcomardquo The Journal of Clinical Investigationvol 122 no 1 pp 403ndash407 2012

[51] E Y Chen andDM Langenau ldquoZebrafishmodels of rhabdom-yosarcomardquoMethods in Cell Biology vol 105 pp 383ndash402 2011

[52] X Le E K Pugach S Hettmer et al ldquoA novel chemicalscreening strategy in zebrafish identifies common pathways inembryogenesis and rhabdomyosarcoma developmentrdquo Devel-opment vol 140 no 11 pp 2354ndash2364 2012

[53] Y Lee R Kawagoe K Sasai et al ldquoLoss of suppressor-of-fusedfunction promotes tumorigenesisrdquoOncogene vol 26 no 44 pp6442ndash6447 2007

[54] C Keller B R Arenkiel C M Coffin N El-Bardeesy R ADePinho and M R Capecchi ldquoAlveolar rhabdomyosarcomasin conditional Pax3Fkhr mice cooperativity of Ink4aARF andTrp53 loss of functionrdquo Genes and Development vol 18 no 21pp 2614ndash2626 2004

[55] S Singh CVinson CMGurley et al ldquoImpairedWnt signalingin embryonal rhabdomyosarcoma cells from p53c-fos doublemutant micerdquo American Journal of Pathology vol 177 no 4 pp2055ndash2066 2010

[56] G J Schaaf J M Ruijter F Van Ruissen et al ldquoFull transcrip-tome analysis of rhabdomyosarcoma normal and fetal skeletalmuscle statistical comparison of multiple SAGE librariesrdquo TheFASEB Journal vol 19 no 3 pp 404ndash406 2005

[57] D S Hawkins A A Gupta and E R Rudzinski ldquoWhat is newin the biology and treatment of pediatric rhabdomyosarcomardquoCurrent Opinion in Pediatrics vol 26 no 1 pp 50ndash56 2014

[58] M Zhang J Truscott and J Davie ldquoLoss of MEF2D expressioninhibits differentiation and contributes to oncogenesis in rhab-domyosarcoma cellsrdquoMolecular Cancer vol 12 no 1 article 1502013

[59] M Ciesla J Dulak and A Jozkowicz ldquomicroRNAs and epi-genetic mechanisms of rhabdomyosarcoma developmentrdquo TheInternational Journal of Biochemistry amp Cell Biology vol 53 pp482ndash492 2014

[60] S Ricaud B Vernus M Duclos et al ldquoInhibition of autocrinesecretion of myostatin enhances terminal differentiation inhuman rhabdomyosarcoma cellsrdquo Oncogene vol 22 no 51 pp8221ndash8232 2003

[61] C S Guo C Degnin T A Fiddler D Stauffer andM JThayerldquoRegulation of MyoD activity and muscle cell differentiation byMDM2 pRb and Sp1rdquoThe Journal of Biological Chemistry vol278 no 25 pp 22615ndash22622 2003

[62] S Rossi E Stoppani P L Puri and A Fanzani ldquoDifferenti-ation of human rhabdomyosarcoma RD cells is regulated byreciprocal functional interactions between myostatin p38 andextracellular regulated kinase signalling pathwaysrdquo EuropeanJournal of Cancer vol 47 no 7 pp 1095ndash1105 2011

[63] Z S Walters B Villarejo-Balcells D Olmos et al ldquoJARID2is a direct target of the PAX3-FOXO1 fusion protein andinhibits myogenic differentiation of rhabdomyosarcoma cellsrdquoOncogene vol 33 no 9 pp 1148ndash1157 2014

[64] T Hosoyama M I Aslam J Abraham et al ldquoIL-4R drives ded-ifferentiation mitogenesis and metastasis in rhabdomyosar-comardquo Clinical Cancer Research vol 17 no 9 pp 2757ndash27662011

[65] F S Dela Cruz ldquoCancer stem cells in pediatric sarcomasrdquoFrontiers in Oncology vol 3 article 168 2013

[66] M J Bissell and W C Hines ldquoWhy donrsquot we get more cancerA proposed role of the microenvironment in restraining cancerprogressionrdquo Nature Medicine vol 17 no 3 pp 320ndash329 2011

[67] E Charytonowicz C Cordon-Cardo I Matushansky and MZiman ldquoAlveolar rhabdomyosarcoma is the cell of origin amesenchymal stem cellrdquoCancer Letters vol 279 no 2 pp 126ndash136 2009

[68] W K Cavenee ldquoMuscling in on rhabdomyosarcomardquo NatureMedicine vol 8 no 11 pp 1200ndash1201 2002

[69] B Zhu and J K Davie ldquoNew insights into signalling-pathwayalterations in rhabdomyosarcomardquo British Journal of Cancervol 112 no 2 pp 227ndash231 2015

[70] M Zhang C M Linardic and D G Kirsch ldquoRAS and ROS inrhabdomyosarcomardquo Cancer Cell vol 24 no 6 pp 689ndash6912013

[71] S C Marianna L Ianzano and G Nicoletti ldquoTumor sup-pressor genes promote rhabdomyosarcoma progression in p53heterozygousrdquo Oncotarget vol 5 no 1 pp 108ndash119 2013

[72] SWang L Guo L Dong et al ldquoTGF-beta1 signal pathwaymaycontribute to rhabdomyosarcoma development by inhibitingdifferentiationrdquo Cancer Science vol 101 no 5 pp 1108ndash11162010

[73] J Wesche K Haglund and E M Haugsten ldquoFibroblast growthfactors and their receptors in cancerrdquo Biochemical Journal vol437 no 2 pp 199ndash213 2011

[74] M Wachtel J Rakic M Okoniewski P Bode F Niggli andB W Schafer ldquoFGFR4 signaling couples to Bim and not Bmfto discriminate subsets of alveolar rhabdomyosarcoma cellsrdquoInternational Journal of Cancer vol 135 no 7 pp 1543ndash15522014

[75] P L Puri ZWu P Zhang et al ldquoInduction of terminal differen-tiation by constitutive activation of p38 MAP kinase in humanrhabdomyosarcoma cellsrdquo Genes and Development vol 14 no5 pp 574ndash584 2000

[76] A M Tremblay E Missiaglia G G Galli et al ldquoThe Hippotransducer YAP1 transforms activated satellite cells and is apotent effector of embryonal rhabdomyosarcoma formationrdquoCancer Cell vol 26 no 2 pp 273ndash287 2014

[77] R N Judson A M Tremblay P Knopp et al ldquoThe hippo path-way member Yap plays a key role in influencing fate decisionsin muscle satellite cellsrdquo Journal of Cell Science vol 125 no 24pp 6009ndash6019 2012

[78] L E S Crose K A Galindo J G Kephart et al ldquoAlve-olar rhabdomyosarcoma-associated PAX3-FOXO1 promotestumorigenesis via Hippo pathway suppressionrdquo Journal ofClinical Investigation vol 124 no 1 pp 285ndash296 2014

[79] B Belyea J G Kephart J Blum D G Kirsch and C MLinardic ldquoEmbryonic signaling pathways and rhabdomyosar-coma contributions to cancer development and opportunitiesfor therapeutic targetingrdquo Sarcoma vol 2012 Article ID 40623913 pages 2012

14 Sarcoma

[80] J Huertas-Martinez S Rello-Varona D Herrero-Martin et alldquoCaveolin-1 is down-regulated in alveolar rhabdomyosarcomasand negatively regulates tumor growthrdquo Oncotarget vol 5 no20 pp 9744ndash9755 2014

[81] S Rossi P L Poliani CMissale EMonti andA Fanzani ldquoCav-eolins in rhabdomyosarcomardquo Journal of Cellular andMolecularMedicine vol 15 no 12 pp 2553ndash2568 2011

[82] P G Tsioli E S Patsouris C Giaginis and S E TheocharisldquoDNA repair systems in rhabdomyosarcomardquo Histology andHistopathology vol 28 no 8 pp 971ndash984 2013

[83] E Gatzidou C Michailidi S Tseleni-Balafouta and S Theo-charis ldquoAn epitome of DNA repair related genes and mecha-nisms in thyroid carcinomardquo Cancer Letters vol 290 no 2 pp139ndash147 2010

[84] R Rota R Ciarapica A Giordano L Miele and F LocatellildquoMicroRNAs in rhabdomyosarcoma pathogenetic implicationsand translational potentialityrdquoMolecular Cancer vol 10 article120 2011

[85] A Gougelet J Perez D Pissaloux et al ldquoMiRNA profiling howto bypass the current difficulties in the diagnosis and treatmentof sarcomasrdquo Sarcoma vol 2011 Article ID 460650 13 pages2011

[86] R Ciarapica G Russo F Verginelli et al ldquoDeregulated expres-sion of miR-26a and Ezh2 in rhabdomyosarcomardquo Cell Cyclevol 8 no 1 pp 172ndash175 2009

[87] L Li A L Sarver S Alamgir and S Subramanian ldquoDownreg-ulation of microRNAs miR-1 -206 and -29 stabilizes PAX3 andCCND2 expression in rhabdomyosarcomardquo Laboratory Investi-gation vol 92 no 4 pp 571ndash583 2012

[88] S-Y Ng G K Bogu B Soh and L W Stanton ldquoThe longnoncoding RNA RMST interacts with SOX2 to regulate neu-rogenesisrdquoMolecular Cell vol 51 no 3 pp 349ndash359 2013

[89] C A Lynch B Tycko T H Bestor and C P Walsh ldquoReac-tivation of a silenced H19 gene in human rhabdomyosarcomaby demethylation of DNA but not by histone hyperacetylationrdquoMolecular Cancer vol 1 article 2 2002

[90] P Onyango and A P Feinberg ldquoA nucleolar protein H19opposite tumor suppressor (HOTS) is a tumor growth inhibitorencoded by a human imprinted H19 antisense transcriptrdquoProceedings of the National Academy of Sciences of the UnitedStates of America vol 108 no 40 pp 16759ndash16764 2011

[91] C E Albacker N Y Storer E M Langdon et al ldquoThe his-tone methyltransferase SUV39H1 suppresses embryonal rhab-domyosarcoma formation in zebrafishrdquo PLoS ONE vol 8 no 5Article ID e64969 2013

[92] N Y Storer R M White A Uong et al ldquoZebrafish rhab-domyosarcoma reflects the developmental stage of oncogeneexpression during myogenesisrdquo Development vol 140 no 14pp 3040ndash3050 2013

[93] M E Hatley W Tang M R Garcia et al ldquoA mouse modelof rhabdomyosarcoma originating from the adipocyte lineagerdquoCancer Cell vol 22 no 4 pp 536ndash546 2012

[94] V Hosur A Kavirayani J Riefler et al ldquoDystrophin and dysfer-lin doublemutantmice a novelmodel for rhabdomyosarcomardquoCancer Genetics vol 205 no 5 pp 232ndash241 2012

[95] W Goessling T E North and L I Zon ldquoNew waves of discov-ery modeling cancer in zebrafishrdquo Journal of Clinical Oncologyvol 25 no 17 pp 2473ndash2479 2007

[96] B P Rubin K Nishijo H-I H Chen et al ldquoEvidence for anunanticipated relationship between undifferentiated pleomor-phic sarcoma and embryonal rhabdomyosarcomardquo Cancer Cellvol 19 no 2 pp 177ndash191 2011

[97] S Z Kaylani J Xu R K Srivastava L Kopelovich J G Presseyand M Athar ldquoRapamycin targeting mTOR and hedgehog sig-naling pathways blocks human rhabdomyosarcoma growth inxenograft murine modelrdquo Biochemical and Biophysical ResearchCommunications vol 435 no 4 pp 557ndash561 2013

[98] S Q Li A T Cheuk J F Shern et al ldquoTargeting wild-type andmutationally activated FGFR4 in rhabdomyosarcoma with theinhibitor ponatinib (AP24534)rdquoPLoSONE vol 8 no 10 ArticleID e76551 2013

[99] K Miyoshi K Kohashi F Fushimi et al ldquoClose correlationbetween CXCR4 and VEGF expression and frequent CXCR7expression in rhabdomyosarcomardquo Human Pathology vol 45no 9 pp 1900ndash1909 2014

[100] ADMarshall F Picchione R I KGeltink andGCGrosveldldquoPAX3-FOXO1 induces up-regulation of Noxa sensitizing alve-olar rhabdomyosarcoma cells to apoptosisrdquo Neoplasia vol 15no 7 pp 738ndash748 2013

[101] F Perut S Avnet C Fotia et al ldquoV-ATPase as an effective thera-peutic target for sarcomasrdquo Experimental Cell Research vol 320no 1 pp 21ndash32 2014

Submit your manuscripts athttpwwwhindawicom

Stem CellsInternational

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

MEDIATORSINFLAMMATION

of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Behavioural Neurology

EndocrinologyInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Disease Markers

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

BioMed Research International

OncologyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Oxidative Medicine and Cellular Longevity

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

PPAR Research

The Scientific World JournalHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Immunology ResearchHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal of

ObesityJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Computational and Mathematical Methods in Medicine

OphthalmologyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Diabetes ResearchJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Research and TreatmentAIDS

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Gastroenterology Research and Practice

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Parkinsonrsquos Disease

Evidence-Based Complementary and Alternative Medicine

Volume 2014Hindawi Publishing Corporationhttpwwwhindawicom

Page 9: Review Article Rhabdomyosarcoma: Advances in Molecular and

Sarcoma 9

Table 5 In vivo animal models of RMS

Model TargetModel origin Reference

Inactivation Expression

DrosophilaPAX-FKHR Bipartite Gal4-UAS

expression systemGalindo et al2006 [49]

PAX7-FOXO1 rols Chromosomal deletionDf(3L)vin5

Avirneni-Vadlamudi et al2012 [50]

Zebrafishrag2 promoter c-Myc kRASG12D

Chen andLangenau 2011[51]

MAPKERK andAKTS6K1 PD98059 TPCK Tg(hsp70-HRASG12V) Le et al 2012

[52]

Mouse

Sufu N-myc Sfrp1 Ptch2and cyclin D1 Sufu+minus Lee et al 2007

[53]

Ink4aArf and Trp53 Pax3FKHR Pax3P3Fpwt Keller et al2004 [54]

Wnt120573-cateninsignaling pathway Wnt2 p53minusminusc-fosminusminus Singh et al 2010

[55]

a tumor growth inhibitor is encoded by an imprinted H19antisense transcript Overexpression of HOTS inhibits RMStumor cell growth Silencing HOTS by RNAi was shown toincrease in vitro colony formation as well as in vivo tumorgrowth of RMS [90]

6 In Vivo Models of RMS

Several in vivo models have recently been developed inDrosophila zebrafish and mice to further understand themolecular and cellular biology of RMS tumorigenesis [49ndash55 91ndash94] (Table 5)

61 Drosophila Models in RMS Despite the evolutionarydistance a strong conservation of genes pathways andregulatory molecular networks have been demonstratedbetween flies and humans Many human disease genes haverelated sequences in Drosophila In contrast to mammalianmodels the generation of Drosophila mutants is easy cheapand fast

For a better understanding of the pathogenic conse-quences of PAX-FKHR expression Drosophila was chosenbecause the animalrsquos transparent outer cuticle allows forsimple real-time monitoring of muscle abnormalities elicitedby PAX-FKHR including subtle or focal changes [49] Themutation in the myoblast fusion gene rolling pebbles (rols)was reported to dominantly suppress PAX-FKHR1-inducedlethality by using a Drosophila model of PAX-FKHR1-mediated transformation [50] PAX-FKHR1 signaling upreg-ulates the TANC1 gene and blocks myoblasts from terminaldifferentiation However downregulating the TANC1 genecould cause RMS cells to lose their neoplastic state undergofusion and form differentiation This novel finding uncov-ered a PAX-FKHR1-TANC1 neoplasia axis collaboratively ina Drosophila model and loss-gain-of-function studies inmammalian platforms [50]

62 Zebrafish Models in RMS In contrast to other mod-els the short tumor onset in zebrafish allows for rapididentification of essential genes in various processes suchas tumor growth self-renewal and maintenance There areother advantages for using zebrafish in the study of humandisease and development including the ease and low cost ofraising large numbers of fish and the highly conserved geneticand biochemical pathways between zebrafish and mammals[95]

As zebrafish RMS is highly similar to human ERMSby using fluorescent transgenic approaches it is more con-venient to understand histogenesis and different aspects oftumorigenesis in RMS A robust zebrafish model of RAS-induced RMS has been established sharing twomorphologicand immunophenotypic features resembling ERMS [51] Inone case both are associated with tissue-restricted geneexpression in RMS while in another both comprise a RAS-induced gene signature Cross-species microarray compar-isons confirm that conserved genetic tumor-specific andtissue-restricted pathways such as RAS and p53 pathwaysdrive RMS growth Tg(hsp70-HRASG12V) zebrafish embryoswere generated to evaluate gene expression that mimics RASpathway activation during tumorigenesis to defineRAS targetgenes [52] Another KRASG12D-induced zebrafish embryonalRMS was utilized to assess the therapeutic effects By theblockage of two major downstream signaling pathwaysMAPKERK and AKTS6K1 the model showed that inhibi-tion of translation initiation suppresses tumor cell prolifera-tion [52]

All the above zebrafish cancer models share similar cellu-lar features and molecular pathways with human RMS espe-cially ERMS and can demonstrate a response to therapiesin a similar manner with human RMS Thus a tremendousopportunity is available to implement these animal modelsinto different steps for novel targeted therapeutic develop-ment

10 Sarcoma

63 Mouse Models in RMS Mouse models established fromin vivo studies may contribute to the understanding of thegenetic basis in tumor development and progression andmayhelp to test the efficacies of novel antineoplastic agents

The importance of PAX3PAX7-FKHR fusion proteins inthe progression of ARMS tumorigenesis is evident from bothin vitro and in vivo studies Transgenic mouse models gener-ated with the PAX-FKHR fusion genemimic the formation ofhuman ARMS [67] A mouse model was developed with het-erozygous loss of Sufu a tumor suppressor inHedgehog (Hh)signaling combined with p53 loss driven ERMS with a lowpenetrance (9) [53] These studies indicate that the modelsconsistently formingARMSnot only need the introduction ofthe PAX-FKHR fusion gene but also need to be accompaniedby other genetic events such as p53 pathway disruption [54]

Compared with ARMS ERMS models are more complexto generate because of their lower tumor penetrance andlonger latency JW41 cells from the p53c-fos double mutantmice models resemble human ERMS cells morphologicallyand express similar characteristic markers [55] The overex-pression of the Wnt2 gene identified in JW41 cells confirmedin human RMS cell lines can allow for further analysis of theWnt signaling pathway Both of these results indicate that thedownregulation of theWnt pathway contributes to resistanceto apoptosis and the inhibition of myogenic differentiation inERMS

Using Ptch1 p53 andor Rb1 conditional mouse modelsand controlling prenatal or postnatal myogenic cell of originRubin et al demonstrated that the loss of p53 in maturingmyoblasts related to tumorigenesis of ERMS [96] They alsoindicated that Rb1 alteration with other oncogenic factorswas strongly associated with an undifferentiated phenotypeacting as a modifier In addition they highlighted a sub-set of p53-deficient ERMSs arising from Myf6-expressingmyoblasts which had the same latency with Pax7-expressingmurine satellite cells in the study

7 Potential Therapeutic Targets in RMS

Detection of genomic imbalances and identification of thecrucial effective genes can contribute to identifying novelpotential biomarkers and relevant targets for clinical therapyin RMS

Survivin-responsive conditionally replicating adenovi-ruses can regulate multiple factors (Survm-CRAs) [41]Although Survm-CRA can efficiently replicate and potentlyinduce cell death in RMS cells the cytotoxic effects are morepronounced in RSC-enriched or RSC-purified cells thanin RSC-exiguous or progeny-purified cells Injections ofSurvm-CRAs into tumor nodules generated by transplantingRSC-enriched cells induce significant death in RMS cells andregression of tumor nodulesThe unique therapeutic featuresof Survm-CRA include not only its therapeutic effectivenessagainst RMS but also its increasing effectiveness against CSCsuggesting that Survm-CRA may be a promising anticanceragent [41]

Rapamycin an inhibitor of mTOR can abrogate RMStumor growth in a xenograft mouse model [97] Thetumors in the rapamycin-treatedmice group show significant

reduction of proliferation and invasiveness and inductionof apoptosis The tumor growth inhibition is simultaneousassociated with the diminution of mTOR and Hh pathwayswhich are implicated in the pathogenesis of RMSThe resultsindicate that using rapamycin either alone or in combinationwith traditional chemotherapeutic drugs may represent apotential targeted agent for therapeutic intervention Ampli-fication and mutational activation of Fibroblast Growth Fac-tor Receptor 4 (FGFR4) in RMS cells promote tumor pro-gression [98] Ponatinib is the most potent FGFR4 inhibitorto block wild-type RMS cell growth mutate FGFR4 throughincreasing apoptosis and suppress the FGFR4 downstreamtarget STAT3 Ponatinib treatment slows tumor growth inRMS mouse models expressing mutated FGFR4 [98] Pona-tinib as an FGFR4 inhibitor shows potential to act as aneffective therapeutic agent for RMS tumorsThe expression ofCXC chemokine receptor 4 (CXCR4) CXCR7 and VascularEndothelial Growth Factor (VEGF) indicates poor prognosisin a variety of malignancies including RMS cell lines Mostsamples in a large series of clinical RMS cases show asso-ciation with high expression of these antagonists regardlessof their subtypes [99] However there are significant corre-lations with high expression of CXCR4 and VEGF in bothERMS and ARMS High VEGF expression is predictive ofadverse prognostic factors in RMS as the expression of theseangiogenesis factors was compared with clinicopathologicalparameters and prognosis Considering their overexpressionin RMS these chemokine receptors and VEGF could providepotential molecular therapeutic targets in RMS Some ARMScell lines have undergone apoptosis in response to antineo-plastic drugs such as bortezomib The proapoptotic BH3only family member NOXA acts as an upregulated proteindownstream PAX3-FKHR resulting in increased cell sensi-tivity to bortezomib Apoptosis in response to bortezomibcan be reversed by shRNA knockdown of Noxa PAX3-FKHR upregulation of Noxa creates a potential therapeuticinsight into inducing apoptosis in ARMS cellsThis apoptosispathway could represent a specific targeted therapy againstPAX3-FKHR-expressing ARMS cells [100]

Proton pump activation resulting from intracellular acid-ificationmay be an energetic mechanism to escape apoptosisV-ATPase is an ATP-driven proton pump acidifying theintracellular compartment and transports protons across theplasma membrane Esomeprazole as a V-ATPase inhibitor inthe PPI administration interferes with intensive ion trans-porter activity of RMS cells and induces remarkable cytotoxi-city RMS expresses a higher level ofV-ATPase comparedwithother sarcomasTherefore RMS should be very susceptible toesomeprazole treatment and V-ATPase could be consideredas a promising selective target for treatment [101]

8 Conclusion and Future Prospects

For RMS the current challenges include how to iden-tify promising novel therapeutics and integrate them intoexisting therapy For better understanding of congenitaland epigenetic modifications in the development of RMSthis review summarizes the recent genome-wide studies onmolecular and genetic alterations to decipher the underlying

Sarcoma 11

tumorigenesis mechanisms There is a link between geneticand epigenetic alterations responsible for a variety of tumorcell growth proliferation differentiation apoptosis andtherapy-resistance mechanisms The identification of theprognostic value of the PAX-FKHR fusion status in RMSis one of the most important shifts in the lineage and therisk assessment of RMS which exerts an oncogenic effectthrough multiple pathways and is incorporated into mostrelevant studies The misbalanced expression of a number ofmiRs involved in the regulation of myogenic differentiationalso implicates the tumorsrsquo escape suppressive mechanismsMoreover we described several in vivo models of RMSto explore underlying histogenesis and different aspects oftumorigenesis Recent work on CSCs has also contributedto understanding the cell origins refining molecular char-acterization and elucidating the underlying basis of refrac-tory responses to current adjuvant treatments of subsets oftumorsThese integrative approaches can provide opportuni-ties to identify diagnostic and prognostic biomarkers appliedto the individual targeted therapy and significantly improveRMS prognosis

Abbreviations

RMS RhabdomyosarcomaARMS Alveolar rhabdomyosarcomaERMS Embryonal rhabdomyosarcomaPRMS Pleomorphic rhabdomyosarcomaSRMS Sclerosingspindle cell

rhabdomyosarcomamiRNA miR MicroRNACSC Cancer stem cellMSC Mesenchymal stem cell

Conflict of Interests

The authors declare that there is no conflict of interestsregarding the publication of this paper

Acknowledgments

This work was supported in part by grants from the Gat-tegno and Wechsler funds Dr Duan was supported in partthrough a grant from Sarcoma Foundation of America (SFA)a grant from National Cancer Institute (NCI)National Insti-tutes of Health (NIH) UO1 CA 151452 and a grant from anAcademic Enrichment Fund of MGH Orthopedic Surgery

References

[1] L Yang T Takimoto and J Fujimoto ldquoPrognostic model forpredicting overall survival in children and adolescents withrhabdomyosarcomardquo BMC Cancer vol 14 no 1 article 6542014

[2] D Egas-Bejar and W W Huh ldquoRhabdomyosarcoma in adoles-cent and young adult patients current perspectivesrdquo AdolescentHealth Medicine andTherapeutics vol 5 pp 115ndash125 2014

[3] J C Breneman E Lyden A S Pappo et al ldquoPrognosticfactors and clinical outcomes in children and adolescents withmetastatic rhabdomyosarcomamdasha report from the intergrouprhabdomyosarcoma study IVrdquo Journal of Clinical Oncology vol21 no 1 pp 78ndash84 2003

[4] V D Soleimani and M A Rudnicki ldquoNew insights into theorigin and the genetic basis of rhabdomyosarcomasrdquo CancerCell vol 19 no 2 pp 157ndash159 2011

[5] T Glass D D Cochrane S R Rassekh K Goddard and JHukin ldquoGrowing teratoma syndrome in intracranial non-ger-minomatous germ cell tumors (iNGGCTs) a risk for secondarymalignant transformationmdasha report of two casesrdquo Childrsquos Ner-vous System vol 30 no 5 pp 953ndash957 2014

[6] J Ji C Eng and K Hemminki ldquoFamilial risk for soft tissuetumors a nation-wide epidemiological study from SwedenrdquoJournal of Cancer Research and Clinical Oncology vol 134 no5 pp 617ndash624 2008

[7] C D M Fletcher J A Bridge P Hogendoorn and F MertensWHO Classification of Tumours of Soft Tissue and Bone vol 5IARC Press Paris France 4th edition 2013

[8] I Sultan I Qaddoumi S Yaser C Rodriguez-Galindo and AFerrari ldquoComparing adult and pediatric rhabdomyosarcoma inthe surveillance epidemiology and end results program 1973 to2005 an analysis of 2600 patientsrdquo Journal of Clinical Oncologyvol 27 no 20 pp 3391ndash3397 2009

[9] B Raney W Huh D Hawkins et al ldquoOutcome of patientswith localized orbital sarcoma who relapsed following treat-ment on Intergroup Rhabdomyosarcoma Study Group (IRSG)Protocols-III and -IV 1984ndash1997 a report from the ChildrenrsquosOncology Grouprdquo Pediatric Blood and Cancer vol 60 no 3 pp371ndash376 2013

[10] W Crist E A Gehan A H Ragab et al ldquoThe third intergrouprhabdomyosarcoma studyrdquo Journal of Clinical Oncology vol 13no 3 pp 610ndash630 1995

[11] C Liu D Li J Jiang et al ldquoAnalysis of molecular cytogeneticalteration in rhabdomyosarcoma by array comparative genomichybridizationrdquo PLoS ONE vol 9 no 4 Article ID e94924 2014

[12] DWilliamson E Missiaglia A de Reynies et al ldquoFusion gene-negative alveolar rhabdomyosarcoma is clinically and molec-ularly indistinguishable from embryonal rhabdomyosarcomardquoJournal of Clinical Oncology vol 28 no 13 pp 2151ndash2158 2010

[13] V Paulson G Chandler D Rakheja et al ldquoHigh-resolutionarray CGH identifies common mechanisms that drive embry-onal rhabdomyosarcoma pathogenesisrdquo Genes Chromosomesand Cancer vol 50 no 6 pp 397ndash408 2011

[14] S Park J Lee I-G Do et al ldquoAberrant CDK4 amplificationin refractory rhabdomyosarcoma as identified by genomicprofilingrdquo Scientific Reports vol 4 article 3623 2014

[15] C Liu D Li J Hu et al ldquoChromosomal and genetic imbalancesin Chinese patients with rhabdomyosarcoma detected by high-resolution array comparative genomic hybridizationrdquo Interna-tional Journal of Clinical and Experimental Pathology vol 7 no2 pp 690ndash698 2014

[16] Q-X Li C-X Liu C-P Chun et al ldquoChromosomal imbalancesrevealed in primary rhabdomyo-sarcomas by comparativegenomic hybridizationrdquo Chinese Medical Journal vol 122 no11 pp 1277ndash1282 2009

[17] E Missiaglia J Selfe M Hamdi et al ldquoGenomic imbalancesin rhabdomyosarcoma cell lines affect expression of genesfrequently altered in primary tumors an approach to identifycandidate genes involved in tumor developmentrdquo Genes Chro-mosomes and Cancer vol 48 no 6 pp 455ndash467 2009

12 Sarcoma

[18] F G Barr F Duan L M Smith et al ldquoGenomic and clinicalanalyses of 2p24 and 12q13-q14 amplification in alveolar rhab-domyosarcoma a report from the childrenrsquos oncology grouprdquoGenes Chromosomes and Cancer vol 48 no 8 pp 661ndash6722009

[19] T Gordon AMcManus J Anderson et al ldquoCytogenetic abnor-malities in 42 rhabdomyosarcoma a United Kingdom CancerCytogenetics Group StudyrdquoMedical and Pediatric Oncology vol36 no 2 pp 259ndash267 2001

[20] X Chen E Stewart A A Shelat et al ldquoTargeting oxidativestress in embryonal rhabdomyosarcomardquo Cancer Cell vol 24no 6 pp 710ndash724 2013

[21] S Lizard-Nacol F Mugneret C Volk C Turc-Carel M Favrotand T Philip ldquoTranslocation (213)(q37q14) in alveolar rhab-domyosarcoma a new caserdquo Cancer Genetics and Cytogeneticsvol 25 no 2 pp 373ndash374 1987

[22] P H B Sorensen J C Lynch S J Qualman et al ldquoPAX3-FKHR and PAX7-FKHR gene fusions are prognostic indicatorsin alveolar rhabdomyosarcoma a report from the childrenrsquosoncology grouprdquo Journal of Clinical Oncology vol 20 no 11 pp2672ndash2679 2002

[23] A D Marshall and G C Grosveld ldquoAlveolar rhabdomyo-sarcomamdashthe molecular drivers of PAX37-FOXO1-inducedtumorigenesisrdquo Skeletal Muscle vol 2 no 1 article 25 2012

[24] E Missiaglia D Williamson J Chisholm et al ldquoPAX3FOXO1fusion gene status is the key prognostic molecular markerin rhabdomyosarcoma and significantly improves current riskstratificationrdquo Journal of Clinical Oncology vol 30 no 14 pp1670ndash1677 2012

[25] J F Shern L Chen J Chmielecki et al ldquoComprehensivegenomic analysis of rhabdomyosarcoma reveals a landscape ofalterations affecting a common genetic axis in fusion-positiveand fusion-negative tumorsrdquo Cancer Discovery vol 4 no 2 pp216ndash231 2014

[26] MWachtel M Dettling E Koscielniak et al ldquoGene expressionsignatures identify rhabdomyosarcoma subtypes and detect anovel t(22)(q35p23) translocation fusing PAX3 to NCOA1rdquoCancer Research vol 64 no 16 pp 5539ndash5545 2004

[27] E Davicioni M J Anderson F G Finckenstein et al ldquoMolec-ular classification of rhabdomyosarcomamdashgenotypic and phe-notypic determinants of diagnosis a report from the ChildrenrsquosOncology Grouprdquo The American Journal of Pathology vol 174no 2 pp 550ndash564 2009

[28] M Visser C Sijmons J Bras et al ldquoAllelotype of pediatric rhab-domyosarcomardquo Oncogene vol 15 no 11 pp 1309ndash1314 1997

[29] R Gil-Benso T San-Miguel R C Callaghan et al ldquoChromo-somal and genetic changes produced in tumoral progression ofembryonal rhabdomyosarcomardquo Histopathology vol 62 no 5pp 816ndash819 2013

[30] S X Skapek J Anderson F G Barr et al ldquoPAX-FOXO1 fusionstatus drives unfavorable outcome for children with rhabdom-yosarcoma a childrenrsquos oncology group reportrdquo Pediatric Bloodand Cancer vol 60 no 9 pp 1411ndash1417 2013

[31] HHosoi N Kakazu E Konishi et al ldquoA novel PAX3 rearrange-ment in embryonal rhabdomyosarcomardquo Cancer Genetics andCytogenetics vol 189 no 2 pp 98ndash104 2009

[32] E Takaoka H Sonobe K Akimaru et al ldquoMultiple sites ofhighly amplified DNA sequences detected bymolecular cytoge-netic analysis inHS-RMS-2 a newpleomorphic rhabdomyosar-coma cell linerdquo American Journal of Cancer Research vol 2 no2 pp 141ndash152 2012

[33] K Szuhai D de Jong W Y Leung C D M Fletcher and P CWHogendoorn ldquoTransactivatingmutation of theMYOD1 geneis a frequent event in adult spindle cell rhabdomyosarcomardquoJournal of Pathology vol 232 no 3 pp 300ndash307 2014

[34] N P Agaram C-L Chen L Zhang M P Laquaglia L Wexlerand C R Antonescu ldquoRecurrent MYOD1 mutations in pedi-atric and adult sclerosing and spindle cell rhabdomyosarcomasEvidence for a common pathogenesisrdquoGenes Chromosomes andCancer vol 53 no 9 pp 779ndash787 2014

[35] H Yoshida M Miyachi K Sakamoto et al ldquoPAX3-NCOA2fusion gene has a dual role in promoting the proliferation andinhibiting the myogenic differentiation of rhabdomyosarcomacellsrdquo Oncogene vol 33 pp 5601ndash5608 2013

[36] J M Mosquera A Sboner L Zhang et al ldquoRecurrent NCOA2gene rearrangements in congenitalinfantile spindle cell rhab-domyosarcomardquo Genes Chromosomes and Cancer vol 52 no6 pp 538ndash550 2013

[37] V Thavaraj A Sethi and L S Arya ldquoIncomplete Beckwith-Wiedemann syndrome in a child with orbital rhabdomyosar-comardquo Indian Pediatrics vol 39 no 3 pp 299ndash304 2002

[38] D Walter S Satheesha P Albrecht et al ldquoCD133 positiveembryonal rhabdomyosarcoma stem-like cell population isenriched in rhabdospheresrdquo PLoS ONE vol 6 no 5 Article IDe19506 2011

[39] Y-X Ren F G Finckenstein D A Abdueva et al ldquoMousemesenchymal stem cells expressing PAX-FKHR form alveolarrhabdomyosarcomas by cooperating with secondary muta-tionsrdquo Cancer Research vol 68 no 16 pp 6587ndash6597 2008

[40] M Salerno S Avnet G Bonuccelli S Hosogi D Granchi andN Baldini ldquoImpairment of lysosomal activity as a therapeuticmodality targeting cancer stem cells of embryonal rhabdom-yosarcoma cell line RDrdquo PLoS ONE vol 9 no 10 Article IDe110340 2014

[41] K Tanoue Y Wang M Ikeda et al ldquoSurvivin-responsive con-ditionally replicating adenovirus kills rhabdomyosarcoma stemcells more efficiently than their progenyrdquo Journal of Transla-tional Medicine vol 12 no 1 article 27 2014

[42] P K Rao EMissiaglia L Shields et al ldquoDistinct roles formiR-1and miR-133a in the proliferation and differentiation of rhab-domyosarcoma cellsrdquo The FASEB Journal vol 24 no 9 pp3427ndash3437 2010

[43] D Yan X D Dong X Chen et al ldquoMicroRNA-1206 targets c-met and inhibits rhabdomyosarcomadevelopmentrdquoThe Journalof Biological Chemistry vol 284 no 43 pp 29596ndash29604 2009

[44] I Marchesi A Giordano and L Bagella ldquoRoles of enhancerof zeste homolog 2 from skeletal muscle differentiation torhabdomyosarcoma carcinogenesisrdquoCell Cycle vol 13 no 4 pp516ndash527 2014

[45] M M Sun J F Li L L Guo et al ldquoTGF-1205731 suppression ofmicroRNA-450b-5p expression a novel mechanism for block-ingmyogenic differentiation of rhabdomyosarcomardquoOncogenevol 33 no 16 pp 2075ndash2086 2014

[46] Y Diao X Guo L Jiang et al ldquoMiR-203 a tumor suppressorfrequently down-regulated by promoter hypermethylation inrhabdomyosarcomardquo The Journal of Biological Chemistry vol289 no 1 pp 529ndash539 2014

[47] H-J Huang J Liu HHua et al ldquoMiR-214 andN-ras regulatoryloop suppresses rhabdomyosarcoma cell growth and xenografttumorigenesisrdquo Oncotarget vol 5 no 8 pp 2161ndash2175 2014

Sarcoma 13

[48] A L Sarver L Li and S Subramanian ldquomicroRNA miR-183functions as an oncogene by targeting the transcription factorEGR1 and promoting tumor cell migrationrdquo Cancer Researchvol 70 no 23 pp 9570ndash9580 2010

[49] R L Galindo J A Allport and E N Olson ldquoA Drosophilamodel of the rhabdomyosarcoma initiator PAX7-FKHRrdquo Pro-ceedings of the National Academy of Sciences of the United Statesof America vol 103 no 36 pp 13439ndash13444 2006

[50] U Avirneni-Vadlamudi K A Galindo T R Endicott V Paul-son S Cameron and R L Galindo ldquoDrosophila and mammal-ian models uncover a role for the myoblast fusion gene TANC1in rhabdomyosarcomardquo The Journal of Clinical Investigationvol 122 no 1 pp 403ndash407 2012

[51] E Y Chen andDM Langenau ldquoZebrafishmodels of rhabdom-yosarcomardquoMethods in Cell Biology vol 105 pp 383ndash402 2011

[52] X Le E K Pugach S Hettmer et al ldquoA novel chemicalscreening strategy in zebrafish identifies common pathways inembryogenesis and rhabdomyosarcoma developmentrdquo Devel-opment vol 140 no 11 pp 2354ndash2364 2012

[53] Y Lee R Kawagoe K Sasai et al ldquoLoss of suppressor-of-fusedfunction promotes tumorigenesisrdquoOncogene vol 26 no 44 pp6442ndash6447 2007

[54] C Keller B R Arenkiel C M Coffin N El-Bardeesy R ADePinho and M R Capecchi ldquoAlveolar rhabdomyosarcomasin conditional Pax3Fkhr mice cooperativity of Ink4aARF andTrp53 loss of functionrdquo Genes and Development vol 18 no 21pp 2614ndash2626 2004

[55] S Singh CVinson CMGurley et al ldquoImpairedWnt signalingin embryonal rhabdomyosarcoma cells from p53c-fos doublemutant micerdquo American Journal of Pathology vol 177 no 4 pp2055ndash2066 2010

[56] G J Schaaf J M Ruijter F Van Ruissen et al ldquoFull transcrip-tome analysis of rhabdomyosarcoma normal and fetal skeletalmuscle statistical comparison of multiple SAGE librariesrdquo TheFASEB Journal vol 19 no 3 pp 404ndash406 2005

[57] D S Hawkins A A Gupta and E R Rudzinski ldquoWhat is newin the biology and treatment of pediatric rhabdomyosarcomardquoCurrent Opinion in Pediatrics vol 26 no 1 pp 50ndash56 2014

[58] M Zhang J Truscott and J Davie ldquoLoss of MEF2D expressioninhibits differentiation and contributes to oncogenesis in rhab-domyosarcoma cellsrdquoMolecular Cancer vol 12 no 1 article 1502013

[59] M Ciesla J Dulak and A Jozkowicz ldquomicroRNAs and epi-genetic mechanisms of rhabdomyosarcoma developmentrdquo TheInternational Journal of Biochemistry amp Cell Biology vol 53 pp482ndash492 2014

[60] S Ricaud B Vernus M Duclos et al ldquoInhibition of autocrinesecretion of myostatin enhances terminal differentiation inhuman rhabdomyosarcoma cellsrdquo Oncogene vol 22 no 51 pp8221ndash8232 2003

[61] C S Guo C Degnin T A Fiddler D Stauffer andM JThayerldquoRegulation of MyoD activity and muscle cell differentiation byMDM2 pRb and Sp1rdquoThe Journal of Biological Chemistry vol278 no 25 pp 22615ndash22622 2003

[62] S Rossi E Stoppani P L Puri and A Fanzani ldquoDifferenti-ation of human rhabdomyosarcoma RD cells is regulated byreciprocal functional interactions between myostatin p38 andextracellular regulated kinase signalling pathwaysrdquo EuropeanJournal of Cancer vol 47 no 7 pp 1095ndash1105 2011

[63] Z S Walters B Villarejo-Balcells D Olmos et al ldquoJARID2is a direct target of the PAX3-FOXO1 fusion protein andinhibits myogenic differentiation of rhabdomyosarcoma cellsrdquoOncogene vol 33 no 9 pp 1148ndash1157 2014

[64] T Hosoyama M I Aslam J Abraham et al ldquoIL-4R drives ded-ifferentiation mitogenesis and metastasis in rhabdomyosar-comardquo Clinical Cancer Research vol 17 no 9 pp 2757ndash27662011

[65] F S Dela Cruz ldquoCancer stem cells in pediatric sarcomasrdquoFrontiers in Oncology vol 3 article 168 2013

[66] M J Bissell and W C Hines ldquoWhy donrsquot we get more cancerA proposed role of the microenvironment in restraining cancerprogressionrdquo Nature Medicine vol 17 no 3 pp 320ndash329 2011

[67] E Charytonowicz C Cordon-Cardo I Matushansky and MZiman ldquoAlveolar rhabdomyosarcoma is the cell of origin amesenchymal stem cellrdquoCancer Letters vol 279 no 2 pp 126ndash136 2009

[68] W K Cavenee ldquoMuscling in on rhabdomyosarcomardquo NatureMedicine vol 8 no 11 pp 1200ndash1201 2002

[69] B Zhu and J K Davie ldquoNew insights into signalling-pathwayalterations in rhabdomyosarcomardquo British Journal of Cancervol 112 no 2 pp 227ndash231 2015

[70] M Zhang C M Linardic and D G Kirsch ldquoRAS and ROS inrhabdomyosarcomardquo Cancer Cell vol 24 no 6 pp 689ndash6912013

[71] S C Marianna L Ianzano and G Nicoletti ldquoTumor sup-pressor genes promote rhabdomyosarcoma progression in p53heterozygousrdquo Oncotarget vol 5 no 1 pp 108ndash119 2013

[72] SWang L Guo L Dong et al ldquoTGF-beta1 signal pathwaymaycontribute to rhabdomyosarcoma development by inhibitingdifferentiationrdquo Cancer Science vol 101 no 5 pp 1108ndash11162010

[73] J Wesche K Haglund and E M Haugsten ldquoFibroblast growthfactors and their receptors in cancerrdquo Biochemical Journal vol437 no 2 pp 199ndash213 2011

[74] M Wachtel J Rakic M Okoniewski P Bode F Niggli andB W Schafer ldquoFGFR4 signaling couples to Bim and not Bmfto discriminate subsets of alveolar rhabdomyosarcoma cellsrdquoInternational Journal of Cancer vol 135 no 7 pp 1543ndash15522014

[75] P L Puri ZWu P Zhang et al ldquoInduction of terminal differen-tiation by constitutive activation of p38 MAP kinase in humanrhabdomyosarcoma cellsrdquo Genes and Development vol 14 no5 pp 574ndash584 2000

[76] A M Tremblay E Missiaglia G G Galli et al ldquoThe Hippotransducer YAP1 transforms activated satellite cells and is apotent effector of embryonal rhabdomyosarcoma formationrdquoCancer Cell vol 26 no 2 pp 273ndash287 2014

[77] R N Judson A M Tremblay P Knopp et al ldquoThe hippo path-way member Yap plays a key role in influencing fate decisionsin muscle satellite cellsrdquo Journal of Cell Science vol 125 no 24pp 6009ndash6019 2012

[78] L E S Crose K A Galindo J G Kephart et al ldquoAlve-olar rhabdomyosarcoma-associated PAX3-FOXO1 promotestumorigenesis via Hippo pathway suppressionrdquo Journal ofClinical Investigation vol 124 no 1 pp 285ndash296 2014

[79] B Belyea J G Kephart J Blum D G Kirsch and C MLinardic ldquoEmbryonic signaling pathways and rhabdomyosar-coma contributions to cancer development and opportunitiesfor therapeutic targetingrdquo Sarcoma vol 2012 Article ID 40623913 pages 2012

14 Sarcoma

[80] J Huertas-Martinez S Rello-Varona D Herrero-Martin et alldquoCaveolin-1 is down-regulated in alveolar rhabdomyosarcomasand negatively regulates tumor growthrdquo Oncotarget vol 5 no20 pp 9744ndash9755 2014

[81] S Rossi P L Poliani CMissale EMonti andA Fanzani ldquoCav-eolins in rhabdomyosarcomardquo Journal of Cellular andMolecularMedicine vol 15 no 12 pp 2553ndash2568 2011

[82] P G Tsioli E S Patsouris C Giaginis and S E TheocharisldquoDNA repair systems in rhabdomyosarcomardquo Histology andHistopathology vol 28 no 8 pp 971ndash984 2013

[83] E Gatzidou C Michailidi S Tseleni-Balafouta and S Theo-charis ldquoAn epitome of DNA repair related genes and mecha-nisms in thyroid carcinomardquo Cancer Letters vol 290 no 2 pp139ndash147 2010

[84] R Rota R Ciarapica A Giordano L Miele and F LocatellildquoMicroRNAs in rhabdomyosarcoma pathogenetic implicationsand translational potentialityrdquoMolecular Cancer vol 10 article120 2011

[85] A Gougelet J Perez D Pissaloux et al ldquoMiRNA profiling howto bypass the current difficulties in the diagnosis and treatmentof sarcomasrdquo Sarcoma vol 2011 Article ID 460650 13 pages2011

[86] R Ciarapica G Russo F Verginelli et al ldquoDeregulated expres-sion of miR-26a and Ezh2 in rhabdomyosarcomardquo Cell Cyclevol 8 no 1 pp 172ndash175 2009

[87] L Li A L Sarver S Alamgir and S Subramanian ldquoDownreg-ulation of microRNAs miR-1 -206 and -29 stabilizes PAX3 andCCND2 expression in rhabdomyosarcomardquo Laboratory Investi-gation vol 92 no 4 pp 571ndash583 2012

[88] S-Y Ng G K Bogu B Soh and L W Stanton ldquoThe longnoncoding RNA RMST interacts with SOX2 to regulate neu-rogenesisrdquoMolecular Cell vol 51 no 3 pp 349ndash359 2013

[89] C A Lynch B Tycko T H Bestor and C P Walsh ldquoReac-tivation of a silenced H19 gene in human rhabdomyosarcomaby demethylation of DNA but not by histone hyperacetylationrdquoMolecular Cancer vol 1 article 2 2002

[90] P Onyango and A P Feinberg ldquoA nucleolar protein H19opposite tumor suppressor (HOTS) is a tumor growth inhibitorencoded by a human imprinted H19 antisense transcriptrdquoProceedings of the National Academy of Sciences of the UnitedStates of America vol 108 no 40 pp 16759ndash16764 2011

[91] C E Albacker N Y Storer E M Langdon et al ldquoThe his-tone methyltransferase SUV39H1 suppresses embryonal rhab-domyosarcoma formation in zebrafishrdquo PLoS ONE vol 8 no 5Article ID e64969 2013

[92] N Y Storer R M White A Uong et al ldquoZebrafish rhab-domyosarcoma reflects the developmental stage of oncogeneexpression during myogenesisrdquo Development vol 140 no 14pp 3040ndash3050 2013

[93] M E Hatley W Tang M R Garcia et al ldquoA mouse modelof rhabdomyosarcoma originating from the adipocyte lineagerdquoCancer Cell vol 22 no 4 pp 536ndash546 2012

[94] V Hosur A Kavirayani J Riefler et al ldquoDystrophin and dysfer-lin doublemutantmice a novelmodel for rhabdomyosarcomardquoCancer Genetics vol 205 no 5 pp 232ndash241 2012

[95] W Goessling T E North and L I Zon ldquoNew waves of discov-ery modeling cancer in zebrafishrdquo Journal of Clinical Oncologyvol 25 no 17 pp 2473ndash2479 2007

[96] B P Rubin K Nishijo H-I H Chen et al ldquoEvidence for anunanticipated relationship between undifferentiated pleomor-phic sarcoma and embryonal rhabdomyosarcomardquo Cancer Cellvol 19 no 2 pp 177ndash191 2011

[97] S Z Kaylani J Xu R K Srivastava L Kopelovich J G Presseyand M Athar ldquoRapamycin targeting mTOR and hedgehog sig-naling pathways blocks human rhabdomyosarcoma growth inxenograft murine modelrdquo Biochemical and Biophysical ResearchCommunications vol 435 no 4 pp 557ndash561 2013

[98] S Q Li A T Cheuk J F Shern et al ldquoTargeting wild-type andmutationally activated FGFR4 in rhabdomyosarcoma with theinhibitor ponatinib (AP24534)rdquoPLoSONE vol 8 no 10 ArticleID e76551 2013

[99] K Miyoshi K Kohashi F Fushimi et al ldquoClose correlationbetween CXCR4 and VEGF expression and frequent CXCR7expression in rhabdomyosarcomardquo Human Pathology vol 45no 9 pp 1900ndash1909 2014

[100] ADMarshall F Picchione R I KGeltink andGCGrosveldldquoPAX3-FOXO1 induces up-regulation of Noxa sensitizing alve-olar rhabdomyosarcoma cells to apoptosisrdquo Neoplasia vol 15no 7 pp 738ndash748 2013

[101] F Perut S Avnet C Fotia et al ldquoV-ATPase as an effective thera-peutic target for sarcomasrdquo Experimental Cell Research vol 320no 1 pp 21ndash32 2014

Submit your manuscripts athttpwwwhindawicom

Stem CellsInternational

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

MEDIATORSINFLAMMATION

of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Behavioural Neurology

EndocrinologyInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Disease Markers

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

BioMed Research International

OncologyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Oxidative Medicine and Cellular Longevity

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

PPAR Research

The Scientific World JournalHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Immunology ResearchHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal of

ObesityJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Computational and Mathematical Methods in Medicine

OphthalmologyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Diabetes ResearchJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Research and TreatmentAIDS

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Gastroenterology Research and Practice

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Parkinsonrsquos Disease

Evidence-Based Complementary and Alternative Medicine

Volume 2014Hindawi Publishing Corporationhttpwwwhindawicom

Page 10: Review Article Rhabdomyosarcoma: Advances in Molecular and

10 Sarcoma

63 Mouse Models in RMS Mouse models established fromin vivo studies may contribute to the understanding of thegenetic basis in tumor development and progression andmayhelp to test the efficacies of novel antineoplastic agents

The importance of PAX3PAX7-FKHR fusion proteins inthe progression of ARMS tumorigenesis is evident from bothin vitro and in vivo studies Transgenic mouse models gener-ated with the PAX-FKHR fusion genemimic the formation ofhuman ARMS [67] A mouse model was developed with het-erozygous loss of Sufu a tumor suppressor inHedgehog (Hh)signaling combined with p53 loss driven ERMS with a lowpenetrance (9) [53] These studies indicate that the modelsconsistently formingARMSnot only need the introduction ofthe PAX-FKHR fusion gene but also need to be accompaniedby other genetic events such as p53 pathway disruption [54]

Compared with ARMS ERMS models are more complexto generate because of their lower tumor penetrance andlonger latency JW41 cells from the p53c-fos double mutantmice models resemble human ERMS cells morphologicallyand express similar characteristic markers [55] The overex-pression of the Wnt2 gene identified in JW41 cells confirmedin human RMS cell lines can allow for further analysis of theWnt signaling pathway Both of these results indicate that thedownregulation of theWnt pathway contributes to resistanceto apoptosis and the inhibition of myogenic differentiation inERMS

Using Ptch1 p53 andor Rb1 conditional mouse modelsand controlling prenatal or postnatal myogenic cell of originRubin et al demonstrated that the loss of p53 in maturingmyoblasts related to tumorigenesis of ERMS [96] They alsoindicated that Rb1 alteration with other oncogenic factorswas strongly associated with an undifferentiated phenotypeacting as a modifier In addition they highlighted a sub-set of p53-deficient ERMSs arising from Myf6-expressingmyoblasts which had the same latency with Pax7-expressingmurine satellite cells in the study

7 Potential Therapeutic Targets in RMS

Detection of genomic imbalances and identification of thecrucial effective genes can contribute to identifying novelpotential biomarkers and relevant targets for clinical therapyin RMS

Survivin-responsive conditionally replicating adenovi-ruses can regulate multiple factors (Survm-CRAs) [41]Although Survm-CRA can efficiently replicate and potentlyinduce cell death in RMS cells the cytotoxic effects are morepronounced in RSC-enriched or RSC-purified cells thanin RSC-exiguous or progeny-purified cells Injections ofSurvm-CRAs into tumor nodules generated by transplantingRSC-enriched cells induce significant death in RMS cells andregression of tumor nodulesThe unique therapeutic featuresof Survm-CRA include not only its therapeutic effectivenessagainst RMS but also its increasing effectiveness against CSCsuggesting that Survm-CRA may be a promising anticanceragent [41]

Rapamycin an inhibitor of mTOR can abrogate RMStumor growth in a xenograft mouse model [97] Thetumors in the rapamycin-treatedmice group show significant

reduction of proliferation and invasiveness and inductionof apoptosis The tumor growth inhibition is simultaneousassociated with the diminution of mTOR and Hh pathwayswhich are implicated in the pathogenesis of RMSThe resultsindicate that using rapamycin either alone or in combinationwith traditional chemotherapeutic drugs may represent apotential targeted agent for therapeutic intervention Ampli-fication and mutational activation of Fibroblast Growth Fac-tor Receptor 4 (FGFR4) in RMS cells promote tumor pro-gression [98] Ponatinib is the most potent FGFR4 inhibitorto block wild-type RMS cell growth mutate FGFR4 throughincreasing apoptosis and suppress the FGFR4 downstreamtarget STAT3 Ponatinib treatment slows tumor growth inRMS mouse models expressing mutated FGFR4 [98] Pona-tinib as an FGFR4 inhibitor shows potential to act as aneffective therapeutic agent for RMS tumorsThe expression ofCXC chemokine receptor 4 (CXCR4) CXCR7 and VascularEndothelial Growth Factor (VEGF) indicates poor prognosisin a variety of malignancies including RMS cell lines Mostsamples in a large series of clinical RMS cases show asso-ciation with high expression of these antagonists regardlessof their subtypes [99] However there are significant corre-lations with high expression of CXCR4 and VEGF in bothERMS and ARMS High VEGF expression is predictive ofadverse prognostic factors in RMS as the expression of theseangiogenesis factors was compared with clinicopathologicalparameters and prognosis Considering their overexpressionin RMS these chemokine receptors and VEGF could providepotential molecular therapeutic targets in RMS Some ARMScell lines have undergone apoptosis in response to antineo-plastic drugs such as bortezomib The proapoptotic BH3only family member NOXA acts as an upregulated proteindownstream PAX3-FKHR resulting in increased cell sensi-tivity to bortezomib Apoptosis in response to bortezomibcan be reversed by shRNA knockdown of Noxa PAX3-FKHR upregulation of Noxa creates a potential therapeuticinsight into inducing apoptosis in ARMS cellsThis apoptosispathway could represent a specific targeted therapy againstPAX3-FKHR-expressing ARMS cells [100]

Proton pump activation resulting from intracellular acid-ificationmay be an energetic mechanism to escape apoptosisV-ATPase is an ATP-driven proton pump acidifying theintracellular compartment and transports protons across theplasma membrane Esomeprazole as a V-ATPase inhibitor inthe PPI administration interferes with intensive ion trans-porter activity of RMS cells and induces remarkable cytotoxi-city RMS expresses a higher level ofV-ATPase comparedwithother sarcomasTherefore RMS should be very susceptible toesomeprazole treatment and V-ATPase could be consideredas a promising selective target for treatment [101]

8 Conclusion and Future Prospects

For RMS the current challenges include how to iden-tify promising novel therapeutics and integrate them intoexisting therapy For better understanding of congenitaland epigenetic modifications in the development of RMSthis review summarizes the recent genome-wide studies onmolecular and genetic alterations to decipher the underlying

Sarcoma 11

tumorigenesis mechanisms There is a link between geneticand epigenetic alterations responsible for a variety of tumorcell growth proliferation differentiation apoptosis andtherapy-resistance mechanisms The identification of theprognostic value of the PAX-FKHR fusion status in RMSis one of the most important shifts in the lineage and therisk assessment of RMS which exerts an oncogenic effectthrough multiple pathways and is incorporated into mostrelevant studies The misbalanced expression of a number ofmiRs involved in the regulation of myogenic differentiationalso implicates the tumorsrsquo escape suppressive mechanismsMoreover we described several in vivo models of RMSto explore underlying histogenesis and different aspects oftumorigenesis Recent work on CSCs has also contributedto understanding the cell origins refining molecular char-acterization and elucidating the underlying basis of refrac-tory responses to current adjuvant treatments of subsets oftumorsThese integrative approaches can provide opportuni-ties to identify diagnostic and prognostic biomarkers appliedto the individual targeted therapy and significantly improveRMS prognosis

Abbreviations

RMS RhabdomyosarcomaARMS Alveolar rhabdomyosarcomaERMS Embryonal rhabdomyosarcomaPRMS Pleomorphic rhabdomyosarcomaSRMS Sclerosingspindle cell

rhabdomyosarcomamiRNA miR MicroRNACSC Cancer stem cellMSC Mesenchymal stem cell

Conflict of Interests

The authors declare that there is no conflict of interestsregarding the publication of this paper

Acknowledgments

This work was supported in part by grants from the Gat-tegno and Wechsler funds Dr Duan was supported in partthrough a grant from Sarcoma Foundation of America (SFA)a grant from National Cancer Institute (NCI)National Insti-tutes of Health (NIH) UO1 CA 151452 and a grant from anAcademic Enrichment Fund of MGH Orthopedic Surgery

References

[1] L Yang T Takimoto and J Fujimoto ldquoPrognostic model forpredicting overall survival in children and adolescents withrhabdomyosarcomardquo BMC Cancer vol 14 no 1 article 6542014

[2] D Egas-Bejar and W W Huh ldquoRhabdomyosarcoma in adoles-cent and young adult patients current perspectivesrdquo AdolescentHealth Medicine andTherapeutics vol 5 pp 115ndash125 2014

[3] J C Breneman E Lyden A S Pappo et al ldquoPrognosticfactors and clinical outcomes in children and adolescents withmetastatic rhabdomyosarcomamdasha report from the intergrouprhabdomyosarcoma study IVrdquo Journal of Clinical Oncology vol21 no 1 pp 78ndash84 2003

[4] V D Soleimani and M A Rudnicki ldquoNew insights into theorigin and the genetic basis of rhabdomyosarcomasrdquo CancerCell vol 19 no 2 pp 157ndash159 2011

[5] T Glass D D Cochrane S R Rassekh K Goddard and JHukin ldquoGrowing teratoma syndrome in intracranial non-ger-minomatous germ cell tumors (iNGGCTs) a risk for secondarymalignant transformationmdasha report of two casesrdquo Childrsquos Ner-vous System vol 30 no 5 pp 953ndash957 2014

[6] J Ji C Eng and K Hemminki ldquoFamilial risk for soft tissuetumors a nation-wide epidemiological study from SwedenrdquoJournal of Cancer Research and Clinical Oncology vol 134 no5 pp 617ndash624 2008

[7] C D M Fletcher J A Bridge P Hogendoorn and F MertensWHO Classification of Tumours of Soft Tissue and Bone vol 5IARC Press Paris France 4th edition 2013

[8] I Sultan I Qaddoumi S Yaser C Rodriguez-Galindo and AFerrari ldquoComparing adult and pediatric rhabdomyosarcoma inthe surveillance epidemiology and end results program 1973 to2005 an analysis of 2600 patientsrdquo Journal of Clinical Oncologyvol 27 no 20 pp 3391ndash3397 2009

[9] B Raney W Huh D Hawkins et al ldquoOutcome of patientswith localized orbital sarcoma who relapsed following treat-ment on Intergroup Rhabdomyosarcoma Study Group (IRSG)Protocols-III and -IV 1984ndash1997 a report from the ChildrenrsquosOncology Grouprdquo Pediatric Blood and Cancer vol 60 no 3 pp371ndash376 2013

[10] W Crist E A Gehan A H Ragab et al ldquoThe third intergrouprhabdomyosarcoma studyrdquo Journal of Clinical Oncology vol 13no 3 pp 610ndash630 1995

[11] C Liu D Li J Jiang et al ldquoAnalysis of molecular cytogeneticalteration in rhabdomyosarcoma by array comparative genomichybridizationrdquo PLoS ONE vol 9 no 4 Article ID e94924 2014

[12] DWilliamson E Missiaglia A de Reynies et al ldquoFusion gene-negative alveolar rhabdomyosarcoma is clinically and molec-ularly indistinguishable from embryonal rhabdomyosarcomardquoJournal of Clinical Oncology vol 28 no 13 pp 2151ndash2158 2010

[13] V Paulson G Chandler D Rakheja et al ldquoHigh-resolutionarray CGH identifies common mechanisms that drive embry-onal rhabdomyosarcoma pathogenesisrdquo Genes Chromosomesand Cancer vol 50 no 6 pp 397ndash408 2011

[14] S Park J Lee I-G Do et al ldquoAberrant CDK4 amplificationin refractory rhabdomyosarcoma as identified by genomicprofilingrdquo Scientific Reports vol 4 article 3623 2014

[15] C Liu D Li J Hu et al ldquoChromosomal and genetic imbalancesin Chinese patients with rhabdomyosarcoma detected by high-resolution array comparative genomic hybridizationrdquo Interna-tional Journal of Clinical and Experimental Pathology vol 7 no2 pp 690ndash698 2014

[16] Q-X Li C-X Liu C-P Chun et al ldquoChromosomal imbalancesrevealed in primary rhabdomyo-sarcomas by comparativegenomic hybridizationrdquo Chinese Medical Journal vol 122 no11 pp 1277ndash1282 2009

[17] E Missiaglia J Selfe M Hamdi et al ldquoGenomic imbalancesin rhabdomyosarcoma cell lines affect expression of genesfrequently altered in primary tumors an approach to identifycandidate genes involved in tumor developmentrdquo Genes Chro-mosomes and Cancer vol 48 no 6 pp 455ndash467 2009

12 Sarcoma

[18] F G Barr F Duan L M Smith et al ldquoGenomic and clinicalanalyses of 2p24 and 12q13-q14 amplification in alveolar rhab-domyosarcoma a report from the childrenrsquos oncology grouprdquoGenes Chromosomes and Cancer vol 48 no 8 pp 661ndash6722009

[19] T Gordon AMcManus J Anderson et al ldquoCytogenetic abnor-malities in 42 rhabdomyosarcoma a United Kingdom CancerCytogenetics Group StudyrdquoMedical and Pediatric Oncology vol36 no 2 pp 259ndash267 2001

[20] X Chen E Stewart A A Shelat et al ldquoTargeting oxidativestress in embryonal rhabdomyosarcomardquo Cancer Cell vol 24no 6 pp 710ndash724 2013

[21] S Lizard-Nacol F Mugneret C Volk C Turc-Carel M Favrotand T Philip ldquoTranslocation (213)(q37q14) in alveolar rhab-domyosarcoma a new caserdquo Cancer Genetics and Cytogeneticsvol 25 no 2 pp 373ndash374 1987

[22] P H B Sorensen J C Lynch S J Qualman et al ldquoPAX3-FKHR and PAX7-FKHR gene fusions are prognostic indicatorsin alveolar rhabdomyosarcoma a report from the childrenrsquosoncology grouprdquo Journal of Clinical Oncology vol 20 no 11 pp2672ndash2679 2002

[23] A D Marshall and G C Grosveld ldquoAlveolar rhabdomyo-sarcomamdashthe molecular drivers of PAX37-FOXO1-inducedtumorigenesisrdquo Skeletal Muscle vol 2 no 1 article 25 2012

[24] E Missiaglia D Williamson J Chisholm et al ldquoPAX3FOXO1fusion gene status is the key prognostic molecular markerin rhabdomyosarcoma and significantly improves current riskstratificationrdquo Journal of Clinical Oncology vol 30 no 14 pp1670ndash1677 2012

[25] J F Shern L Chen J Chmielecki et al ldquoComprehensivegenomic analysis of rhabdomyosarcoma reveals a landscape ofalterations affecting a common genetic axis in fusion-positiveand fusion-negative tumorsrdquo Cancer Discovery vol 4 no 2 pp216ndash231 2014

[26] MWachtel M Dettling E Koscielniak et al ldquoGene expressionsignatures identify rhabdomyosarcoma subtypes and detect anovel t(22)(q35p23) translocation fusing PAX3 to NCOA1rdquoCancer Research vol 64 no 16 pp 5539ndash5545 2004

[27] E Davicioni M J Anderson F G Finckenstein et al ldquoMolec-ular classification of rhabdomyosarcomamdashgenotypic and phe-notypic determinants of diagnosis a report from the ChildrenrsquosOncology Grouprdquo The American Journal of Pathology vol 174no 2 pp 550ndash564 2009

[28] M Visser C Sijmons J Bras et al ldquoAllelotype of pediatric rhab-domyosarcomardquo Oncogene vol 15 no 11 pp 1309ndash1314 1997

[29] R Gil-Benso T San-Miguel R C Callaghan et al ldquoChromo-somal and genetic changes produced in tumoral progression ofembryonal rhabdomyosarcomardquo Histopathology vol 62 no 5pp 816ndash819 2013

[30] S X Skapek J Anderson F G Barr et al ldquoPAX-FOXO1 fusionstatus drives unfavorable outcome for children with rhabdom-yosarcoma a childrenrsquos oncology group reportrdquo Pediatric Bloodand Cancer vol 60 no 9 pp 1411ndash1417 2013

[31] HHosoi N Kakazu E Konishi et al ldquoA novel PAX3 rearrange-ment in embryonal rhabdomyosarcomardquo Cancer Genetics andCytogenetics vol 189 no 2 pp 98ndash104 2009

[32] E Takaoka H Sonobe K Akimaru et al ldquoMultiple sites ofhighly amplified DNA sequences detected bymolecular cytoge-netic analysis inHS-RMS-2 a newpleomorphic rhabdomyosar-coma cell linerdquo American Journal of Cancer Research vol 2 no2 pp 141ndash152 2012

[33] K Szuhai D de Jong W Y Leung C D M Fletcher and P CWHogendoorn ldquoTransactivatingmutation of theMYOD1 geneis a frequent event in adult spindle cell rhabdomyosarcomardquoJournal of Pathology vol 232 no 3 pp 300ndash307 2014

[34] N P Agaram C-L Chen L Zhang M P Laquaglia L Wexlerand C R Antonescu ldquoRecurrent MYOD1 mutations in pedi-atric and adult sclerosing and spindle cell rhabdomyosarcomasEvidence for a common pathogenesisrdquoGenes Chromosomes andCancer vol 53 no 9 pp 779ndash787 2014

[35] H Yoshida M Miyachi K Sakamoto et al ldquoPAX3-NCOA2fusion gene has a dual role in promoting the proliferation andinhibiting the myogenic differentiation of rhabdomyosarcomacellsrdquo Oncogene vol 33 pp 5601ndash5608 2013

[36] J M Mosquera A Sboner L Zhang et al ldquoRecurrent NCOA2gene rearrangements in congenitalinfantile spindle cell rhab-domyosarcomardquo Genes Chromosomes and Cancer vol 52 no6 pp 538ndash550 2013

[37] V Thavaraj A Sethi and L S Arya ldquoIncomplete Beckwith-Wiedemann syndrome in a child with orbital rhabdomyosar-comardquo Indian Pediatrics vol 39 no 3 pp 299ndash304 2002

[38] D Walter S Satheesha P Albrecht et al ldquoCD133 positiveembryonal rhabdomyosarcoma stem-like cell population isenriched in rhabdospheresrdquo PLoS ONE vol 6 no 5 Article IDe19506 2011

[39] Y-X Ren F G Finckenstein D A Abdueva et al ldquoMousemesenchymal stem cells expressing PAX-FKHR form alveolarrhabdomyosarcomas by cooperating with secondary muta-tionsrdquo Cancer Research vol 68 no 16 pp 6587ndash6597 2008

[40] M Salerno S Avnet G Bonuccelli S Hosogi D Granchi andN Baldini ldquoImpairment of lysosomal activity as a therapeuticmodality targeting cancer stem cells of embryonal rhabdom-yosarcoma cell line RDrdquo PLoS ONE vol 9 no 10 Article IDe110340 2014

[41] K Tanoue Y Wang M Ikeda et al ldquoSurvivin-responsive con-ditionally replicating adenovirus kills rhabdomyosarcoma stemcells more efficiently than their progenyrdquo Journal of Transla-tional Medicine vol 12 no 1 article 27 2014

[42] P K Rao EMissiaglia L Shields et al ldquoDistinct roles formiR-1and miR-133a in the proliferation and differentiation of rhab-domyosarcoma cellsrdquo The FASEB Journal vol 24 no 9 pp3427ndash3437 2010

[43] D Yan X D Dong X Chen et al ldquoMicroRNA-1206 targets c-met and inhibits rhabdomyosarcomadevelopmentrdquoThe Journalof Biological Chemistry vol 284 no 43 pp 29596ndash29604 2009

[44] I Marchesi A Giordano and L Bagella ldquoRoles of enhancerof zeste homolog 2 from skeletal muscle differentiation torhabdomyosarcoma carcinogenesisrdquoCell Cycle vol 13 no 4 pp516ndash527 2014

[45] M M Sun J F Li L L Guo et al ldquoTGF-1205731 suppression ofmicroRNA-450b-5p expression a novel mechanism for block-ingmyogenic differentiation of rhabdomyosarcomardquoOncogenevol 33 no 16 pp 2075ndash2086 2014

[46] Y Diao X Guo L Jiang et al ldquoMiR-203 a tumor suppressorfrequently down-regulated by promoter hypermethylation inrhabdomyosarcomardquo The Journal of Biological Chemistry vol289 no 1 pp 529ndash539 2014

[47] H-J Huang J Liu HHua et al ldquoMiR-214 andN-ras regulatoryloop suppresses rhabdomyosarcoma cell growth and xenografttumorigenesisrdquo Oncotarget vol 5 no 8 pp 2161ndash2175 2014

Sarcoma 13

[48] A L Sarver L Li and S Subramanian ldquomicroRNA miR-183functions as an oncogene by targeting the transcription factorEGR1 and promoting tumor cell migrationrdquo Cancer Researchvol 70 no 23 pp 9570ndash9580 2010

[49] R L Galindo J A Allport and E N Olson ldquoA Drosophilamodel of the rhabdomyosarcoma initiator PAX7-FKHRrdquo Pro-ceedings of the National Academy of Sciences of the United Statesof America vol 103 no 36 pp 13439ndash13444 2006

[50] U Avirneni-Vadlamudi K A Galindo T R Endicott V Paul-son S Cameron and R L Galindo ldquoDrosophila and mammal-ian models uncover a role for the myoblast fusion gene TANC1in rhabdomyosarcomardquo The Journal of Clinical Investigationvol 122 no 1 pp 403ndash407 2012

[51] E Y Chen andDM Langenau ldquoZebrafishmodels of rhabdom-yosarcomardquoMethods in Cell Biology vol 105 pp 383ndash402 2011

[52] X Le E K Pugach S Hettmer et al ldquoA novel chemicalscreening strategy in zebrafish identifies common pathways inembryogenesis and rhabdomyosarcoma developmentrdquo Devel-opment vol 140 no 11 pp 2354ndash2364 2012

[53] Y Lee R Kawagoe K Sasai et al ldquoLoss of suppressor-of-fusedfunction promotes tumorigenesisrdquoOncogene vol 26 no 44 pp6442ndash6447 2007

[54] C Keller B R Arenkiel C M Coffin N El-Bardeesy R ADePinho and M R Capecchi ldquoAlveolar rhabdomyosarcomasin conditional Pax3Fkhr mice cooperativity of Ink4aARF andTrp53 loss of functionrdquo Genes and Development vol 18 no 21pp 2614ndash2626 2004

[55] S Singh CVinson CMGurley et al ldquoImpairedWnt signalingin embryonal rhabdomyosarcoma cells from p53c-fos doublemutant micerdquo American Journal of Pathology vol 177 no 4 pp2055ndash2066 2010

[56] G J Schaaf J M Ruijter F Van Ruissen et al ldquoFull transcrip-tome analysis of rhabdomyosarcoma normal and fetal skeletalmuscle statistical comparison of multiple SAGE librariesrdquo TheFASEB Journal vol 19 no 3 pp 404ndash406 2005

[57] D S Hawkins A A Gupta and E R Rudzinski ldquoWhat is newin the biology and treatment of pediatric rhabdomyosarcomardquoCurrent Opinion in Pediatrics vol 26 no 1 pp 50ndash56 2014

[58] M Zhang J Truscott and J Davie ldquoLoss of MEF2D expressioninhibits differentiation and contributes to oncogenesis in rhab-domyosarcoma cellsrdquoMolecular Cancer vol 12 no 1 article 1502013

[59] M Ciesla J Dulak and A Jozkowicz ldquomicroRNAs and epi-genetic mechanisms of rhabdomyosarcoma developmentrdquo TheInternational Journal of Biochemistry amp Cell Biology vol 53 pp482ndash492 2014

[60] S Ricaud B Vernus M Duclos et al ldquoInhibition of autocrinesecretion of myostatin enhances terminal differentiation inhuman rhabdomyosarcoma cellsrdquo Oncogene vol 22 no 51 pp8221ndash8232 2003

[61] C S Guo C Degnin T A Fiddler D Stauffer andM JThayerldquoRegulation of MyoD activity and muscle cell differentiation byMDM2 pRb and Sp1rdquoThe Journal of Biological Chemistry vol278 no 25 pp 22615ndash22622 2003

[62] S Rossi E Stoppani P L Puri and A Fanzani ldquoDifferenti-ation of human rhabdomyosarcoma RD cells is regulated byreciprocal functional interactions between myostatin p38 andextracellular regulated kinase signalling pathwaysrdquo EuropeanJournal of Cancer vol 47 no 7 pp 1095ndash1105 2011

[63] Z S Walters B Villarejo-Balcells D Olmos et al ldquoJARID2is a direct target of the PAX3-FOXO1 fusion protein andinhibits myogenic differentiation of rhabdomyosarcoma cellsrdquoOncogene vol 33 no 9 pp 1148ndash1157 2014

[64] T Hosoyama M I Aslam J Abraham et al ldquoIL-4R drives ded-ifferentiation mitogenesis and metastasis in rhabdomyosar-comardquo Clinical Cancer Research vol 17 no 9 pp 2757ndash27662011

[65] F S Dela Cruz ldquoCancer stem cells in pediatric sarcomasrdquoFrontiers in Oncology vol 3 article 168 2013

[66] M J Bissell and W C Hines ldquoWhy donrsquot we get more cancerA proposed role of the microenvironment in restraining cancerprogressionrdquo Nature Medicine vol 17 no 3 pp 320ndash329 2011

[67] E Charytonowicz C Cordon-Cardo I Matushansky and MZiman ldquoAlveolar rhabdomyosarcoma is the cell of origin amesenchymal stem cellrdquoCancer Letters vol 279 no 2 pp 126ndash136 2009

[68] W K Cavenee ldquoMuscling in on rhabdomyosarcomardquo NatureMedicine vol 8 no 11 pp 1200ndash1201 2002

[69] B Zhu and J K Davie ldquoNew insights into signalling-pathwayalterations in rhabdomyosarcomardquo British Journal of Cancervol 112 no 2 pp 227ndash231 2015

[70] M Zhang C M Linardic and D G Kirsch ldquoRAS and ROS inrhabdomyosarcomardquo Cancer Cell vol 24 no 6 pp 689ndash6912013

[71] S C Marianna L Ianzano and G Nicoletti ldquoTumor sup-pressor genes promote rhabdomyosarcoma progression in p53heterozygousrdquo Oncotarget vol 5 no 1 pp 108ndash119 2013

[72] SWang L Guo L Dong et al ldquoTGF-beta1 signal pathwaymaycontribute to rhabdomyosarcoma development by inhibitingdifferentiationrdquo Cancer Science vol 101 no 5 pp 1108ndash11162010

[73] J Wesche K Haglund and E M Haugsten ldquoFibroblast growthfactors and their receptors in cancerrdquo Biochemical Journal vol437 no 2 pp 199ndash213 2011

[74] M Wachtel J Rakic M Okoniewski P Bode F Niggli andB W Schafer ldquoFGFR4 signaling couples to Bim and not Bmfto discriminate subsets of alveolar rhabdomyosarcoma cellsrdquoInternational Journal of Cancer vol 135 no 7 pp 1543ndash15522014

[75] P L Puri ZWu P Zhang et al ldquoInduction of terminal differen-tiation by constitutive activation of p38 MAP kinase in humanrhabdomyosarcoma cellsrdquo Genes and Development vol 14 no5 pp 574ndash584 2000

[76] A M Tremblay E Missiaglia G G Galli et al ldquoThe Hippotransducer YAP1 transforms activated satellite cells and is apotent effector of embryonal rhabdomyosarcoma formationrdquoCancer Cell vol 26 no 2 pp 273ndash287 2014

[77] R N Judson A M Tremblay P Knopp et al ldquoThe hippo path-way member Yap plays a key role in influencing fate decisionsin muscle satellite cellsrdquo Journal of Cell Science vol 125 no 24pp 6009ndash6019 2012

[78] L E S Crose K A Galindo J G Kephart et al ldquoAlve-olar rhabdomyosarcoma-associated PAX3-FOXO1 promotestumorigenesis via Hippo pathway suppressionrdquo Journal ofClinical Investigation vol 124 no 1 pp 285ndash296 2014

[79] B Belyea J G Kephart J Blum D G Kirsch and C MLinardic ldquoEmbryonic signaling pathways and rhabdomyosar-coma contributions to cancer development and opportunitiesfor therapeutic targetingrdquo Sarcoma vol 2012 Article ID 40623913 pages 2012

14 Sarcoma

[80] J Huertas-Martinez S Rello-Varona D Herrero-Martin et alldquoCaveolin-1 is down-regulated in alveolar rhabdomyosarcomasand negatively regulates tumor growthrdquo Oncotarget vol 5 no20 pp 9744ndash9755 2014

[81] S Rossi P L Poliani CMissale EMonti andA Fanzani ldquoCav-eolins in rhabdomyosarcomardquo Journal of Cellular andMolecularMedicine vol 15 no 12 pp 2553ndash2568 2011

[82] P G Tsioli E S Patsouris C Giaginis and S E TheocharisldquoDNA repair systems in rhabdomyosarcomardquo Histology andHistopathology vol 28 no 8 pp 971ndash984 2013

[83] E Gatzidou C Michailidi S Tseleni-Balafouta and S Theo-charis ldquoAn epitome of DNA repair related genes and mecha-nisms in thyroid carcinomardquo Cancer Letters vol 290 no 2 pp139ndash147 2010

[84] R Rota R Ciarapica A Giordano L Miele and F LocatellildquoMicroRNAs in rhabdomyosarcoma pathogenetic implicationsand translational potentialityrdquoMolecular Cancer vol 10 article120 2011

[85] A Gougelet J Perez D Pissaloux et al ldquoMiRNA profiling howto bypass the current difficulties in the diagnosis and treatmentof sarcomasrdquo Sarcoma vol 2011 Article ID 460650 13 pages2011

[86] R Ciarapica G Russo F Verginelli et al ldquoDeregulated expres-sion of miR-26a and Ezh2 in rhabdomyosarcomardquo Cell Cyclevol 8 no 1 pp 172ndash175 2009

[87] L Li A L Sarver S Alamgir and S Subramanian ldquoDownreg-ulation of microRNAs miR-1 -206 and -29 stabilizes PAX3 andCCND2 expression in rhabdomyosarcomardquo Laboratory Investi-gation vol 92 no 4 pp 571ndash583 2012

[88] S-Y Ng G K Bogu B Soh and L W Stanton ldquoThe longnoncoding RNA RMST interacts with SOX2 to regulate neu-rogenesisrdquoMolecular Cell vol 51 no 3 pp 349ndash359 2013

[89] C A Lynch B Tycko T H Bestor and C P Walsh ldquoReac-tivation of a silenced H19 gene in human rhabdomyosarcomaby demethylation of DNA but not by histone hyperacetylationrdquoMolecular Cancer vol 1 article 2 2002

[90] P Onyango and A P Feinberg ldquoA nucleolar protein H19opposite tumor suppressor (HOTS) is a tumor growth inhibitorencoded by a human imprinted H19 antisense transcriptrdquoProceedings of the National Academy of Sciences of the UnitedStates of America vol 108 no 40 pp 16759ndash16764 2011

[91] C E Albacker N Y Storer E M Langdon et al ldquoThe his-tone methyltransferase SUV39H1 suppresses embryonal rhab-domyosarcoma formation in zebrafishrdquo PLoS ONE vol 8 no 5Article ID e64969 2013

[92] N Y Storer R M White A Uong et al ldquoZebrafish rhab-domyosarcoma reflects the developmental stage of oncogeneexpression during myogenesisrdquo Development vol 140 no 14pp 3040ndash3050 2013

[93] M E Hatley W Tang M R Garcia et al ldquoA mouse modelof rhabdomyosarcoma originating from the adipocyte lineagerdquoCancer Cell vol 22 no 4 pp 536ndash546 2012

[94] V Hosur A Kavirayani J Riefler et al ldquoDystrophin and dysfer-lin doublemutantmice a novelmodel for rhabdomyosarcomardquoCancer Genetics vol 205 no 5 pp 232ndash241 2012

[95] W Goessling T E North and L I Zon ldquoNew waves of discov-ery modeling cancer in zebrafishrdquo Journal of Clinical Oncologyvol 25 no 17 pp 2473ndash2479 2007

[96] B P Rubin K Nishijo H-I H Chen et al ldquoEvidence for anunanticipated relationship between undifferentiated pleomor-phic sarcoma and embryonal rhabdomyosarcomardquo Cancer Cellvol 19 no 2 pp 177ndash191 2011

[97] S Z Kaylani J Xu R K Srivastava L Kopelovich J G Presseyand M Athar ldquoRapamycin targeting mTOR and hedgehog sig-naling pathways blocks human rhabdomyosarcoma growth inxenograft murine modelrdquo Biochemical and Biophysical ResearchCommunications vol 435 no 4 pp 557ndash561 2013

[98] S Q Li A T Cheuk J F Shern et al ldquoTargeting wild-type andmutationally activated FGFR4 in rhabdomyosarcoma with theinhibitor ponatinib (AP24534)rdquoPLoSONE vol 8 no 10 ArticleID e76551 2013

[99] K Miyoshi K Kohashi F Fushimi et al ldquoClose correlationbetween CXCR4 and VEGF expression and frequent CXCR7expression in rhabdomyosarcomardquo Human Pathology vol 45no 9 pp 1900ndash1909 2014

[100] ADMarshall F Picchione R I KGeltink andGCGrosveldldquoPAX3-FOXO1 induces up-regulation of Noxa sensitizing alve-olar rhabdomyosarcoma cells to apoptosisrdquo Neoplasia vol 15no 7 pp 738ndash748 2013

[101] F Perut S Avnet C Fotia et al ldquoV-ATPase as an effective thera-peutic target for sarcomasrdquo Experimental Cell Research vol 320no 1 pp 21ndash32 2014

Submit your manuscripts athttpwwwhindawicom

Stem CellsInternational

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

MEDIATORSINFLAMMATION

of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Behavioural Neurology

EndocrinologyInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Disease Markers

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

BioMed Research International

OncologyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Oxidative Medicine and Cellular Longevity

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

PPAR Research

The Scientific World JournalHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Immunology ResearchHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal of

ObesityJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Computational and Mathematical Methods in Medicine

OphthalmologyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Diabetes ResearchJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Research and TreatmentAIDS

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Gastroenterology Research and Practice

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Parkinsonrsquos Disease

Evidence-Based Complementary and Alternative Medicine

Volume 2014Hindawi Publishing Corporationhttpwwwhindawicom

Page 11: Review Article Rhabdomyosarcoma: Advances in Molecular and

Sarcoma 11

tumorigenesis mechanisms There is a link between geneticand epigenetic alterations responsible for a variety of tumorcell growth proliferation differentiation apoptosis andtherapy-resistance mechanisms The identification of theprognostic value of the PAX-FKHR fusion status in RMSis one of the most important shifts in the lineage and therisk assessment of RMS which exerts an oncogenic effectthrough multiple pathways and is incorporated into mostrelevant studies The misbalanced expression of a number ofmiRs involved in the regulation of myogenic differentiationalso implicates the tumorsrsquo escape suppressive mechanismsMoreover we described several in vivo models of RMSto explore underlying histogenesis and different aspects oftumorigenesis Recent work on CSCs has also contributedto understanding the cell origins refining molecular char-acterization and elucidating the underlying basis of refrac-tory responses to current adjuvant treatments of subsets oftumorsThese integrative approaches can provide opportuni-ties to identify diagnostic and prognostic biomarkers appliedto the individual targeted therapy and significantly improveRMS prognosis

Abbreviations

RMS RhabdomyosarcomaARMS Alveolar rhabdomyosarcomaERMS Embryonal rhabdomyosarcomaPRMS Pleomorphic rhabdomyosarcomaSRMS Sclerosingspindle cell

rhabdomyosarcomamiRNA miR MicroRNACSC Cancer stem cellMSC Mesenchymal stem cell

Conflict of Interests

The authors declare that there is no conflict of interestsregarding the publication of this paper

Acknowledgments

This work was supported in part by grants from the Gat-tegno and Wechsler funds Dr Duan was supported in partthrough a grant from Sarcoma Foundation of America (SFA)a grant from National Cancer Institute (NCI)National Insti-tutes of Health (NIH) UO1 CA 151452 and a grant from anAcademic Enrichment Fund of MGH Orthopedic Surgery

References

[1] L Yang T Takimoto and J Fujimoto ldquoPrognostic model forpredicting overall survival in children and adolescents withrhabdomyosarcomardquo BMC Cancer vol 14 no 1 article 6542014

[2] D Egas-Bejar and W W Huh ldquoRhabdomyosarcoma in adoles-cent and young adult patients current perspectivesrdquo AdolescentHealth Medicine andTherapeutics vol 5 pp 115ndash125 2014

[3] J C Breneman E Lyden A S Pappo et al ldquoPrognosticfactors and clinical outcomes in children and adolescents withmetastatic rhabdomyosarcomamdasha report from the intergrouprhabdomyosarcoma study IVrdquo Journal of Clinical Oncology vol21 no 1 pp 78ndash84 2003

[4] V D Soleimani and M A Rudnicki ldquoNew insights into theorigin and the genetic basis of rhabdomyosarcomasrdquo CancerCell vol 19 no 2 pp 157ndash159 2011

[5] T Glass D D Cochrane S R Rassekh K Goddard and JHukin ldquoGrowing teratoma syndrome in intracranial non-ger-minomatous germ cell tumors (iNGGCTs) a risk for secondarymalignant transformationmdasha report of two casesrdquo Childrsquos Ner-vous System vol 30 no 5 pp 953ndash957 2014

[6] J Ji C Eng and K Hemminki ldquoFamilial risk for soft tissuetumors a nation-wide epidemiological study from SwedenrdquoJournal of Cancer Research and Clinical Oncology vol 134 no5 pp 617ndash624 2008

[7] C D M Fletcher J A Bridge P Hogendoorn and F MertensWHO Classification of Tumours of Soft Tissue and Bone vol 5IARC Press Paris France 4th edition 2013

[8] I Sultan I Qaddoumi S Yaser C Rodriguez-Galindo and AFerrari ldquoComparing adult and pediatric rhabdomyosarcoma inthe surveillance epidemiology and end results program 1973 to2005 an analysis of 2600 patientsrdquo Journal of Clinical Oncologyvol 27 no 20 pp 3391ndash3397 2009

[9] B Raney W Huh D Hawkins et al ldquoOutcome of patientswith localized orbital sarcoma who relapsed following treat-ment on Intergroup Rhabdomyosarcoma Study Group (IRSG)Protocols-III and -IV 1984ndash1997 a report from the ChildrenrsquosOncology Grouprdquo Pediatric Blood and Cancer vol 60 no 3 pp371ndash376 2013

[10] W Crist E A Gehan A H Ragab et al ldquoThe third intergrouprhabdomyosarcoma studyrdquo Journal of Clinical Oncology vol 13no 3 pp 610ndash630 1995

[11] C Liu D Li J Jiang et al ldquoAnalysis of molecular cytogeneticalteration in rhabdomyosarcoma by array comparative genomichybridizationrdquo PLoS ONE vol 9 no 4 Article ID e94924 2014

[12] DWilliamson E Missiaglia A de Reynies et al ldquoFusion gene-negative alveolar rhabdomyosarcoma is clinically and molec-ularly indistinguishable from embryonal rhabdomyosarcomardquoJournal of Clinical Oncology vol 28 no 13 pp 2151ndash2158 2010

[13] V Paulson G Chandler D Rakheja et al ldquoHigh-resolutionarray CGH identifies common mechanisms that drive embry-onal rhabdomyosarcoma pathogenesisrdquo Genes Chromosomesand Cancer vol 50 no 6 pp 397ndash408 2011

[14] S Park J Lee I-G Do et al ldquoAberrant CDK4 amplificationin refractory rhabdomyosarcoma as identified by genomicprofilingrdquo Scientific Reports vol 4 article 3623 2014

[15] C Liu D Li J Hu et al ldquoChromosomal and genetic imbalancesin Chinese patients with rhabdomyosarcoma detected by high-resolution array comparative genomic hybridizationrdquo Interna-tional Journal of Clinical and Experimental Pathology vol 7 no2 pp 690ndash698 2014

[16] Q-X Li C-X Liu C-P Chun et al ldquoChromosomal imbalancesrevealed in primary rhabdomyo-sarcomas by comparativegenomic hybridizationrdquo Chinese Medical Journal vol 122 no11 pp 1277ndash1282 2009

[17] E Missiaglia J Selfe M Hamdi et al ldquoGenomic imbalancesin rhabdomyosarcoma cell lines affect expression of genesfrequently altered in primary tumors an approach to identifycandidate genes involved in tumor developmentrdquo Genes Chro-mosomes and Cancer vol 48 no 6 pp 455ndash467 2009

12 Sarcoma

[18] F G Barr F Duan L M Smith et al ldquoGenomic and clinicalanalyses of 2p24 and 12q13-q14 amplification in alveolar rhab-domyosarcoma a report from the childrenrsquos oncology grouprdquoGenes Chromosomes and Cancer vol 48 no 8 pp 661ndash6722009

[19] T Gordon AMcManus J Anderson et al ldquoCytogenetic abnor-malities in 42 rhabdomyosarcoma a United Kingdom CancerCytogenetics Group StudyrdquoMedical and Pediatric Oncology vol36 no 2 pp 259ndash267 2001

[20] X Chen E Stewart A A Shelat et al ldquoTargeting oxidativestress in embryonal rhabdomyosarcomardquo Cancer Cell vol 24no 6 pp 710ndash724 2013

[21] S Lizard-Nacol F Mugneret C Volk C Turc-Carel M Favrotand T Philip ldquoTranslocation (213)(q37q14) in alveolar rhab-domyosarcoma a new caserdquo Cancer Genetics and Cytogeneticsvol 25 no 2 pp 373ndash374 1987

[22] P H B Sorensen J C Lynch S J Qualman et al ldquoPAX3-FKHR and PAX7-FKHR gene fusions are prognostic indicatorsin alveolar rhabdomyosarcoma a report from the childrenrsquosoncology grouprdquo Journal of Clinical Oncology vol 20 no 11 pp2672ndash2679 2002

[23] A D Marshall and G C Grosveld ldquoAlveolar rhabdomyo-sarcomamdashthe molecular drivers of PAX37-FOXO1-inducedtumorigenesisrdquo Skeletal Muscle vol 2 no 1 article 25 2012

[24] E Missiaglia D Williamson J Chisholm et al ldquoPAX3FOXO1fusion gene status is the key prognostic molecular markerin rhabdomyosarcoma and significantly improves current riskstratificationrdquo Journal of Clinical Oncology vol 30 no 14 pp1670ndash1677 2012

[25] J F Shern L Chen J Chmielecki et al ldquoComprehensivegenomic analysis of rhabdomyosarcoma reveals a landscape ofalterations affecting a common genetic axis in fusion-positiveand fusion-negative tumorsrdquo Cancer Discovery vol 4 no 2 pp216ndash231 2014

[26] MWachtel M Dettling E Koscielniak et al ldquoGene expressionsignatures identify rhabdomyosarcoma subtypes and detect anovel t(22)(q35p23) translocation fusing PAX3 to NCOA1rdquoCancer Research vol 64 no 16 pp 5539ndash5545 2004

[27] E Davicioni M J Anderson F G Finckenstein et al ldquoMolec-ular classification of rhabdomyosarcomamdashgenotypic and phe-notypic determinants of diagnosis a report from the ChildrenrsquosOncology Grouprdquo The American Journal of Pathology vol 174no 2 pp 550ndash564 2009

[28] M Visser C Sijmons J Bras et al ldquoAllelotype of pediatric rhab-domyosarcomardquo Oncogene vol 15 no 11 pp 1309ndash1314 1997

[29] R Gil-Benso T San-Miguel R C Callaghan et al ldquoChromo-somal and genetic changes produced in tumoral progression ofembryonal rhabdomyosarcomardquo Histopathology vol 62 no 5pp 816ndash819 2013

[30] S X Skapek J Anderson F G Barr et al ldquoPAX-FOXO1 fusionstatus drives unfavorable outcome for children with rhabdom-yosarcoma a childrenrsquos oncology group reportrdquo Pediatric Bloodand Cancer vol 60 no 9 pp 1411ndash1417 2013

[31] HHosoi N Kakazu E Konishi et al ldquoA novel PAX3 rearrange-ment in embryonal rhabdomyosarcomardquo Cancer Genetics andCytogenetics vol 189 no 2 pp 98ndash104 2009

[32] E Takaoka H Sonobe K Akimaru et al ldquoMultiple sites ofhighly amplified DNA sequences detected bymolecular cytoge-netic analysis inHS-RMS-2 a newpleomorphic rhabdomyosar-coma cell linerdquo American Journal of Cancer Research vol 2 no2 pp 141ndash152 2012

[33] K Szuhai D de Jong W Y Leung C D M Fletcher and P CWHogendoorn ldquoTransactivatingmutation of theMYOD1 geneis a frequent event in adult spindle cell rhabdomyosarcomardquoJournal of Pathology vol 232 no 3 pp 300ndash307 2014

[34] N P Agaram C-L Chen L Zhang M P Laquaglia L Wexlerand C R Antonescu ldquoRecurrent MYOD1 mutations in pedi-atric and adult sclerosing and spindle cell rhabdomyosarcomasEvidence for a common pathogenesisrdquoGenes Chromosomes andCancer vol 53 no 9 pp 779ndash787 2014

[35] H Yoshida M Miyachi K Sakamoto et al ldquoPAX3-NCOA2fusion gene has a dual role in promoting the proliferation andinhibiting the myogenic differentiation of rhabdomyosarcomacellsrdquo Oncogene vol 33 pp 5601ndash5608 2013

[36] J M Mosquera A Sboner L Zhang et al ldquoRecurrent NCOA2gene rearrangements in congenitalinfantile spindle cell rhab-domyosarcomardquo Genes Chromosomes and Cancer vol 52 no6 pp 538ndash550 2013

[37] V Thavaraj A Sethi and L S Arya ldquoIncomplete Beckwith-Wiedemann syndrome in a child with orbital rhabdomyosar-comardquo Indian Pediatrics vol 39 no 3 pp 299ndash304 2002

[38] D Walter S Satheesha P Albrecht et al ldquoCD133 positiveembryonal rhabdomyosarcoma stem-like cell population isenriched in rhabdospheresrdquo PLoS ONE vol 6 no 5 Article IDe19506 2011

[39] Y-X Ren F G Finckenstein D A Abdueva et al ldquoMousemesenchymal stem cells expressing PAX-FKHR form alveolarrhabdomyosarcomas by cooperating with secondary muta-tionsrdquo Cancer Research vol 68 no 16 pp 6587ndash6597 2008

[40] M Salerno S Avnet G Bonuccelli S Hosogi D Granchi andN Baldini ldquoImpairment of lysosomal activity as a therapeuticmodality targeting cancer stem cells of embryonal rhabdom-yosarcoma cell line RDrdquo PLoS ONE vol 9 no 10 Article IDe110340 2014

[41] K Tanoue Y Wang M Ikeda et al ldquoSurvivin-responsive con-ditionally replicating adenovirus kills rhabdomyosarcoma stemcells more efficiently than their progenyrdquo Journal of Transla-tional Medicine vol 12 no 1 article 27 2014

[42] P K Rao EMissiaglia L Shields et al ldquoDistinct roles formiR-1and miR-133a in the proliferation and differentiation of rhab-domyosarcoma cellsrdquo The FASEB Journal vol 24 no 9 pp3427ndash3437 2010

[43] D Yan X D Dong X Chen et al ldquoMicroRNA-1206 targets c-met and inhibits rhabdomyosarcomadevelopmentrdquoThe Journalof Biological Chemistry vol 284 no 43 pp 29596ndash29604 2009

[44] I Marchesi A Giordano and L Bagella ldquoRoles of enhancerof zeste homolog 2 from skeletal muscle differentiation torhabdomyosarcoma carcinogenesisrdquoCell Cycle vol 13 no 4 pp516ndash527 2014

[45] M M Sun J F Li L L Guo et al ldquoTGF-1205731 suppression ofmicroRNA-450b-5p expression a novel mechanism for block-ingmyogenic differentiation of rhabdomyosarcomardquoOncogenevol 33 no 16 pp 2075ndash2086 2014

[46] Y Diao X Guo L Jiang et al ldquoMiR-203 a tumor suppressorfrequently down-regulated by promoter hypermethylation inrhabdomyosarcomardquo The Journal of Biological Chemistry vol289 no 1 pp 529ndash539 2014

[47] H-J Huang J Liu HHua et al ldquoMiR-214 andN-ras regulatoryloop suppresses rhabdomyosarcoma cell growth and xenografttumorigenesisrdquo Oncotarget vol 5 no 8 pp 2161ndash2175 2014

Sarcoma 13

[48] A L Sarver L Li and S Subramanian ldquomicroRNA miR-183functions as an oncogene by targeting the transcription factorEGR1 and promoting tumor cell migrationrdquo Cancer Researchvol 70 no 23 pp 9570ndash9580 2010

[49] R L Galindo J A Allport and E N Olson ldquoA Drosophilamodel of the rhabdomyosarcoma initiator PAX7-FKHRrdquo Pro-ceedings of the National Academy of Sciences of the United Statesof America vol 103 no 36 pp 13439ndash13444 2006

[50] U Avirneni-Vadlamudi K A Galindo T R Endicott V Paul-son S Cameron and R L Galindo ldquoDrosophila and mammal-ian models uncover a role for the myoblast fusion gene TANC1in rhabdomyosarcomardquo The Journal of Clinical Investigationvol 122 no 1 pp 403ndash407 2012

[51] E Y Chen andDM Langenau ldquoZebrafishmodels of rhabdom-yosarcomardquoMethods in Cell Biology vol 105 pp 383ndash402 2011

[52] X Le E K Pugach S Hettmer et al ldquoA novel chemicalscreening strategy in zebrafish identifies common pathways inembryogenesis and rhabdomyosarcoma developmentrdquo Devel-opment vol 140 no 11 pp 2354ndash2364 2012

[53] Y Lee R Kawagoe K Sasai et al ldquoLoss of suppressor-of-fusedfunction promotes tumorigenesisrdquoOncogene vol 26 no 44 pp6442ndash6447 2007

[54] C Keller B R Arenkiel C M Coffin N El-Bardeesy R ADePinho and M R Capecchi ldquoAlveolar rhabdomyosarcomasin conditional Pax3Fkhr mice cooperativity of Ink4aARF andTrp53 loss of functionrdquo Genes and Development vol 18 no 21pp 2614ndash2626 2004

[55] S Singh CVinson CMGurley et al ldquoImpairedWnt signalingin embryonal rhabdomyosarcoma cells from p53c-fos doublemutant micerdquo American Journal of Pathology vol 177 no 4 pp2055ndash2066 2010

[56] G J Schaaf J M Ruijter F Van Ruissen et al ldquoFull transcrip-tome analysis of rhabdomyosarcoma normal and fetal skeletalmuscle statistical comparison of multiple SAGE librariesrdquo TheFASEB Journal vol 19 no 3 pp 404ndash406 2005

[57] D S Hawkins A A Gupta and E R Rudzinski ldquoWhat is newin the biology and treatment of pediatric rhabdomyosarcomardquoCurrent Opinion in Pediatrics vol 26 no 1 pp 50ndash56 2014

[58] M Zhang J Truscott and J Davie ldquoLoss of MEF2D expressioninhibits differentiation and contributes to oncogenesis in rhab-domyosarcoma cellsrdquoMolecular Cancer vol 12 no 1 article 1502013

[59] M Ciesla J Dulak and A Jozkowicz ldquomicroRNAs and epi-genetic mechanisms of rhabdomyosarcoma developmentrdquo TheInternational Journal of Biochemistry amp Cell Biology vol 53 pp482ndash492 2014

[60] S Ricaud B Vernus M Duclos et al ldquoInhibition of autocrinesecretion of myostatin enhances terminal differentiation inhuman rhabdomyosarcoma cellsrdquo Oncogene vol 22 no 51 pp8221ndash8232 2003

[61] C S Guo C Degnin T A Fiddler D Stauffer andM JThayerldquoRegulation of MyoD activity and muscle cell differentiation byMDM2 pRb and Sp1rdquoThe Journal of Biological Chemistry vol278 no 25 pp 22615ndash22622 2003

[62] S Rossi E Stoppani P L Puri and A Fanzani ldquoDifferenti-ation of human rhabdomyosarcoma RD cells is regulated byreciprocal functional interactions between myostatin p38 andextracellular regulated kinase signalling pathwaysrdquo EuropeanJournal of Cancer vol 47 no 7 pp 1095ndash1105 2011

[63] Z S Walters B Villarejo-Balcells D Olmos et al ldquoJARID2is a direct target of the PAX3-FOXO1 fusion protein andinhibits myogenic differentiation of rhabdomyosarcoma cellsrdquoOncogene vol 33 no 9 pp 1148ndash1157 2014

[64] T Hosoyama M I Aslam J Abraham et al ldquoIL-4R drives ded-ifferentiation mitogenesis and metastasis in rhabdomyosar-comardquo Clinical Cancer Research vol 17 no 9 pp 2757ndash27662011

[65] F S Dela Cruz ldquoCancer stem cells in pediatric sarcomasrdquoFrontiers in Oncology vol 3 article 168 2013

[66] M J Bissell and W C Hines ldquoWhy donrsquot we get more cancerA proposed role of the microenvironment in restraining cancerprogressionrdquo Nature Medicine vol 17 no 3 pp 320ndash329 2011

[67] E Charytonowicz C Cordon-Cardo I Matushansky and MZiman ldquoAlveolar rhabdomyosarcoma is the cell of origin amesenchymal stem cellrdquoCancer Letters vol 279 no 2 pp 126ndash136 2009

[68] W K Cavenee ldquoMuscling in on rhabdomyosarcomardquo NatureMedicine vol 8 no 11 pp 1200ndash1201 2002

[69] B Zhu and J K Davie ldquoNew insights into signalling-pathwayalterations in rhabdomyosarcomardquo British Journal of Cancervol 112 no 2 pp 227ndash231 2015

[70] M Zhang C M Linardic and D G Kirsch ldquoRAS and ROS inrhabdomyosarcomardquo Cancer Cell vol 24 no 6 pp 689ndash6912013

[71] S C Marianna L Ianzano and G Nicoletti ldquoTumor sup-pressor genes promote rhabdomyosarcoma progression in p53heterozygousrdquo Oncotarget vol 5 no 1 pp 108ndash119 2013

[72] SWang L Guo L Dong et al ldquoTGF-beta1 signal pathwaymaycontribute to rhabdomyosarcoma development by inhibitingdifferentiationrdquo Cancer Science vol 101 no 5 pp 1108ndash11162010

[73] J Wesche K Haglund and E M Haugsten ldquoFibroblast growthfactors and their receptors in cancerrdquo Biochemical Journal vol437 no 2 pp 199ndash213 2011

[74] M Wachtel J Rakic M Okoniewski P Bode F Niggli andB W Schafer ldquoFGFR4 signaling couples to Bim and not Bmfto discriminate subsets of alveolar rhabdomyosarcoma cellsrdquoInternational Journal of Cancer vol 135 no 7 pp 1543ndash15522014

[75] P L Puri ZWu P Zhang et al ldquoInduction of terminal differen-tiation by constitutive activation of p38 MAP kinase in humanrhabdomyosarcoma cellsrdquo Genes and Development vol 14 no5 pp 574ndash584 2000

[76] A M Tremblay E Missiaglia G G Galli et al ldquoThe Hippotransducer YAP1 transforms activated satellite cells and is apotent effector of embryonal rhabdomyosarcoma formationrdquoCancer Cell vol 26 no 2 pp 273ndash287 2014

[77] R N Judson A M Tremblay P Knopp et al ldquoThe hippo path-way member Yap plays a key role in influencing fate decisionsin muscle satellite cellsrdquo Journal of Cell Science vol 125 no 24pp 6009ndash6019 2012

[78] L E S Crose K A Galindo J G Kephart et al ldquoAlve-olar rhabdomyosarcoma-associated PAX3-FOXO1 promotestumorigenesis via Hippo pathway suppressionrdquo Journal ofClinical Investigation vol 124 no 1 pp 285ndash296 2014

[79] B Belyea J G Kephart J Blum D G Kirsch and C MLinardic ldquoEmbryonic signaling pathways and rhabdomyosar-coma contributions to cancer development and opportunitiesfor therapeutic targetingrdquo Sarcoma vol 2012 Article ID 40623913 pages 2012

14 Sarcoma

[80] J Huertas-Martinez S Rello-Varona D Herrero-Martin et alldquoCaveolin-1 is down-regulated in alveolar rhabdomyosarcomasand negatively regulates tumor growthrdquo Oncotarget vol 5 no20 pp 9744ndash9755 2014

[81] S Rossi P L Poliani CMissale EMonti andA Fanzani ldquoCav-eolins in rhabdomyosarcomardquo Journal of Cellular andMolecularMedicine vol 15 no 12 pp 2553ndash2568 2011

[82] P G Tsioli E S Patsouris C Giaginis and S E TheocharisldquoDNA repair systems in rhabdomyosarcomardquo Histology andHistopathology vol 28 no 8 pp 971ndash984 2013

[83] E Gatzidou C Michailidi S Tseleni-Balafouta and S Theo-charis ldquoAn epitome of DNA repair related genes and mecha-nisms in thyroid carcinomardquo Cancer Letters vol 290 no 2 pp139ndash147 2010

[84] R Rota R Ciarapica A Giordano L Miele and F LocatellildquoMicroRNAs in rhabdomyosarcoma pathogenetic implicationsand translational potentialityrdquoMolecular Cancer vol 10 article120 2011

[85] A Gougelet J Perez D Pissaloux et al ldquoMiRNA profiling howto bypass the current difficulties in the diagnosis and treatmentof sarcomasrdquo Sarcoma vol 2011 Article ID 460650 13 pages2011

[86] R Ciarapica G Russo F Verginelli et al ldquoDeregulated expres-sion of miR-26a and Ezh2 in rhabdomyosarcomardquo Cell Cyclevol 8 no 1 pp 172ndash175 2009

[87] L Li A L Sarver S Alamgir and S Subramanian ldquoDownreg-ulation of microRNAs miR-1 -206 and -29 stabilizes PAX3 andCCND2 expression in rhabdomyosarcomardquo Laboratory Investi-gation vol 92 no 4 pp 571ndash583 2012

[88] S-Y Ng G K Bogu B Soh and L W Stanton ldquoThe longnoncoding RNA RMST interacts with SOX2 to regulate neu-rogenesisrdquoMolecular Cell vol 51 no 3 pp 349ndash359 2013

[89] C A Lynch B Tycko T H Bestor and C P Walsh ldquoReac-tivation of a silenced H19 gene in human rhabdomyosarcomaby demethylation of DNA but not by histone hyperacetylationrdquoMolecular Cancer vol 1 article 2 2002

[90] P Onyango and A P Feinberg ldquoA nucleolar protein H19opposite tumor suppressor (HOTS) is a tumor growth inhibitorencoded by a human imprinted H19 antisense transcriptrdquoProceedings of the National Academy of Sciences of the UnitedStates of America vol 108 no 40 pp 16759ndash16764 2011

[91] C E Albacker N Y Storer E M Langdon et al ldquoThe his-tone methyltransferase SUV39H1 suppresses embryonal rhab-domyosarcoma formation in zebrafishrdquo PLoS ONE vol 8 no 5Article ID e64969 2013

[92] N Y Storer R M White A Uong et al ldquoZebrafish rhab-domyosarcoma reflects the developmental stage of oncogeneexpression during myogenesisrdquo Development vol 140 no 14pp 3040ndash3050 2013

[93] M E Hatley W Tang M R Garcia et al ldquoA mouse modelof rhabdomyosarcoma originating from the adipocyte lineagerdquoCancer Cell vol 22 no 4 pp 536ndash546 2012

[94] V Hosur A Kavirayani J Riefler et al ldquoDystrophin and dysfer-lin doublemutantmice a novelmodel for rhabdomyosarcomardquoCancer Genetics vol 205 no 5 pp 232ndash241 2012

[95] W Goessling T E North and L I Zon ldquoNew waves of discov-ery modeling cancer in zebrafishrdquo Journal of Clinical Oncologyvol 25 no 17 pp 2473ndash2479 2007

[96] B P Rubin K Nishijo H-I H Chen et al ldquoEvidence for anunanticipated relationship between undifferentiated pleomor-phic sarcoma and embryonal rhabdomyosarcomardquo Cancer Cellvol 19 no 2 pp 177ndash191 2011

[97] S Z Kaylani J Xu R K Srivastava L Kopelovich J G Presseyand M Athar ldquoRapamycin targeting mTOR and hedgehog sig-naling pathways blocks human rhabdomyosarcoma growth inxenograft murine modelrdquo Biochemical and Biophysical ResearchCommunications vol 435 no 4 pp 557ndash561 2013

[98] S Q Li A T Cheuk J F Shern et al ldquoTargeting wild-type andmutationally activated FGFR4 in rhabdomyosarcoma with theinhibitor ponatinib (AP24534)rdquoPLoSONE vol 8 no 10 ArticleID e76551 2013

[99] K Miyoshi K Kohashi F Fushimi et al ldquoClose correlationbetween CXCR4 and VEGF expression and frequent CXCR7expression in rhabdomyosarcomardquo Human Pathology vol 45no 9 pp 1900ndash1909 2014

[100] ADMarshall F Picchione R I KGeltink andGCGrosveldldquoPAX3-FOXO1 induces up-regulation of Noxa sensitizing alve-olar rhabdomyosarcoma cells to apoptosisrdquo Neoplasia vol 15no 7 pp 738ndash748 2013

[101] F Perut S Avnet C Fotia et al ldquoV-ATPase as an effective thera-peutic target for sarcomasrdquo Experimental Cell Research vol 320no 1 pp 21ndash32 2014

Submit your manuscripts athttpwwwhindawicom

Stem CellsInternational

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

MEDIATORSINFLAMMATION

of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Behavioural Neurology

EndocrinologyInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Disease Markers

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

BioMed Research International

OncologyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Oxidative Medicine and Cellular Longevity

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

PPAR Research

The Scientific World JournalHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Immunology ResearchHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal of

ObesityJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Computational and Mathematical Methods in Medicine

OphthalmologyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Diabetes ResearchJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Research and TreatmentAIDS

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Gastroenterology Research and Practice

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Parkinsonrsquos Disease

Evidence-Based Complementary and Alternative Medicine

Volume 2014Hindawi Publishing Corporationhttpwwwhindawicom

Page 12: Review Article Rhabdomyosarcoma: Advances in Molecular and

12 Sarcoma

[18] F G Barr F Duan L M Smith et al ldquoGenomic and clinicalanalyses of 2p24 and 12q13-q14 amplification in alveolar rhab-domyosarcoma a report from the childrenrsquos oncology grouprdquoGenes Chromosomes and Cancer vol 48 no 8 pp 661ndash6722009

[19] T Gordon AMcManus J Anderson et al ldquoCytogenetic abnor-malities in 42 rhabdomyosarcoma a United Kingdom CancerCytogenetics Group StudyrdquoMedical and Pediatric Oncology vol36 no 2 pp 259ndash267 2001

[20] X Chen E Stewart A A Shelat et al ldquoTargeting oxidativestress in embryonal rhabdomyosarcomardquo Cancer Cell vol 24no 6 pp 710ndash724 2013

[21] S Lizard-Nacol F Mugneret C Volk C Turc-Carel M Favrotand T Philip ldquoTranslocation (213)(q37q14) in alveolar rhab-domyosarcoma a new caserdquo Cancer Genetics and Cytogeneticsvol 25 no 2 pp 373ndash374 1987

[22] P H B Sorensen J C Lynch S J Qualman et al ldquoPAX3-FKHR and PAX7-FKHR gene fusions are prognostic indicatorsin alveolar rhabdomyosarcoma a report from the childrenrsquosoncology grouprdquo Journal of Clinical Oncology vol 20 no 11 pp2672ndash2679 2002

[23] A D Marshall and G C Grosveld ldquoAlveolar rhabdomyo-sarcomamdashthe molecular drivers of PAX37-FOXO1-inducedtumorigenesisrdquo Skeletal Muscle vol 2 no 1 article 25 2012

[24] E Missiaglia D Williamson J Chisholm et al ldquoPAX3FOXO1fusion gene status is the key prognostic molecular markerin rhabdomyosarcoma and significantly improves current riskstratificationrdquo Journal of Clinical Oncology vol 30 no 14 pp1670ndash1677 2012

[25] J F Shern L Chen J Chmielecki et al ldquoComprehensivegenomic analysis of rhabdomyosarcoma reveals a landscape ofalterations affecting a common genetic axis in fusion-positiveand fusion-negative tumorsrdquo Cancer Discovery vol 4 no 2 pp216ndash231 2014

[26] MWachtel M Dettling E Koscielniak et al ldquoGene expressionsignatures identify rhabdomyosarcoma subtypes and detect anovel t(22)(q35p23) translocation fusing PAX3 to NCOA1rdquoCancer Research vol 64 no 16 pp 5539ndash5545 2004

[27] E Davicioni M J Anderson F G Finckenstein et al ldquoMolec-ular classification of rhabdomyosarcomamdashgenotypic and phe-notypic determinants of diagnosis a report from the ChildrenrsquosOncology Grouprdquo The American Journal of Pathology vol 174no 2 pp 550ndash564 2009

[28] M Visser C Sijmons J Bras et al ldquoAllelotype of pediatric rhab-domyosarcomardquo Oncogene vol 15 no 11 pp 1309ndash1314 1997

[29] R Gil-Benso T San-Miguel R C Callaghan et al ldquoChromo-somal and genetic changes produced in tumoral progression ofembryonal rhabdomyosarcomardquo Histopathology vol 62 no 5pp 816ndash819 2013

[30] S X Skapek J Anderson F G Barr et al ldquoPAX-FOXO1 fusionstatus drives unfavorable outcome for children with rhabdom-yosarcoma a childrenrsquos oncology group reportrdquo Pediatric Bloodand Cancer vol 60 no 9 pp 1411ndash1417 2013

[31] HHosoi N Kakazu E Konishi et al ldquoA novel PAX3 rearrange-ment in embryonal rhabdomyosarcomardquo Cancer Genetics andCytogenetics vol 189 no 2 pp 98ndash104 2009

[32] E Takaoka H Sonobe K Akimaru et al ldquoMultiple sites ofhighly amplified DNA sequences detected bymolecular cytoge-netic analysis inHS-RMS-2 a newpleomorphic rhabdomyosar-coma cell linerdquo American Journal of Cancer Research vol 2 no2 pp 141ndash152 2012

[33] K Szuhai D de Jong W Y Leung C D M Fletcher and P CWHogendoorn ldquoTransactivatingmutation of theMYOD1 geneis a frequent event in adult spindle cell rhabdomyosarcomardquoJournal of Pathology vol 232 no 3 pp 300ndash307 2014

[34] N P Agaram C-L Chen L Zhang M P Laquaglia L Wexlerand C R Antonescu ldquoRecurrent MYOD1 mutations in pedi-atric and adult sclerosing and spindle cell rhabdomyosarcomasEvidence for a common pathogenesisrdquoGenes Chromosomes andCancer vol 53 no 9 pp 779ndash787 2014

[35] H Yoshida M Miyachi K Sakamoto et al ldquoPAX3-NCOA2fusion gene has a dual role in promoting the proliferation andinhibiting the myogenic differentiation of rhabdomyosarcomacellsrdquo Oncogene vol 33 pp 5601ndash5608 2013

[36] J M Mosquera A Sboner L Zhang et al ldquoRecurrent NCOA2gene rearrangements in congenitalinfantile spindle cell rhab-domyosarcomardquo Genes Chromosomes and Cancer vol 52 no6 pp 538ndash550 2013

[37] V Thavaraj A Sethi and L S Arya ldquoIncomplete Beckwith-Wiedemann syndrome in a child with orbital rhabdomyosar-comardquo Indian Pediatrics vol 39 no 3 pp 299ndash304 2002

[38] D Walter S Satheesha P Albrecht et al ldquoCD133 positiveembryonal rhabdomyosarcoma stem-like cell population isenriched in rhabdospheresrdquo PLoS ONE vol 6 no 5 Article IDe19506 2011

[39] Y-X Ren F G Finckenstein D A Abdueva et al ldquoMousemesenchymal stem cells expressing PAX-FKHR form alveolarrhabdomyosarcomas by cooperating with secondary muta-tionsrdquo Cancer Research vol 68 no 16 pp 6587ndash6597 2008

[40] M Salerno S Avnet G Bonuccelli S Hosogi D Granchi andN Baldini ldquoImpairment of lysosomal activity as a therapeuticmodality targeting cancer stem cells of embryonal rhabdom-yosarcoma cell line RDrdquo PLoS ONE vol 9 no 10 Article IDe110340 2014

[41] K Tanoue Y Wang M Ikeda et al ldquoSurvivin-responsive con-ditionally replicating adenovirus kills rhabdomyosarcoma stemcells more efficiently than their progenyrdquo Journal of Transla-tional Medicine vol 12 no 1 article 27 2014

[42] P K Rao EMissiaglia L Shields et al ldquoDistinct roles formiR-1and miR-133a in the proliferation and differentiation of rhab-domyosarcoma cellsrdquo The FASEB Journal vol 24 no 9 pp3427ndash3437 2010

[43] D Yan X D Dong X Chen et al ldquoMicroRNA-1206 targets c-met and inhibits rhabdomyosarcomadevelopmentrdquoThe Journalof Biological Chemistry vol 284 no 43 pp 29596ndash29604 2009

[44] I Marchesi A Giordano and L Bagella ldquoRoles of enhancerof zeste homolog 2 from skeletal muscle differentiation torhabdomyosarcoma carcinogenesisrdquoCell Cycle vol 13 no 4 pp516ndash527 2014

[45] M M Sun J F Li L L Guo et al ldquoTGF-1205731 suppression ofmicroRNA-450b-5p expression a novel mechanism for block-ingmyogenic differentiation of rhabdomyosarcomardquoOncogenevol 33 no 16 pp 2075ndash2086 2014

[46] Y Diao X Guo L Jiang et al ldquoMiR-203 a tumor suppressorfrequently down-regulated by promoter hypermethylation inrhabdomyosarcomardquo The Journal of Biological Chemistry vol289 no 1 pp 529ndash539 2014

[47] H-J Huang J Liu HHua et al ldquoMiR-214 andN-ras regulatoryloop suppresses rhabdomyosarcoma cell growth and xenografttumorigenesisrdquo Oncotarget vol 5 no 8 pp 2161ndash2175 2014

Sarcoma 13

[48] A L Sarver L Li and S Subramanian ldquomicroRNA miR-183functions as an oncogene by targeting the transcription factorEGR1 and promoting tumor cell migrationrdquo Cancer Researchvol 70 no 23 pp 9570ndash9580 2010

[49] R L Galindo J A Allport and E N Olson ldquoA Drosophilamodel of the rhabdomyosarcoma initiator PAX7-FKHRrdquo Pro-ceedings of the National Academy of Sciences of the United Statesof America vol 103 no 36 pp 13439ndash13444 2006

[50] U Avirneni-Vadlamudi K A Galindo T R Endicott V Paul-son S Cameron and R L Galindo ldquoDrosophila and mammal-ian models uncover a role for the myoblast fusion gene TANC1in rhabdomyosarcomardquo The Journal of Clinical Investigationvol 122 no 1 pp 403ndash407 2012

[51] E Y Chen andDM Langenau ldquoZebrafishmodels of rhabdom-yosarcomardquoMethods in Cell Biology vol 105 pp 383ndash402 2011

[52] X Le E K Pugach S Hettmer et al ldquoA novel chemicalscreening strategy in zebrafish identifies common pathways inembryogenesis and rhabdomyosarcoma developmentrdquo Devel-opment vol 140 no 11 pp 2354ndash2364 2012

[53] Y Lee R Kawagoe K Sasai et al ldquoLoss of suppressor-of-fusedfunction promotes tumorigenesisrdquoOncogene vol 26 no 44 pp6442ndash6447 2007

[54] C Keller B R Arenkiel C M Coffin N El-Bardeesy R ADePinho and M R Capecchi ldquoAlveolar rhabdomyosarcomasin conditional Pax3Fkhr mice cooperativity of Ink4aARF andTrp53 loss of functionrdquo Genes and Development vol 18 no 21pp 2614ndash2626 2004

[55] S Singh CVinson CMGurley et al ldquoImpairedWnt signalingin embryonal rhabdomyosarcoma cells from p53c-fos doublemutant micerdquo American Journal of Pathology vol 177 no 4 pp2055ndash2066 2010

[56] G J Schaaf J M Ruijter F Van Ruissen et al ldquoFull transcrip-tome analysis of rhabdomyosarcoma normal and fetal skeletalmuscle statistical comparison of multiple SAGE librariesrdquo TheFASEB Journal vol 19 no 3 pp 404ndash406 2005

[57] D S Hawkins A A Gupta and E R Rudzinski ldquoWhat is newin the biology and treatment of pediatric rhabdomyosarcomardquoCurrent Opinion in Pediatrics vol 26 no 1 pp 50ndash56 2014

[58] M Zhang J Truscott and J Davie ldquoLoss of MEF2D expressioninhibits differentiation and contributes to oncogenesis in rhab-domyosarcoma cellsrdquoMolecular Cancer vol 12 no 1 article 1502013

[59] M Ciesla J Dulak and A Jozkowicz ldquomicroRNAs and epi-genetic mechanisms of rhabdomyosarcoma developmentrdquo TheInternational Journal of Biochemistry amp Cell Biology vol 53 pp482ndash492 2014

[60] S Ricaud B Vernus M Duclos et al ldquoInhibition of autocrinesecretion of myostatin enhances terminal differentiation inhuman rhabdomyosarcoma cellsrdquo Oncogene vol 22 no 51 pp8221ndash8232 2003

[61] C S Guo C Degnin T A Fiddler D Stauffer andM JThayerldquoRegulation of MyoD activity and muscle cell differentiation byMDM2 pRb and Sp1rdquoThe Journal of Biological Chemistry vol278 no 25 pp 22615ndash22622 2003

[62] S Rossi E Stoppani P L Puri and A Fanzani ldquoDifferenti-ation of human rhabdomyosarcoma RD cells is regulated byreciprocal functional interactions between myostatin p38 andextracellular regulated kinase signalling pathwaysrdquo EuropeanJournal of Cancer vol 47 no 7 pp 1095ndash1105 2011

[63] Z S Walters B Villarejo-Balcells D Olmos et al ldquoJARID2is a direct target of the PAX3-FOXO1 fusion protein andinhibits myogenic differentiation of rhabdomyosarcoma cellsrdquoOncogene vol 33 no 9 pp 1148ndash1157 2014

[64] T Hosoyama M I Aslam J Abraham et al ldquoIL-4R drives ded-ifferentiation mitogenesis and metastasis in rhabdomyosar-comardquo Clinical Cancer Research vol 17 no 9 pp 2757ndash27662011

[65] F S Dela Cruz ldquoCancer stem cells in pediatric sarcomasrdquoFrontiers in Oncology vol 3 article 168 2013

[66] M J Bissell and W C Hines ldquoWhy donrsquot we get more cancerA proposed role of the microenvironment in restraining cancerprogressionrdquo Nature Medicine vol 17 no 3 pp 320ndash329 2011

[67] E Charytonowicz C Cordon-Cardo I Matushansky and MZiman ldquoAlveolar rhabdomyosarcoma is the cell of origin amesenchymal stem cellrdquoCancer Letters vol 279 no 2 pp 126ndash136 2009

[68] W K Cavenee ldquoMuscling in on rhabdomyosarcomardquo NatureMedicine vol 8 no 11 pp 1200ndash1201 2002

[69] B Zhu and J K Davie ldquoNew insights into signalling-pathwayalterations in rhabdomyosarcomardquo British Journal of Cancervol 112 no 2 pp 227ndash231 2015

[70] M Zhang C M Linardic and D G Kirsch ldquoRAS and ROS inrhabdomyosarcomardquo Cancer Cell vol 24 no 6 pp 689ndash6912013

[71] S C Marianna L Ianzano and G Nicoletti ldquoTumor sup-pressor genes promote rhabdomyosarcoma progression in p53heterozygousrdquo Oncotarget vol 5 no 1 pp 108ndash119 2013

[72] SWang L Guo L Dong et al ldquoTGF-beta1 signal pathwaymaycontribute to rhabdomyosarcoma development by inhibitingdifferentiationrdquo Cancer Science vol 101 no 5 pp 1108ndash11162010

[73] J Wesche K Haglund and E M Haugsten ldquoFibroblast growthfactors and their receptors in cancerrdquo Biochemical Journal vol437 no 2 pp 199ndash213 2011

[74] M Wachtel J Rakic M Okoniewski P Bode F Niggli andB W Schafer ldquoFGFR4 signaling couples to Bim and not Bmfto discriminate subsets of alveolar rhabdomyosarcoma cellsrdquoInternational Journal of Cancer vol 135 no 7 pp 1543ndash15522014

[75] P L Puri ZWu P Zhang et al ldquoInduction of terminal differen-tiation by constitutive activation of p38 MAP kinase in humanrhabdomyosarcoma cellsrdquo Genes and Development vol 14 no5 pp 574ndash584 2000

[76] A M Tremblay E Missiaglia G G Galli et al ldquoThe Hippotransducer YAP1 transforms activated satellite cells and is apotent effector of embryonal rhabdomyosarcoma formationrdquoCancer Cell vol 26 no 2 pp 273ndash287 2014

[77] R N Judson A M Tremblay P Knopp et al ldquoThe hippo path-way member Yap plays a key role in influencing fate decisionsin muscle satellite cellsrdquo Journal of Cell Science vol 125 no 24pp 6009ndash6019 2012

[78] L E S Crose K A Galindo J G Kephart et al ldquoAlve-olar rhabdomyosarcoma-associated PAX3-FOXO1 promotestumorigenesis via Hippo pathway suppressionrdquo Journal ofClinical Investigation vol 124 no 1 pp 285ndash296 2014

[79] B Belyea J G Kephart J Blum D G Kirsch and C MLinardic ldquoEmbryonic signaling pathways and rhabdomyosar-coma contributions to cancer development and opportunitiesfor therapeutic targetingrdquo Sarcoma vol 2012 Article ID 40623913 pages 2012

14 Sarcoma

[80] J Huertas-Martinez S Rello-Varona D Herrero-Martin et alldquoCaveolin-1 is down-regulated in alveolar rhabdomyosarcomasand negatively regulates tumor growthrdquo Oncotarget vol 5 no20 pp 9744ndash9755 2014

[81] S Rossi P L Poliani CMissale EMonti andA Fanzani ldquoCav-eolins in rhabdomyosarcomardquo Journal of Cellular andMolecularMedicine vol 15 no 12 pp 2553ndash2568 2011

[82] P G Tsioli E S Patsouris C Giaginis and S E TheocharisldquoDNA repair systems in rhabdomyosarcomardquo Histology andHistopathology vol 28 no 8 pp 971ndash984 2013

[83] E Gatzidou C Michailidi S Tseleni-Balafouta and S Theo-charis ldquoAn epitome of DNA repair related genes and mecha-nisms in thyroid carcinomardquo Cancer Letters vol 290 no 2 pp139ndash147 2010

[84] R Rota R Ciarapica A Giordano L Miele and F LocatellildquoMicroRNAs in rhabdomyosarcoma pathogenetic implicationsand translational potentialityrdquoMolecular Cancer vol 10 article120 2011

[85] A Gougelet J Perez D Pissaloux et al ldquoMiRNA profiling howto bypass the current difficulties in the diagnosis and treatmentof sarcomasrdquo Sarcoma vol 2011 Article ID 460650 13 pages2011

[86] R Ciarapica G Russo F Verginelli et al ldquoDeregulated expres-sion of miR-26a and Ezh2 in rhabdomyosarcomardquo Cell Cyclevol 8 no 1 pp 172ndash175 2009

[87] L Li A L Sarver S Alamgir and S Subramanian ldquoDownreg-ulation of microRNAs miR-1 -206 and -29 stabilizes PAX3 andCCND2 expression in rhabdomyosarcomardquo Laboratory Investi-gation vol 92 no 4 pp 571ndash583 2012

[88] S-Y Ng G K Bogu B Soh and L W Stanton ldquoThe longnoncoding RNA RMST interacts with SOX2 to regulate neu-rogenesisrdquoMolecular Cell vol 51 no 3 pp 349ndash359 2013

[89] C A Lynch B Tycko T H Bestor and C P Walsh ldquoReac-tivation of a silenced H19 gene in human rhabdomyosarcomaby demethylation of DNA but not by histone hyperacetylationrdquoMolecular Cancer vol 1 article 2 2002

[90] P Onyango and A P Feinberg ldquoA nucleolar protein H19opposite tumor suppressor (HOTS) is a tumor growth inhibitorencoded by a human imprinted H19 antisense transcriptrdquoProceedings of the National Academy of Sciences of the UnitedStates of America vol 108 no 40 pp 16759ndash16764 2011

[91] C E Albacker N Y Storer E M Langdon et al ldquoThe his-tone methyltransferase SUV39H1 suppresses embryonal rhab-domyosarcoma formation in zebrafishrdquo PLoS ONE vol 8 no 5Article ID e64969 2013

[92] N Y Storer R M White A Uong et al ldquoZebrafish rhab-domyosarcoma reflects the developmental stage of oncogeneexpression during myogenesisrdquo Development vol 140 no 14pp 3040ndash3050 2013

[93] M E Hatley W Tang M R Garcia et al ldquoA mouse modelof rhabdomyosarcoma originating from the adipocyte lineagerdquoCancer Cell vol 22 no 4 pp 536ndash546 2012

[94] V Hosur A Kavirayani J Riefler et al ldquoDystrophin and dysfer-lin doublemutantmice a novelmodel for rhabdomyosarcomardquoCancer Genetics vol 205 no 5 pp 232ndash241 2012

[95] W Goessling T E North and L I Zon ldquoNew waves of discov-ery modeling cancer in zebrafishrdquo Journal of Clinical Oncologyvol 25 no 17 pp 2473ndash2479 2007

[96] B P Rubin K Nishijo H-I H Chen et al ldquoEvidence for anunanticipated relationship between undifferentiated pleomor-phic sarcoma and embryonal rhabdomyosarcomardquo Cancer Cellvol 19 no 2 pp 177ndash191 2011

[97] S Z Kaylani J Xu R K Srivastava L Kopelovich J G Presseyand M Athar ldquoRapamycin targeting mTOR and hedgehog sig-naling pathways blocks human rhabdomyosarcoma growth inxenograft murine modelrdquo Biochemical and Biophysical ResearchCommunications vol 435 no 4 pp 557ndash561 2013

[98] S Q Li A T Cheuk J F Shern et al ldquoTargeting wild-type andmutationally activated FGFR4 in rhabdomyosarcoma with theinhibitor ponatinib (AP24534)rdquoPLoSONE vol 8 no 10 ArticleID e76551 2013

[99] K Miyoshi K Kohashi F Fushimi et al ldquoClose correlationbetween CXCR4 and VEGF expression and frequent CXCR7expression in rhabdomyosarcomardquo Human Pathology vol 45no 9 pp 1900ndash1909 2014

[100] ADMarshall F Picchione R I KGeltink andGCGrosveldldquoPAX3-FOXO1 induces up-regulation of Noxa sensitizing alve-olar rhabdomyosarcoma cells to apoptosisrdquo Neoplasia vol 15no 7 pp 738ndash748 2013

[101] F Perut S Avnet C Fotia et al ldquoV-ATPase as an effective thera-peutic target for sarcomasrdquo Experimental Cell Research vol 320no 1 pp 21ndash32 2014

Submit your manuscripts athttpwwwhindawicom

Stem CellsInternational

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

MEDIATORSINFLAMMATION

of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Behavioural Neurology

EndocrinologyInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Disease Markers

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

BioMed Research International

OncologyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Oxidative Medicine and Cellular Longevity

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

PPAR Research

The Scientific World JournalHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Immunology ResearchHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal of

ObesityJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Computational and Mathematical Methods in Medicine

OphthalmologyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Diabetes ResearchJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Research and TreatmentAIDS

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Gastroenterology Research and Practice

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Parkinsonrsquos Disease

Evidence-Based Complementary and Alternative Medicine

Volume 2014Hindawi Publishing Corporationhttpwwwhindawicom

Page 13: Review Article Rhabdomyosarcoma: Advances in Molecular and

Sarcoma 13

[48] A L Sarver L Li and S Subramanian ldquomicroRNA miR-183functions as an oncogene by targeting the transcription factorEGR1 and promoting tumor cell migrationrdquo Cancer Researchvol 70 no 23 pp 9570ndash9580 2010

[49] R L Galindo J A Allport and E N Olson ldquoA Drosophilamodel of the rhabdomyosarcoma initiator PAX7-FKHRrdquo Pro-ceedings of the National Academy of Sciences of the United Statesof America vol 103 no 36 pp 13439ndash13444 2006

[50] U Avirneni-Vadlamudi K A Galindo T R Endicott V Paul-son S Cameron and R L Galindo ldquoDrosophila and mammal-ian models uncover a role for the myoblast fusion gene TANC1in rhabdomyosarcomardquo The Journal of Clinical Investigationvol 122 no 1 pp 403ndash407 2012

[51] E Y Chen andDM Langenau ldquoZebrafishmodels of rhabdom-yosarcomardquoMethods in Cell Biology vol 105 pp 383ndash402 2011

[52] X Le E K Pugach S Hettmer et al ldquoA novel chemicalscreening strategy in zebrafish identifies common pathways inembryogenesis and rhabdomyosarcoma developmentrdquo Devel-opment vol 140 no 11 pp 2354ndash2364 2012

[53] Y Lee R Kawagoe K Sasai et al ldquoLoss of suppressor-of-fusedfunction promotes tumorigenesisrdquoOncogene vol 26 no 44 pp6442ndash6447 2007

[54] C Keller B R Arenkiel C M Coffin N El-Bardeesy R ADePinho and M R Capecchi ldquoAlveolar rhabdomyosarcomasin conditional Pax3Fkhr mice cooperativity of Ink4aARF andTrp53 loss of functionrdquo Genes and Development vol 18 no 21pp 2614ndash2626 2004

[55] S Singh CVinson CMGurley et al ldquoImpairedWnt signalingin embryonal rhabdomyosarcoma cells from p53c-fos doublemutant micerdquo American Journal of Pathology vol 177 no 4 pp2055ndash2066 2010

[56] G J Schaaf J M Ruijter F Van Ruissen et al ldquoFull transcrip-tome analysis of rhabdomyosarcoma normal and fetal skeletalmuscle statistical comparison of multiple SAGE librariesrdquo TheFASEB Journal vol 19 no 3 pp 404ndash406 2005

[57] D S Hawkins A A Gupta and E R Rudzinski ldquoWhat is newin the biology and treatment of pediatric rhabdomyosarcomardquoCurrent Opinion in Pediatrics vol 26 no 1 pp 50ndash56 2014

[58] M Zhang J Truscott and J Davie ldquoLoss of MEF2D expressioninhibits differentiation and contributes to oncogenesis in rhab-domyosarcoma cellsrdquoMolecular Cancer vol 12 no 1 article 1502013

[59] M Ciesla J Dulak and A Jozkowicz ldquomicroRNAs and epi-genetic mechanisms of rhabdomyosarcoma developmentrdquo TheInternational Journal of Biochemistry amp Cell Biology vol 53 pp482ndash492 2014

[60] S Ricaud B Vernus M Duclos et al ldquoInhibition of autocrinesecretion of myostatin enhances terminal differentiation inhuman rhabdomyosarcoma cellsrdquo Oncogene vol 22 no 51 pp8221ndash8232 2003

[61] C S Guo C Degnin T A Fiddler D Stauffer andM JThayerldquoRegulation of MyoD activity and muscle cell differentiation byMDM2 pRb and Sp1rdquoThe Journal of Biological Chemistry vol278 no 25 pp 22615ndash22622 2003

[62] S Rossi E Stoppani P L Puri and A Fanzani ldquoDifferenti-ation of human rhabdomyosarcoma RD cells is regulated byreciprocal functional interactions between myostatin p38 andextracellular regulated kinase signalling pathwaysrdquo EuropeanJournal of Cancer vol 47 no 7 pp 1095ndash1105 2011

[63] Z S Walters B Villarejo-Balcells D Olmos et al ldquoJARID2is a direct target of the PAX3-FOXO1 fusion protein andinhibits myogenic differentiation of rhabdomyosarcoma cellsrdquoOncogene vol 33 no 9 pp 1148ndash1157 2014

[64] T Hosoyama M I Aslam J Abraham et al ldquoIL-4R drives ded-ifferentiation mitogenesis and metastasis in rhabdomyosar-comardquo Clinical Cancer Research vol 17 no 9 pp 2757ndash27662011

[65] F S Dela Cruz ldquoCancer stem cells in pediatric sarcomasrdquoFrontiers in Oncology vol 3 article 168 2013

[66] M J Bissell and W C Hines ldquoWhy donrsquot we get more cancerA proposed role of the microenvironment in restraining cancerprogressionrdquo Nature Medicine vol 17 no 3 pp 320ndash329 2011

[67] E Charytonowicz C Cordon-Cardo I Matushansky and MZiman ldquoAlveolar rhabdomyosarcoma is the cell of origin amesenchymal stem cellrdquoCancer Letters vol 279 no 2 pp 126ndash136 2009

[68] W K Cavenee ldquoMuscling in on rhabdomyosarcomardquo NatureMedicine vol 8 no 11 pp 1200ndash1201 2002

[69] B Zhu and J K Davie ldquoNew insights into signalling-pathwayalterations in rhabdomyosarcomardquo British Journal of Cancervol 112 no 2 pp 227ndash231 2015

[70] M Zhang C M Linardic and D G Kirsch ldquoRAS and ROS inrhabdomyosarcomardquo Cancer Cell vol 24 no 6 pp 689ndash6912013

[71] S C Marianna L Ianzano and G Nicoletti ldquoTumor sup-pressor genes promote rhabdomyosarcoma progression in p53heterozygousrdquo Oncotarget vol 5 no 1 pp 108ndash119 2013

[72] SWang L Guo L Dong et al ldquoTGF-beta1 signal pathwaymaycontribute to rhabdomyosarcoma development by inhibitingdifferentiationrdquo Cancer Science vol 101 no 5 pp 1108ndash11162010

[73] J Wesche K Haglund and E M Haugsten ldquoFibroblast growthfactors and their receptors in cancerrdquo Biochemical Journal vol437 no 2 pp 199ndash213 2011

[74] M Wachtel J Rakic M Okoniewski P Bode F Niggli andB W Schafer ldquoFGFR4 signaling couples to Bim and not Bmfto discriminate subsets of alveolar rhabdomyosarcoma cellsrdquoInternational Journal of Cancer vol 135 no 7 pp 1543ndash15522014

[75] P L Puri ZWu P Zhang et al ldquoInduction of terminal differen-tiation by constitutive activation of p38 MAP kinase in humanrhabdomyosarcoma cellsrdquo Genes and Development vol 14 no5 pp 574ndash584 2000

[76] A M Tremblay E Missiaglia G G Galli et al ldquoThe Hippotransducer YAP1 transforms activated satellite cells and is apotent effector of embryonal rhabdomyosarcoma formationrdquoCancer Cell vol 26 no 2 pp 273ndash287 2014

[77] R N Judson A M Tremblay P Knopp et al ldquoThe hippo path-way member Yap plays a key role in influencing fate decisionsin muscle satellite cellsrdquo Journal of Cell Science vol 125 no 24pp 6009ndash6019 2012

[78] L E S Crose K A Galindo J G Kephart et al ldquoAlve-olar rhabdomyosarcoma-associated PAX3-FOXO1 promotestumorigenesis via Hippo pathway suppressionrdquo Journal ofClinical Investigation vol 124 no 1 pp 285ndash296 2014

[79] B Belyea J G Kephart J Blum D G Kirsch and C MLinardic ldquoEmbryonic signaling pathways and rhabdomyosar-coma contributions to cancer development and opportunitiesfor therapeutic targetingrdquo Sarcoma vol 2012 Article ID 40623913 pages 2012

14 Sarcoma

[80] J Huertas-Martinez S Rello-Varona D Herrero-Martin et alldquoCaveolin-1 is down-regulated in alveolar rhabdomyosarcomasand negatively regulates tumor growthrdquo Oncotarget vol 5 no20 pp 9744ndash9755 2014

[81] S Rossi P L Poliani CMissale EMonti andA Fanzani ldquoCav-eolins in rhabdomyosarcomardquo Journal of Cellular andMolecularMedicine vol 15 no 12 pp 2553ndash2568 2011

[82] P G Tsioli E S Patsouris C Giaginis and S E TheocharisldquoDNA repair systems in rhabdomyosarcomardquo Histology andHistopathology vol 28 no 8 pp 971ndash984 2013

[83] E Gatzidou C Michailidi S Tseleni-Balafouta and S Theo-charis ldquoAn epitome of DNA repair related genes and mecha-nisms in thyroid carcinomardquo Cancer Letters vol 290 no 2 pp139ndash147 2010

[84] R Rota R Ciarapica A Giordano L Miele and F LocatellildquoMicroRNAs in rhabdomyosarcoma pathogenetic implicationsand translational potentialityrdquoMolecular Cancer vol 10 article120 2011

[85] A Gougelet J Perez D Pissaloux et al ldquoMiRNA profiling howto bypass the current difficulties in the diagnosis and treatmentof sarcomasrdquo Sarcoma vol 2011 Article ID 460650 13 pages2011

[86] R Ciarapica G Russo F Verginelli et al ldquoDeregulated expres-sion of miR-26a and Ezh2 in rhabdomyosarcomardquo Cell Cyclevol 8 no 1 pp 172ndash175 2009

[87] L Li A L Sarver S Alamgir and S Subramanian ldquoDownreg-ulation of microRNAs miR-1 -206 and -29 stabilizes PAX3 andCCND2 expression in rhabdomyosarcomardquo Laboratory Investi-gation vol 92 no 4 pp 571ndash583 2012

[88] S-Y Ng G K Bogu B Soh and L W Stanton ldquoThe longnoncoding RNA RMST interacts with SOX2 to regulate neu-rogenesisrdquoMolecular Cell vol 51 no 3 pp 349ndash359 2013

[89] C A Lynch B Tycko T H Bestor and C P Walsh ldquoReac-tivation of a silenced H19 gene in human rhabdomyosarcomaby demethylation of DNA but not by histone hyperacetylationrdquoMolecular Cancer vol 1 article 2 2002

[90] P Onyango and A P Feinberg ldquoA nucleolar protein H19opposite tumor suppressor (HOTS) is a tumor growth inhibitorencoded by a human imprinted H19 antisense transcriptrdquoProceedings of the National Academy of Sciences of the UnitedStates of America vol 108 no 40 pp 16759ndash16764 2011

[91] C E Albacker N Y Storer E M Langdon et al ldquoThe his-tone methyltransferase SUV39H1 suppresses embryonal rhab-domyosarcoma formation in zebrafishrdquo PLoS ONE vol 8 no 5Article ID e64969 2013

[92] N Y Storer R M White A Uong et al ldquoZebrafish rhab-domyosarcoma reflects the developmental stage of oncogeneexpression during myogenesisrdquo Development vol 140 no 14pp 3040ndash3050 2013

[93] M E Hatley W Tang M R Garcia et al ldquoA mouse modelof rhabdomyosarcoma originating from the adipocyte lineagerdquoCancer Cell vol 22 no 4 pp 536ndash546 2012

[94] V Hosur A Kavirayani J Riefler et al ldquoDystrophin and dysfer-lin doublemutantmice a novelmodel for rhabdomyosarcomardquoCancer Genetics vol 205 no 5 pp 232ndash241 2012

[95] W Goessling T E North and L I Zon ldquoNew waves of discov-ery modeling cancer in zebrafishrdquo Journal of Clinical Oncologyvol 25 no 17 pp 2473ndash2479 2007

[96] B P Rubin K Nishijo H-I H Chen et al ldquoEvidence for anunanticipated relationship between undifferentiated pleomor-phic sarcoma and embryonal rhabdomyosarcomardquo Cancer Cellvol 19 no 2 pp 177ndash191 2011

[97] S Z Kaylani J Xu R K Srivastava L Kopelovich J G Presseyand M Athar ldquoRapamycin targeting mTOR and hedgehog sig-naling pathways blocks human rhabdomyosarcoma growth inxenograft murine modelrdquo Biochemical and Biophysical ResearchCommunications vol 435 no 4 pp 557ndash561 2013

[98] S Q Li A T Cheuk J F Shern et al ldquoTargeting wild-type andmutationally activated FGFR4 in rhabdomyosarcoma with theinhibitor ponatinib (AP24534)rdquoPLoSONE vol 8 no 10 ArticleID e76551 2013

[99] K Miyoshi K Kohashi F Fushimi et al ldquoClose correlationbetween CXCR4 and VEGF expression and frequent CXCR7expression in rhabdomyosarcomardquo Human Pathology vol 45no 9 pp 1900ndash1909 2014

[100] ADMarshall F Picchione R I KGeltink andGCGrosveldldquoPAX3-FOXO1 induces up-regulation of Noxa sensitizing alve-olar rhabdomyosarcoma cells to apoptosisrdquo Neoplasia vol 15no 7 pp 738ndash748 2013

[101] F Perut S Avnet C Fotia et al ldquoV-ATPase as an effective thera-peutic target for sarcomasrdquo Experimental Cell Research vol 320no 1 pp 21ndash32 2014

Submit your manuscripts athttpwwwhindawicom

Stem CellsInternational

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

MEDIATORSINFLAMMATION

of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Behavioural Neurology

EndocrinologyInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Disease Markers

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

BioMed Research International

OncologyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Oxidative Medicine and Cellular Longevity

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

PPAR Research

The Scientific World JournalHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Immunology ResearchHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal of

ObesityJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Computational and Mathematical Methods in Medicine

OphthalmologyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Diabetes ResearchJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Research and TreatmentAIDS

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Gastroenterology Research and Practice

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Parkinsonrsquos Disease

Evidence-Based Complementary and Alternative Medicine

Volume 2014Hindawi Publishing Corporationhttpwwwhindawicom

Page 14: Review Article Rhabdomyosarcoma: Advances in Molecular and

14 Sarcoma

[80] J Huertas-Martinez S Rello-Varona D Herrero-Martin et alldquoCaveolin-1 is down-regulated in alveolar rhabdomyosarcomasand negatively regulates tumor growthrdquo Oncotarget vol 5 no20 pp 9744ndash9755 2014

[81] S Rossi P L Poliani CMissale EMonti andA Fanzani ldquoCav-eolins in rhabdomyosarcomardquo Journal of Cellular andMolecularMedicine vol 15 no 12 pp 2553ndash2568 2011

[82] P G Tsioli E S Patsouris C Giaginis and S E TheocharisldquoDNA repair systems in rhabdomyosarcomardquo Histology andHistopathology vol 28 no 8 pp 971ndash984 2013

[83] E Gatzidou C Michailidi S Tseleni-Balafouta and S Theo-charis ldquoAn epitome of DNA repair related genes and mecha-nisms in thyroid carcinomardquo Cancer Letters vol 290 no 2 pp139ndash147 2010

[84] R Rota R Ciarapica A Giordano L Miele and F LocatellildquoMicroRNAs in rhabdomyosarcoma pathogenetic implicationsand translational potentialityrdquoMolecular Cancer vol 10 article120 2011

[85] A Gougelet J Perez D Pissaloux et al ldquoMiRNA profiling howto bypass the current difficulties in the diagnosis and treatmentof sarcomasrdquo Sarcoma vol 2011 Article ID 460650 13 pages2011

[86] R Ciarapica G Russo F Verginelli et al ldquoDeregulated expres-sion of miR-26a and Ezh2 in rhabdomyosarcomardquo Cell Cyclevol 8 no 1 pp 172ndash175 2009

[87] L Li A L Sarver S Alamgir and S Subramanian ldquoDownreg-ulation of microRNAs miR-1 -206 and -29 stabilizes PAX3 andCCND2 expression in rhabdomyosarcomardquo Laboratory Investi-gation vol 92 no 4 pp 571ndash583 2012

[88] S-Y Ng G K Bogu B Soh and L W Stanton ldquoThe longnoncoding RNA RMST interacts with SOX2 to regulate neu-rogenesisrdquoMolecular Cell vol 51 no 3 pp 349ndash359 2013

[89] C A Lynch B Tycko T H Bestor and C P Walsh ldquoReac-tivation of a silenced H19 gene in human rhabdomyosarcomaby demethylation of DNA but not by histone hyperacetylationrdquoMolecular Cancer vol 1 article 2 2002

[90] P Onyango and A P Feinberg ldquoA nucleolar protein H19opposite tumor suppressor (HOTS) is a tumor growth inhibitorencoded by a human imprinted H19 antisense transcriptrdquoProceedings of the National Academy of Sciences of the UnitedStates of America vol 108 no 40 pp 16759ndash16764 2011

[91] C E Albacker N Y Storer E M Langdon et al ldquoThe his-tone methyltransferase SUV39H1 suppresses embryonal rhab-domyosarcoma formation in zebrafishrdquo PLoS ONE vol 8 no 5Article ID e64969 2013

[92] N Y Storer R M White A Uong et al ldquoZebrafish rhab-domyosarcoma reflects the developmental stage of oncogeneexpression during myogenesisrdquo Development vol 140 no 14pp 3040ndash3050 2013

[93] M E Hatley W Tang M R Garcia et al ldquoA mouse modelof rhabdomyosarcoma originating from the adipocyte lineagerdquoCancer Cell vol 22 no 4 pp 536ndash546 2012

[94] V Hosur A Kavirayani J Riefler et al ldquoDystrophin and dysfer-lin doublemutantmice a novelmodel for rhabdomyosarcomardquoCancer Genetics vol 205 no 5 pp 232ndash241 2012

[95] W Goessling T E North and L I Zon ldquoNew waves of discov-ery modeling cancer in zebrafishrdquo Journal of Clinical Oncologyvol 25 no 17 pp 2473ndash2479 2007

[96] B P Rubin K Nishijo H-I H Chen et al ldquoEvidence for anunanticipated relationship between undifferentiated pleomor-phic sarcoma and embryonal rhabdomyosarcomardquo Cancer Cellvol 19 no 2 pp 177ndash191 2011

[97] S Z Kaylani J Xu R K Srivastava L Kopelovich J G Presseyand M Athar ldquoRapamycin targeting mTOR and hedgehog sig-naling pathways blocks human rhabdomyosarcoma growth inxenograft murine modelrdquo Biochemical and Biophysical ResearchCommunications vol 435 no 4 pp 557ndash561 2013

[98] S Q Li A T Cheuk J F Shern et al ldquoTargeting wild-type andmutationally activated FGFR4 in rhabdomyosarcoma with theinhibitor ponatinib (AP24534)rdquoPLoSONE vol 8 no 10 ArticleID e76551 2013

[99] K Miyoshi K Kohashi F Fushimi et al ldquoClose correlationbetween CXCR4 and VEGF expression and frequent CXCR7expression in rhabdomyosarcomardquo Human Pathology vol 45no 9 pp 1900ndash1909 2014

[100] ADMarshall F Picchione R I KGeltink andGCGrosveldldquoPAX3-FOXO1 induces up-regulation of Noxa sensitizing alve-olar rhabdomyosarcoma cells to apoptosisrdquo Neoplasia vol 15no 7 pp 738ndash748 2013

[101] F Perut S Avnet C Fotia et al ldquoV-ATPase as an effective thera-peutic target for sarcomasrdquo Experimental Cell Research vol 320no 1 pp 21ndash32 2014

Submit your manuscripts athttpwwwhindawicom

Stem CellsInternational

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

MEDIATORSINFLAMMATION

of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Behavioural Neurology

EndocrinologyInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Disease Markers

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

BioMed Research International

OncologyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Oxidative Medicine and Cellular Longevity

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

PPAR Research

The Scientific World JournalHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Immunology ResearchHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal of

ObesityJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Computational and Mathematical Methods in Medicine

OphthalmologyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Diabetes ResearchJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Research and TreatmentAIDS

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Gastroenterology Research and Practice

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Parkinsonrsquos Disease

Evidence-Based Complementary and Alternative Medicine

Volume 2014Hindawi Publishing Corporationhttpwwwhindawicom

Page 15: Review Article Rhabdomyosarcoma: Advances in Molecular and

Submit your manuscripts athttpwwwhindawicom

Stem CellsInternational

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

MEDIATORSINFLAMMATION

of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Behavioural Neurology

EndocrinologyInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Disease Markers

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

BioMed Research International

OncologyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Oxidative Medicine and Cellular Longevity

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

PPAR Research

The Scientific World JournalHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Immunology ResearchHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal of

ObesityJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Computational and Mathematical Methods in Medicine

OphthalmologyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Diabetes ResearchJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Research and TreatmentAIDS

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Gastroenterology Research and Practice

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Parkinsonrsquos Disease

Evidence-Based Complementary and Alternative Medicine

Volume 2014Hindawi Publishing Corporationhttpwwwhindawicom